AU767640B2 - Vertebrate embryonic pattern-inducing hedgehog-like proteins - Google Patents

Vertebrate embryonic pattern-inducing hedgehog-like proteins Download PDF

Info

Publication number
AU767640B2
AU767640B2 AU87319/01A AU8731901A AU767640B2 AU 767640 B2 AU767640 B2 AU 767640B2 AU 87319/01 A AU87319/01 A AU 87319/01A AU 8731901 A AU8731901 A AU 8731901A AU 767640 B2 AU767640 B2 AU 767640B2
Authority
AU
Australia
Prior art keywords
seq
hedgehog
polypeptide
cells
amino acid
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.)
Expired
Application number
AU87319/01A
Other versions
AU8731901A (en
Inventor
Philip Ingham
Andrew P. Mcmahon
Clifford J. Tabin
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.)
Cancer Research Horizons Ltd
Harvard College
Original Assignee
Imperial Cancer Research Technology Ltd
Harvard College
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/356,060 external-priority patent/US5844079A/en
Priority claimed from AU16451/99A external-priority patent/AU736492C/en
Application filed by Imperial Cancer Research Technology Ltd, Harvard College filed Critical Imperial Cancer Research Technology Ltd
Priority to AU87319/01A priority Critical patent/AU767640B2/en
Publication of AU8731901A publication Critical patent/AU8731901A/en
Application granted granted Critical
Publication of AU767640B2 publication Critical patent/AU767640B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Description

AUSTRALIA
PATENTS ACT 1990 DIVISIONAL APPLICATION NAME OF APPLICANTS: President and Fellows of Harvard College AND Imperial Cancer Research Technology Ltd ADDRESS FOR SERVICE: DAVIES COLLISON CAVE Patent Attorneys 1 Little Collins Street Melbourne, 3000.
INVENTION TITLE: "Vertebrate embryonic pattern-inducing hedgehog-like proteins" The following statement is a full description of this invention, including the best method of performing it known to us: -la- VERTEBRATE EMBRYONIC PATTERN-INDUCING HEDGEHOG-LIKE
PROTEINS.
Background of the Invention Any discussion of the prior art throughout the specification should in no way be considered as an admission that such prior art is widely known or forms part of common general knowledge in the field.
Pattern formation is the activity by which embryonic cells form ordered spatial arrangements of differentiated tissues. The physical complexity of higher organisms arises during embryogenesis through the interplay of cell-intrinsic lineage and cellextrinsic signalling. Inductive interactions are essential to embryonic patterning in vertebrate development from the earliest establishment of the body plan, to the patterning of the organ systems, to the generation of diverse cell types during tissue differentiation (Davidson, (1990) Development 108: 365-389; Gurdon, (1992) Cell 68: 185-199; Jessell, T.M. et al., (1992) Cell 68: 257-270). The effects of developmental cell interactions are varied. Typically, responding cells are diverted from one route of cell differentiation to another by inducing cells that differ from both the uninduced and induced states of the responding cells (inductions). Sometimes cells induce their neighbors to differentiate like themselves (homoiogenetic induction); in S.other cases a cell inhibits its neighbors from differentiating like itself. Cell interactions 20 in early development may be sequential, such that an initial induction between two cell types leads to a progressive amplification of diversity. Moreover, inductive interactions ,oooccur not only in embryos, but in adult cells as well, and can act to establish and maintain morphogenetic patterns as well as induce differentiation Gurdon (1992) Cell 68: 185-199).
The origin of the nervous system in all vertebrates can be traced to the end of gastrulation. At this time, the ectoderm in the dorsal side of the embryo changes its fate S°from epidermal to neural. The newly formed neuroectoderm thickens to form a flattened structure called the neural plate which is characterized, in some vertebrates, by a central .groove (neural groove) and thickened lateral edges (neural folds). At its early stages of differentiation, the neural plate already exhibits signs of regional differentiation along its o. anterior posterior and mediolateral axis The neural folds eventually fuse at the dorsal midline to form the neural tube which will differentiate into brain at its anterior end and spinal cord at its posterior end. Closure of the neural tube creates 500230206_ .Doc/BSW lbdorsal/ventral differences by virtue of previous mediolateral differentiation. Thus, at the end ofneurulation, the neural tube has a clear anterior-posterior dorsal ventral (D- V) and mediolateral polarities (see, for example, Principles in Neural Science 3 rd), eds. Kandel, Schwartz and Jessell, Elsevier Science Publishing Company: NY, 1991; and Developmental Biology 3 rd), ed. S.F. Gilbert, Sinauer Associates: Sunderland MA, 1991). Inductive interactions that define the fate of cells within the neural tube establish the initial pattern of the embryonic vertebrate nervous system. In the spinal cord, the identify of cell types is controlled, in part, by signals from two a o 500230206_1 DOC/BSW midline cell groups, the notochord and floor plate, that induce neural plate cells to differentiate into floor plate, motor neurons, and other ventral neuronal types (van Straaten et al. (1988) Anat. Embryol. 177:317-324; Placzek et al. (1993) Development 117:205-218; Yamada et al. (1991) Cell 64:035-647; and Hatta et al. (1991) Nature 350:339-341). In addition, signals from the floor plate are responsible for the orientation and direction of commissural neuron outgrowth (Placzek, M. et al., (1990) Development 110: 19-30). Besides patterning the neural tube, the notochord and floorplate are also responsible for producing signals which control the patterning of the somites by inhibiting differentiation of dorsal somite derivatives in the ventral regions (Brand-Saberi, B. et al., (1993) Anat. Embryol. 188: 239-245; Porquie, O. et al., (1993) Proc. Natl. Acad Sci. USA 90: 5242-5246).
Another important signaling center exists in the posterior mesenchyme of developing limb buds, called the Zone of Polarizing Activity, or "ZPA". When tissue from the posterior region of the limb bud is grafted to the anterior border of a second limb bud, the resultant limb will develop with additional digits in a mirror-image sequence along the anteroposterior axis (Saunders and Gasseling, (1968) Epithelial-Mesenchymal Interaction, pp. 78-97). This finding has led to the model that the ZPA is responsible for normal anteroposterior patterning in the limb. The ZPA has been hypothesized to function by releasing a signal, termed a "morphogen", which forms a gradient across the early embryonic bud. According to this S model, the fate of cells at different distances from the ZPA is determined by the local concentration of the morphogen, with specific thresholds of the morphogen inducing successive structures (Wolpert, (1969) Theor. Biol. 25:1-47). This is supported by the finding that the extent of digit duplication is proportional to the number of implanted ZPA cells (Tickle, (1981) Nature 254:199-202).
A candidate for the putative ZPA morphogen was identified by the discovery that a 25 source of retinoic acid can result in the same type of mirror-image digit duplications when placed in the anterior of a limb bud (Tickle et al., (1982) Nature 296:564-565; Summerbell, (1983) J. Embryol 78:269-289). The response to exogenous retinoic acid is concentration dependent as the morphogen model demands (Tickle et al., (1985) Dev. Biol. 109:82-95).
Moreover, a differential distribution of retinoic acid exists across the limb bud, with a higher 30 concentration in the ZPA region (Thaller and Eichele, (1987) Nature 327:625-628).
Recent evidence, however, has indicated that retinoic acid is unlikely to be the endogenous factor responsible for ZPA activity (reviewed in Brockes, (1991) Nature 350:15; Tabin, (1991) Cell 66:199-217). It is now believed that rather than directly mimicking an endogenous signal, retinoic acid implants act by inducing an ectopic ZPA. The anterior limb tissue just distal to a retinoic acid implant and directly under the ectoderm has been demonstrated to acquire ZPA activity by serially transplanting that tissue to another limb bud (Summerbell and Harvey, (1983) Limb Development and Regeneration pp. 109-118; Wanek -3et al., (1991) Nature 350:81-83). Conversely, the tissue next to a ZPA graft does not gain ZPA activity (Smith, (1979) J. Embryol 52:105-113). Exogenous retinoic acid would thus appear to act upstream of the ZPA in limb patterning.
The immediate downstream targets of ZPA action are not known. However, one important set of genes which are ectopically activated during ZPA-induced pattern duplications are the 5' genes of the Hoxd cluster. These genes are normally expressed in a nested pattern emanating from the posterior margin of the limb bud (Dolle et al., (1989) Nature 342:767-772; Izpisua-Belmonte et al., (1991) Nature 350:585-589). This nested pattern of Hox gene expression has been directly demonstrated to determine the identity of the structures produced along the anteroposterior axis of the limb (Morgan et al., (1993) Nature 358:236-239). As this would predict, ZPA grafts which produce mirror-image duplication of structures at an anatomical level first lead to the ectopic activation of the Hoxd genes in a mirror-image duplication at the molecular level.
(Nohno et al., (1991) Cell 64:1197-1205; Izpisua-Belmonte et al., (1991) Nature 350:585-589). The molecular signals which regulate the expression of these important genes are currently not understood.
Summary of the Invention According to a first aspect, the present invention provides an isolated nucleic acid encoding a polypeptide including a hedgehog amino acid sequence at least S 20 identical to at least one ofSEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to apatched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
SAccording to a second aspect, the present invention provides an isolated nucleic acid which hybridizes under highly stringent conditions to a nucleic acid sequence selected from SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID 30 NO: 6, and SEQ ID NO: 7, which nucleic acid encodes a hedgehog amino acid sequence that binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation 5002302061 .Doc/BSW -3aof chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
According to a third aspect, the present invention provides a method for inducing a cell to differentiate to a neuronal cell phenotype, including contacting said cell with a polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
According to a fourth aspect, the present invention provides a method of modulating skeletogenesis including contacting a target tissue with an amount of a polypeptide effective to cause one or both of chondrogenesis and osteogenesis in the target tissue, said polypeptide including a hedgehog amino acid sequence at least identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which 20 hedgehog amino acid sequence binds to apatched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
According to a fifth aspect, the present invention provides a method for treating a degenerative disorder of the nervous system characterized by neuronal cell death, including administering to a patient a therapeutically effective amount of a pharmaceutical preparation of a polypeptide, thereby causing, relative to the absence of treatment, prolonged survival of neural cells in said patient, said polypeptide including a 30 hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates 500230206_ iDOc/BSW 3b survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, (v) regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
According to a sixth aspect, the present invention provides a method for modulating one or more of growth, differentiation and survival of a neuronal cell, including contacting said cell with an effective amount of a polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, (v) regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
According to a seventh aspect, the present invention provides a method for promoting survival of mammalian neuronal cells responsive to hedgehog induction, including treating the cell with an amount of a hedgehog polypeptide effective to increase the rate of survival of the neuronal cells, said polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID 20 NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of S differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
According to an eighth aspect, the present invention provides a method for promoting growth of mammalian neuronal stem cells, including treating the cell with an amount of a polypeptide effective to increase the rate of growth of the neuronal stem cells, said polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ 30 ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ 500230206 l.DOC/BSW 3c lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
According to a ninth aspect, the present invention provides a method for preventing, treating or reducing the severity of a neurodegenerative disorder, including administering to a subject a therapeutically effective amount of a polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, (v) regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
According to a tenth aspect, the present invention provides a method for preventing, treating or reducing the severity of an acute, subacute or chronic injury to the nervous system in a subject, including administering to a subject a amount of a polypeptide therapeutically effective to alter, relative to the absence of treatment, at least one of rate of growth, (ii) differentiation, or (iii) survival of one or more neuronal cell-types in said subject, said polypeptide including a hedgehog amino acid sequence at y: least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 20 SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
According to an eleventh aspect, the present invention provides a method for modulating the growth state of a chondrocytic cell including contacting the cell with a polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 30 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to apatched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces 500230206 I.DOC/BSW -3dexpression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
According to a twelfth aspect, the present invention provides a method for treating a skeletogenic disorder, including administering to a patient an amount of a hedgehog polypeptide effective to alleviate conditions of the disorder, said polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
According to a thirteenth aspect, the present invention provides use of a pharmaceutical preparation of a polypeptide, said polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a "patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila for the manufacture of a medicament for treating a degenerative disorder of the nervous system characterized by neuronal cell death.
According to a fourteenth aspect, the present invention provides use of a polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal 30 cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in 500230206 1.DOC/BSW -3etransgenic drosophila for the manufacture of a medicament for preventing, treating or reducing the severity of a neurodegenerative disorder.
According to a fifteenth aspect, the present invention provides use of a polypeptide therapeutically effective to alter, relative to the absence of treatment, at least one of rate of growth, (ii) differentiation, or (iii) survival of one or more neuronal cell-types in said subject, said polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to apatched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila for the manufacture of a medicament for preventing, treating or reducing the severity of an acute, subacute or chronic injury to the nervous system in a subject.
According to a sixteenth aspect, the present invention provides use of a hedgehog polypeptide said polypeptide including a hedgehog amino acid sequence at least identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to apatched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila for the manufacture of a medicament for treating a skeletogenic disorder.
The present invention relates to the discovery of a novel family of proteins present in vertebrate organisms, referred to hereinafter as "hedgehog" proteins, which proteins have apparent broad involvement in the formation and maintenance of ordered Sspatial arrangements of differentiated tissues in vertebrates, and can be used to generate 30 and/or maintain an array of different vertebrate tissue both in vitro and in vivo.
In general, the invention features hedgehog polypeptides, preferably substantially pure preparations of one or more of the subject hedgehog polypeptides. The invention also provides recombinantly produced hedgehog polypeptides. In preferred 500230206 1.DOC/BSW 3fembodiments the polypeptide has a biological activity including: an ability to modulate proliferation, survival and/or differentiation of mesodermally-derived tissue, such as tissue derived from dorsal mesoderm; the ability to modulate proliferation, survival and/or differentiation of ectodermally-derived tissue, such as tissue derived from the neural tube, neural crest, or head mesenchyme; the ability to modulate proliferation, survival and/or differentiation of endodermally-derived tissue, such as tissue derived from the primitive gut. Moreover, in preferred embodiments, the subject hedgehog proteins have the ability to induce expression of secondary signaling molecules, such as members of the Transforming Growth Factor P family, as well as members of the fibroblast growth factor (FGF) family.
a e *a
C
e a *a *o/ 500230206 i.Doc/BSW In a certain embodiments, the polypeptide is identical with or homologous to a Sonic hedgehog (Shh) polypeptide, such as a mammalian Shh represented by SEQ ID Nos: 13 or 11, an avian Shh represented by SEQ ID No: 8, or a fish Shh represented by SEQ ID No: 12. For instance, the Shh polypeptide preferably has an amino acid sequence at least homologous to a polypeptide represented by any of SEQ ID Nos: 8, 11, 12 or 13, though polypeptides with higher sequence homologies of, for example, 80%, 90% or 95% are also contemplated. Exemplary Shh proteins are represented by SEQ ID No. 40. The Shh polypeptide can comprise a full length protein, such as represented in the sequence listings, or it can comprise a fragment of, for instance, at least 5, 10, 20, 50, 100 or 150 amino acids in length. Preferred hedgehog polypeptides include Shh sequences corresponding approximately to the natural proteolytic fragments of the hedgehog proteins, such as from about Cys-24 through Glu-188, or from about Asn-189 through Ala-475 of the human Shh protein, or analogous fragments thereto.
In other embodiments, the polypeptide is identical with or homologous to an Indian hedgehog (Ihh) polypeptide, such as a human Ihh represented by SEQ ID No: 14, or a mouse Ihh represented by SEQ ID No: 10. For instance, the Ihh polypeptide preferably has an amino acid sequence at least 70% homologous to a polypeptide represented by either of SEQ ID Nos: 10 or 14, though Ihh polypeptides with higher sequence homologies of, for example.
90% or 95% are also contemplated. The polypeptide can comprise the full length protein represented by in part by these sequences, or it can comprise a fragment of, for S* instance, at least 5, 10, 20, 50, 100 or 150 amino acids in length. Preferred Ihh polypeptides o comprise an N-terminal fragment including Arg-1 through Glu-94, or a C-terminal fragment including His-95 through Ser-3312 of the human Ihh represented by SEQ ID No: 14, or analogous fragments thereto.
-25 In still further embodiments, the polypeptide is identical with or homologous to a Desert hedgehog (Dhh) polypeptide, such as a mouse Dhh represented by SEQ ID No: 9. For instance, the Dhh polypeptide preferably has an amino acid sequence at least homologous to a polypeptide represented by SEQ ID No: 9, though Dhh polypeptides with higher sequence homologies of, for example, 80%, 90% or 95% are also contemplated. The *30 polypeptide can comprise the full length protein represented by this sequence, or it can comprise a fragment of, for instance, at least 5, 10, 20, 50, 100 or 150 amino acids in length.
Preferred Dhh polypeptides comprise Dhh sequences corresponding to the N-terminal portion of the protein, e.g. Cys-23 through Asp-189 or Asn-190 through Gly-396 of SEQ ID No: 9, or analogous fragments thereto.
Moreover, as described below, the hedgehog polypeptide can be either an agonist mimics), or alternatively, an antagonist of a biological activity of a naturally occurring form of the protein, the polypeptide is able to modulate differentiation and/or growth and/or survival of a cell responsive to authentic hedgehog proteins. Homologs of the subject hedgehog proteins include versions of the 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.
The hedgehog polypeptides of the present invention can be glycosylated, or conversely, by choice of the expression system or by modification of the protein sequence to preclude glycosylation, reduced carbohydrate analogs can also be provided. Glycosylated forms include derivatization with glycosaminoglycan chains. Likewise, hedgehog polypeptides can be generated which lack an endogenous signal sequence (though this is typically cleaved off even if present in the pro-form of the protein).
The subject proteins can also be provided as chimeric molecules, such as in the form of fusion proteins. For instance, the hedgehog protein can be provided as a recombinant fusion protein which includes a second polypeptide portion, a second polypeptide having an amino acid sequence unrelated to hedgehog, e.g. the second polypeptide portion is glutathione-S-transferase, e.g. the second polypeptide portion is an enzymatic activity such as alkaline phosphatase, e.g. the second polypeptide portion is an epitope tag.
Yet another aspect of the present invention concerns an immunogen comprising a hedgehog polypeptide in an immunogenic preparation, the immunogen being capable of eliciting an immune response specific for a hedgehog polypeptide; e.g. a humoral response, 20 e.g. an antibody response; e.g. a cellular response. In preferred embodiments, the immunogen Scomprising an antigenic determinant, e.g. a unique determinant, from a protein represented by one of SEQ ID Nos. 8-14.
A still further aspect of the present invention features antibodies and antibody preparations specifically reactive with an epitope of the hedgehog immunogen.
Another aspect of the present invention provides a substantially isolated nucleic acid having a nucleotide sequence which encodes a hedgehog polypeptide. In preferred embodiments, the encoded polypeptide specifically agonizes or antagonizes inductive events mediated by wild-type hedgehog proteins. The coding sequence of the nucleic acid can S-comprise a sequence which is identical to a coding sequence represented in one of SEQ ID Nos: 1-7, or it can merely be homologous to one or more of those sequences. For instance, the hedgehog encoding sequence preferably has a sequence at least 70% homologous to a nucleotide sequence in one or more of SEQ ID Nos: 1-7, though higher sequence homologies of, for example, 80%, 90% or 95% are also contemplated. The polypeptide encoded by the nucleic acid can comprise an amino acid sequence represented in one of SEQ ID Nos: 8-14 such as one of those full length proteins, or it can comprise a fragment of that nucleic acid, which fragment may, for instance, encode a fragment which is, for example, at least 5, 10, or 100 amino acids in length. The polypeptide encoded by the nucleic acid can be either an agonist mimics), or alternatively, an antagonist of a biological activity of a naturally occurring form of a hedgehog protein.
Furthermore, in certain preferred embodiments, the subject hedgehog nucleic acid will include a transcriptional regulatory sequence, e.g. at least one of a transcriptional promoter or transcriptional enhancer sequence, which regulatory sequence is operably linked to the hedgehog gene sequence. Such regulatory sequences can be used in to render the hedgehog gene sequence suitable for use as an expression vector.
In yet a further preferred embodiment, the nucleic acid hybridizes under stringent conditions to a nucleic acid probe corresponding to at least 12 consecutive nucleotides of one or more of SEQ ID Nos: 1-7; though preferably corresponding to at least 20 consecutive nucleotides; and more preferably corresponding to at least 40, 50 or 75 consecutive nucleotides of one or more of SEQ ID Nos: 1-7.
The invention also features transgenic non-human animals, e.g. mice, rats, rabbits, chickens, frogs or pigs, having a transgene, animals which include (and preferably express) a heterologous form of a hedgehog gene described herein, or which misexpress an endogenous hedgehog gene, an animal in which expression of one or more of the subject hedgehog proteins is disrupted. Such a transgenic animal can serve as an animal model for studying cellular and tissue disorders comprising mutated or mis-expressed hedgehog alleles S* 20 or for use in drug screening.
The invention also provides a probe/primer comprising a substantially purified oligonucleotide, wherein the oligonucleotide comprises a region of nucleotide sequence 2 which hybridizes under stringent conditions to at least 10 consecutive nucleotides of sense or antisense sequence of SEQ ID No: 1, or naturally occurring mutants thereof. Nucleic acid probes which are specific for each of the classes of vertebrate hedgehog proteins are contemplated by the present invention, e.g. probes which can discern between nucleic acid encoding an Shh versus an Ihh versus a Dhh versus an Mhh. In preferred embodiments, the probe/primer further includes a label group attached thereto and able to be detected. The label group can be selected, from a group consisting of radioisotopes, fluorescent 30 compounds, enzymes, and enzyme co-factors. Probes of the invention can be used as a part of a diagnostic test kit for identifying dysfunctions associated with mis-expression of a hedgehog protein, such as for detecting in a sample of cells isolated from a patient, a level of a nucleic acid encoding a subject hedgehog protein; e.g. measuring a hedgehog mRNA level in a cell, or determining whether a genomic hedgehog gene has been mutated or deleted.
Preferably, the oligonucleotide is at least 10 nucleotides in length, though primers of 20, 100, or 150 nucleotides in length are also contemplated.
In yet another aspect, the invention provides an assay for screening test compounds for inhibitors, or alternatively, potentiators, of an interaction between a hedgehog protein and a hedgehog receptor. An exemplary method includes the steps of combining a hedgehog receptor, either soluble or membrane bound (including whole cells), a hedgehog polypeptide, and a test compound, under conditions wherein, but for the test compound, the hedgehog protein and the hedgehog receptor are able to interact; and (ii) detecting the formation of a complex which includes the hedgehog protein and the receptor either by directly quantitating the complex or by measuring inductive effects of the hedgehog protein. A statistically significant change, such as a decrease, in the formation of the complex in the presence of a test compound (relative to what is seen in the absence of the test compound) is indicative of a modulation, inhibition, of the interaction between the hedgehog protein and the receptor.
Another aspect of the present invention relates to a method of inducing and/or maintaining a differentiated state, causing proliferation, and/or enhancing survival of a cell (from a vertebrate organism) responsive to a hedgehog protein, by contacting the cells with a hedgehog agonist. For example, the present method is applicable to cell culture technique, such as in the culturing of neuronal and other cells whose survival or differentiative state is dependent on hedgehog function. Moreover, hedgehog agonists and antagonists can be used for therapeutic intervention, such as to enhance survival and maintenance of neurons and other neural cells in both the central nervous system and the peripheral nervous system, as oo* 20 well as to influence other vertebrate organogenic pathways, such as other ectodermal O*o patterning, as well as certain mesodermal and endodermal differentiation processes. In addition to the vertebrate hedgehog-like proteins, the present invention further contemplates the use of Drosophila Hedgehog (Dros-HH) to induce cells and tissue of vertebrate organisms in similar fashion to the subject hedgehog proteins.
25 Another aspect of the present invention provides a method of determining if a subject, e.g. a human patient, is at risk for a disorder characterized by unwanted cell proliferation or aberrant control of differentiation. The method includes detecting, in a tissue of the subject, i the presence or absence of a genetic lesion characterized by at least one of a mutation of a gene encoding a hedgehog protein, e.g. represented in SEQ ID No: 2, or a homolog thereof; 30 or (ii) the mis-expression of a hedgehog gene. In preferred embodiments, detecting the genetic lesion includes ascertaining the existence of at least one of: a deletion of one or more nucleotides from a hedgehog gene; an addition of one or more nucleotides to the gene, a substitution of one or more nucleotides of the gene, a gross chromosomal rearrangement of the gene; an alteration in the level of a messenger RNA transcript of the gene; the presence of a non-wild type splicing pattern of a messenger RNA transcript of the gene; or a non-wild type level of the protein.
-8- For example, detecting the genetic lesion can include providing a probe/primer including an oligonucleotide containing a region of nucleotide sequence which hybridizes to a sense or antisense sequence of a hedgehog gene, e.g. a nucleic acid represented in one of SEQ ID Nos: 1-7, or naturally occurring mutants thereof, or 5' or 3' flanking sequences naturally associated with the hedgehog gene; (ii) exposing the probe/primer to nucleic acid of the tissue; and (iii) detecting, by hybridization of the probe/primer to the nucleic acid, the presence or absence of the genetic lesion; e.g.
wherein detecting the lesion comprises utilizing the probe/primer to determine the nucleotide sequence of the hedgehog gene and, optionally, of the flanking nucleic acid sequences. For instance, the probe/primer can be employed in a polymerase chain reaction (PCR) or in a ligation chain reaction (LCR). In alternate embodiments, the level of a hedgehog protein is detected in an immunoassay using an antibody which is specifically immunoreactive with the hedgehog protein.
The practice of the present invention will employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained filly in the literature. See, for example, Molecular Cloning A Laboratory Manual, 2 n d Ed., ed. By Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1989); DNA Cloning, Volumes I and II 20 N. Glover ed., 1985); Oligonucleotide Synthesis J. Gait ed., 1984); Mullis et al.
U.S. Patent No. 4,683,195; Nucleic Acid Hybridization Hames S. J. Higgins eds. 1984); Transcription And Translation Hames S. J. Higgins eds. 1984); Culture ofAnimal Cells I. Freshney, Alan R. Liss, Inc., 1987); Immobilized Cells And Enzymes (IRL Press, 1986); B. Perbal, A Practical Guide to Molecular Cloning (1984); the treatise, Methods In Enzymology (Academic Press, Inc., Gene Transfer Vectors For Mammalian Cells H. Miller and M. P. Calos eds., 1987, Cold Spring Harbor Laboratory); Methods in Enzymology, Vols. 154 and 155 (Wu et al. eds.), Immunochemical Methods In Cell And Molecular Biology (Mayer and Walker, eds., Academic Press, London, 1987); Handbook of Experimental Immunology, Volumes I-IV 30 M. Weir and C. C. Blackwell, eds., 1986); Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1986).
500230206 l.DOC/BSW 8a- Other features and advantages of the invention will be apparent from the following detailed description, and from the claims.
Unless the context clearly requires otherwise, throughout the description and the claims, the words 'comprise', 'comprising', and the like are to be construed in an inclusive sense as opposed to an exclusive or exhaustive sense; that is to say, in the sense of "including, but not limited to".
BriefDescription of the Drawings Figure 1 represents the amino acid sequences of two chick hh clones, chicken hedgehog-A (pCHA; SEQ ID No:35) and chicken hedgehog-B (pCHB; SEQ ID No:36).
These clones were obtained using degenerate primers corresponding to the underlined amino 0* o/o 500230206_ .DOc/BSW 9 acid residues of the Drosophila sequence (corresponding to residues 161-232 of SEQ ID No:34) also shown in Figure 1, followed by nested PCR using chicken genomic DNA.
Figure 2 is an alignment comparing the amino acid sequences of chick Shh (SEQ ID No:8) with its Drosophila homolog (SEQ ID No:34). Shh residues 1-26 correspond to the proposed signal peptide. Identical residues are enclosed by boxes and gaps in order to highlight similarity. The nucleotide sequence of Shh has been submitted to Genbank.
Figure 3 is a hydropathy plot for the predicted chick Shh protein, generated by the methods of Kyte and Doolittle (1982). The values of hydrophobicity are plotted against the amino acid positions. Negative values predict a hydrophobic domain of the protein.
Figure 4 is an alignment comparing the amino acid sequences of various hh proteins.
The white region on the amino terminus of chicken Shh corresponds to the putative signal peptide. The black box refers to a highly conserved region from aa residues 26-207 of SEQ ID No:8). The arrows point to exon boundaries in the Drosophila gene (Lee et al. (1992) Cell 71: 33-50). In each case, the proteins are compared to chicken Shh (SEQ ID No:8) and the percent amino acid identity is indicated in each region's box.
Figure 5A is a "pileup" alignment of predicted amino acid sequences which compares Drosophila hh (D-hh; SEQ ID No:34), mouse hh (M-Dhh; SEQ ID No:9; M-Ihh; SEQ ID No: 10; M-Shh; SEQ ID No:l chicken hh (C-Shh; SEQ ID No:8), and zebrafish hh (Z-Shh; SEQ ID No:12). The predicted hydrophobic transmembrane/signal sequences are indicated 20 in italics and the predicted signal sequence processing site is arrowed. The positions of introns interrupting the Drosophila hh and M-Dhh open reading frames are indicated by arrowheads. All amino acids shared among the six predicted hh proteins are indicated in bold. Figure 5B is a sequence alignment of the N-terminal portion of vertebrate hedgehog proteins, and the predicted degenerate sequence "CON" (SEQ ID No: 41).
Figure 6 is an inter- and cross-species comparison of amino acid identities among the predicted processed hh proteins shown in Figure 5A. All values are percentages. Figures in parentheses represent similarities allowing for conservative amino acid substitutions.
Figure 7 is a representation of the DNA constructs used in transgenic studies to study Sectopic expression of chick Shh in mouse embryos. Constructs were generated for ectopic expression of cDNA clones in the Wnt-l expression domain and tested in transgenic mice embryos using a lac-Z reporter (pWEXP-lacZ (used as a control)) and a chick Shh reporter (pWEXP-CShh). The pWEXP-CShh construct contained two tandem head to tail copies of a chick Shh cDNA. The results of WEXP2-CShh transgenic studies are shown in Table 1.
Figure 8 is a model for anterioposterior limb patterning and the Zone of Polarizing Activity (ZPA), based on Saunders and Gasseling (1968). The left portion of the diagram
/O
schematizes a stage 20 limb bud. The somites are illustrated as blocks along the left margin of the limb bud; right portion of the same panel illustrates the mature wing. The hatched region on the posterior limb is the ZPA. Normally, the developed wing contains three digits II, III, and IV. The figure further shows the result of transplanting a ZPA from one limb bud to the anterior margin of another. The mature limb now contains six digits IV, III, II, II, III, and IV in a mirror-image duplication of the normal pattern. The large arrows in both panels represent the signal produced by the ZPA which acts to specify digit identity.
Figures 9A and 9B illustrate the comparison of zebrafish Shh (Z-Shh) and Drosophila hh (hh) amino acid sequences. Figure 9A is an alignment of zebrafish Shh and Drosophila hh amino acid sequences. Identical amino acids are linked by vertical bars. Dots indicate gaps introduced for optimal alignment. Putative transmembrane/signal peptide sequences are underlined (Kyte and Doolittle (1982) J Mol Biol 157:133-148). The position of exon boundaries in the Drosophila gene are indicated by arrowheads. The region of highest similarity between Z-Shh and hh overlaps exon 2. Figure 9B is a schematic comparison of Z- Shh and Drosophila hh. Black boxes indicate the position of the putative transmembrane/signal peptide sequences. relative to the amino-terminus. Sequence homologies were scored by taking into account the alignment of chemically similar amino acids and percentage of homology in the boxed regions is indicated.
Figure 10 is an alignment of partial predicted amino acid sequences from three 20 different zebrafish hh homologs. One of these sequences corresponds to Shh, while the other two define additional hh homologs in zebrafish, named hh(a) and hh(b). Amino acid identities among the three partial homologs are indicated by vertical bars.
Figure 11 is a schematic representations of chick and mouse Shh proteins. The putative signal peptides and Asn-linked glycosylation sites are shown. The numbers refer to 25 amino acid positions.
Figure 12 is a schematic representation of myc-tagged Shh constructs. The positions S of the c-myc epitope tags are shown, as is the predicted position of the proteolytic cleavage site. The shaded area following the signal peptide of the carboxy terminal tagged construct represents the region included in the Glutathione-S-transferase fusion protein used to generate antisera in rabbits.
Figure 13 is a schematic diagram of Shh processing. Illustrated are cleavage of the signal peptide (black box), glycosylation at the predicted Asn residue and the secondary proteolytic cleavage. The question marks indicate that the precise site of proteolytic cleavage has not been determined. The different symbols representing the carbohydrate moiety indicated maturation of this structure in the Golgi apparatus. The dashed arrow leading from
I/
the signal peptide cleaved protein indicates that secretion of this species may be an artifact of the incomplete proteolytic processing of Shh seen in Xenopus oocytes and cos cells.
Figure 14 is a schematic diagram of a model for the coordinated growth and patterning of the limb. Sonic is proposed to signal directly to the mesoderm to induce expression of the Hoxd and Bmp-2 genes. The induction of these mesodermal genes requires competence signals from the overlying AER. One such signal is apparently Fgf-4.
Expression of Fgf-4 in the AER can be induced by Sonic providing an indirect signaling pathway from Sonic to the mesoderm. FGFs also maintain expression of Sonic in the ZPA, thereby completing a positive feedback loop which controls the relative positions of the signaling centers. While Fgf-4 provides competence signals to the mesoderm, it also promotes mesodermal proliferation. Thus patterning of the mesoderm is dependent on the same signals which promote its proliferation. This mechanism inextricably integrates limb patterning with outgrowth.
Figure 15 is a schematic diagram of patterning of the Drosophila and vertebrate gut.
Regulatory interactions responsible for patterning of Drosophila midgut are compared to a model for patterning of the vertebrate hindgut based on expression data. Morphologic regional distinctions are indicated to the left (A and genes expressed in the visceral S 20 mesoderm are in the center panel, those in the gut lumenal endoderm are on the right.
HOMIHox gene expression domains are boxed. Regionally expressing secreted gene *products are indicated by lines. Arrows indicate activating interactions, barred lines, inhibiting interactions. Regulatory interactions in Drosophila gut have been established by genetic studies except for the relationship between dpp and hedgehog, which is 25 hypothesized based on their interactions in the Drosophila imaginal discs, hedgehog appears S to be a signal from the endoderm to the mesoderm, and that dpp is expressed in the mesoderm.
o Figure 16 is a schematic diagram of chromosomal locations of Ihh, Shh and Dhh in the mouse genome. The loci were mapped by interspecific backcross analysis. The segregation patterns of the loci and flanking genes in backcross animals that were typed for all loci are shown above the chromosome maps. For individual pairs of loci more animals were typed. Each column represents the chromosome identified in the backcross progeny that was inherited from the (C57BL/6J x M. spretus) Fl parent. The shaded boxes represent the presence of a C57BL/6J allele and white boxes represent the presence of a M spretus allele. The number of the offsprings inheriting each type of chromosome is listed at the bottom of each column. Partial chromosome linkage maps showing location of Ihh, Shh and Dhh in relation too linked genes is shown. The number of recombinant N 2 animals is presented over total number of N 2 animals typed to the left of the chromosome maps between each pair of loci. The recombinant frequencies, expressed as genetic distance in centimorgans one standard error) are also shown. When no recombination between loci was detected, the upper 95% confidence limit of the recombination distance is indicated in parentheses. Gene order was determined by minimizing the number of recombinant events required to explain the allele distribution patterns. The position of loci in human chromosomes can be obtained from GDB (Genome Data Base), a computerized database of human linkage information maintained by the William H. Welch Medical Library of the John Hopkins University (Baltimore, MD).
Detailed Description of the Invention Embryonic inductive signals are key regulatory proteins that function in vertebrate pattern formation, and are present in important signaling centers known to operate embryonically to define the organization of the vertebrate embryo. For example, these signaling structures include the notochord, a transient structure which initiates the formation of the nervous system and helps to define the different types of neurons within it. The notochord also regulates mesodermal patterning along the body axis. Another distinct group of cells having apparent signaling activity is the floorplate of the neural tube (the precursor of the spinal cord and brain) which also signals the differentiation of different nerve cell types.
20 It is also generally believed that the region of mesoderm at the bottom of the buds which form the limbs (called the Zone of Polarizing Activity or ZPA) operates as a signaling center by secreting a morphogen which ultimately produces the correct patterning of the developing limbs.
The present invention concerns the discovery that proteins encoded by a family of vertebrate genes, termed here hedgehog-related genes, comprise the signals produced by these embryonic patterning centers. As described herein, each of the disclosed vertebrate hedgehog (hh) homologs exhibits spatially and temporally restricted expression domains indicative of important roles in embryonic patterning. For instance, the results provided below indicate that vertebrate hh genes are expressed in the posterior limb bud, Hensen's node, the early notochord, the floor plate of the neural tube, the fore- and hindgut and their derivatives. These are all important signaling centers known to be required for proper patterning of surrounding embryonic tissues.
The Hedgehog family of vertebrate inter-cellular signaling molecules provided by the present invention consists of at least four members. Three of these members, herein referred to as Desert hedgehog (Dhh), Sonic hedgehog (Shh) and Indian hedgehog (Ihh), exist in all vertebrates, including fish, birds, and mammals. A fourth member, herein referred to as '3 Moonrat hedgehog (Mhh), appears specific to fish. According to the appended sequence listing, (see also Table 1) 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; and a human Ihh polypeptide is encoded by SEQ ID No:7.
Table 1 Guide to vertebrate hedgehog sequences Nucleotide Amino Acid Chicken Shh SEQ ID No. 1 SEQ ID No. 8 Mouse Dhh SEQ ID No. 2 SEQ ID No. 9 Mouse Ihh SEQ ID No. 3 SEQ ID No. Mouse Shh SEQ ID No. 4 SEQ ID No. 11 Zebrafish Shh SEQ ID No. 5 SEQ ID No. 12 Human Shh SEQ ID No. 6 SEQ ID No. 13 Human Ihh SEQ ID No. 7 SEQ ID No. 14 Certain of the vertebrate Hedgehog proteins (hh) of the present invention are defined by SEQ ID Nos:8-14 and can be cloned from vertebrate organisms including fish, avian and S mammalian sources. These proteins are distinct from the Drosophila protein referred to in Sto* the literature as a hedgehog protein which, for clarity, will be referred to hereinafter as "Droso* 15 HH". In addition to the sequence variation between the various hh homologs, the vertebrate 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. Further processing of the mature form apparently occurs in some instances to yield biologically active fragments of the protein. For instance, sonic hedgehog undergoes additional proteolytic processing to yield two peptides of approximately 19 kDa and 27 kDa, both of which are secreted. In addition to proteolytic fragmentation, the vertebrate hedgehog proteins can also be modified post-translationally, i such as by glycosylation, though bacterially produced unglycosylated) forms of the proteins apparently still maintain at least some of the activity of the native protein.
As described in the following examples, the cDNA clones provided by the present invention were first obtained by screening a mouse genomic library with a partial Drosophila hh cDNA clone Positive plaques were identified and one mouse clone was selected.
This clone was then used as a probe to obtain a genomic clone containing the full coding sequence of the Mouse Dhh gene. As described in the attached Examples, Northern blots and /q in situ hybridization demonstrated that Mouse Dhh is expressed in the testes, and potentially the ovaries, and is also associated with sensory neurons of the head and trunk. Dhh is clearly a secreted factor expressed by Sertoli cells in the male testes, which is required for maintenance of the male germ line as probably a mitotic and survival factor. Dhh mutants are male sterile. Furthermore, Dhh is expressed as one of the first signs of differentiation of the gonad, thus Dhh may be a target of the sex determining gene, Sry. Interestingly, no expression was detected on the nerve cell bodies themselves (only the axons), indicating that Dhh is likely produced by the Shwann cells.
In order to obtain cDNA clones encoding chicken hh genes, degenerate oligonucleotides were designed corresponding to the amino and carboxy ends of Drosophila hh exon 2. As described in the Examples below, these oligonucleotides were used to isolate PCR fragments from chicken genomic DNA. These fragments were then cloned and sequenced. Ten clones yielded two different hh homologs, chicken Dhh and chicken Shh.
The chicken Shh clone was then used to screen a stage 21/22 limb bud cDNA library which yielded a full length Shh clone.
In order to identify other vertebrate hedgehog homologs, the chicken clones (Dhh and Shh) were used to probe a genomic southern blot containing chicken DNA. As described below, genomic DNA was cut with various enzymes which do not cleave within the probe sequences. The DNA was run on a gel and transferred to a nylon filter. Probes were derived 20 by ligating each 220 bp clone into a concatomer and then labeling with a random primer kit.
The blots were hybridized and washed at low stringency. In each case, three hybridizing bands were observed following autoradiography, one of which was significantly more intense (a different band with each probe), indicating that there are at least three vertebrate hh genes.
Additional cDNA and genomic screens carried out have yielded clones of three hh homologs 25 from chickens and mice (Shh, Dhh and Ihh), and four hh homologs from zebrafish (Shh, Dhh, Ihh and Mhh). Weaker hybridization signals suggested that the gene family may be even larger. Moreover, a number of weakly hybridizing genomic clones have been isolated.
4 Subsequently, the same probes derived from chicken hedgehog homologs have been utilized to screen a human genomic library. PCR fragments derived from the human genomic library 30 were then sequenced, and PCR probes derived from the human sequences were used to screen human fetal cDNA libraries. Full-length cDNA encoding human sonic hedgehog protein (Shh) and partial cDNA encoding human Indian hedgehog protein (Ihh) were isolated from the fetal library, and represent a source of recombinant human hedgehog proteins.
To order to determine the expression patterns of the various vertebrate hh homologs, in situ hybridizations were performed in developing embryos of chicken, mice and fish. As described in the Examples below, the resulting expression patterns of each hh homolog were similar across each species and revealed that hh genes are expressed in a number of important embryonic signaling centers. For example, Shh is expressed in Hensen's node, the notochord, the ventral floorplate of the developing neural tube, and the ZPA at the base of the limb buds.
Shh is also expressed in differntiated motor neurons in the embryonic mouse (at 11.5 days post fertilization), therefore, Shh may play a role in later stages of motor neuron development, perhaps in proliferation, but more likely in survival of this cell population. Ihh is expressed in the embryonic yolksac and hindgut, and appear also to be involved in chondrogenesis; Dhh is expressed in the testes; and Mhh (only in zebrafish) is expressed in the notochord and in certain cranial nerves.
Furthermore, experimental evidence indicates that certain hedgehog proteins initiate expression of secondary signaling molecules, including Bmp-2 (a TGF-p relative) in the mesoderm and Fgf-4 in the ectoderm. The mesoderm requires ectodermally-derived competence factor(s), which include Fgf-4, to activate target gene expression in response to hedgehog signaling. The expression of, for example, Sonic and Fgf-4 is coordinately regulated by a positive feedback loop operating between the posterior mesoderm and the overlying AER, which is the ridge ofpseudostratified epithelium extending antero-posteriorly along the distal margin of the bud. These data provide a basis for understanding the integration of growth and patterning in the developing limb which can have important implications in the treatment of bone disorders described in greater detail herein.
To determine the role hedgehog proteins plays in inductive interactions between the 20 endoderm and mesoderm, which are critical to gut morphogenesis, in situ hybridizations and recombinant retroviral injections were performed in developing chick embryos. The ventral mesoderm is induced to undergo gut-specific differentiation by the adjacent endoderm. As described in Examples below, at the earliest stages of chick gut formation Shh is expressed by the endoderm, and BMP-4 (a TGF-P relative) is expressed in the adjacent visceral mesoderm.
25 Ectopic expression of Sonic is sufficient to induce expression of BMP-4 in visceral mesoderm, suggesting that Sonic serves as an inductive signal from the endoderm to the mesoderm. Subsequent organ-specific endodermal differentiation depends on regional inductive signal from the visceral mesoderm. Hox genes are expressed in the undifferentiated chick hind gut mesoderm with boundaries corresponding to morphologic borders, suggesting 30 a role in regulating gut morphogenesis.
Accordingly, certain aspects of the present invention relate to nucleic acids encoding vertebrate hedgehog proteins, the hedgehog proteins themselves, antibodies immunoreactive with hh proteins, and preparations of such compositions. Moreover, the present invention provides diagnostic and therapeutic assays and reagents for detecting and treating disorders involving, for example, aberrant expression of vertebrate hedgehog homologs. In addition, drug discovery assays are provided for identifying agents which can modulate the binding of vertebrate hedgehog homologues to hedgehog-binding moieties (such as hedgehog receptors, ligands, or other extracellular matrix components). Such agents can be useful therapeutically to alter the growth and/or differentiation of a cell. Other aspects of the invention are described below or will be apparent to those skilled in the art in light of the present disclosure.
For convenience, certain terms employed in the specification, examples, and appended claims are collected here.
As used herein, the term "nucleic acid" refers to polynucleotides such as deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA). The term should also be understood to include, as equivalents, analogs of either RNA or DNA made from nucleotide analogs, and, as applicable to the embodiment being described, single (sense or antisense) and double-stranded polynucleotides.
As used herein, the term "gene" or "recombinant gene" refers to a nucleic acid comprising an open reading frame encoding one of the vertebrate hh polypeptides of the present invention, including both exon and (optionally) intron sequences. A "recombinant gene" refers to nucleic acid encoding a vertebrate hh polypeptide and comprising vertebrate hh-encoding exon sequences, though it may optionally include intron sequences which are either derived from a chromosomal vertebrate hh gene or from ar unrelated chromosomal gene. Exemplary recombinant genes encoding the subject vertebrate hh polypeptide are represented by SEQ ID No:l, SEQ ID No:2, SEQ ID No:3, SEQ ID No:4, SEQ ID S: 20 SEQ ID No:6 or SEQ ID No:7. The term "intron" refers to a DNA sequence present in a given vertebrate hh gene which is not translated into protein and is generally found between exons.
As used herein, the term "transfection" means the introduction of a nucleic acid, an expression vector, into a recipient cell by nucleic acid-mediated gene transfer.
"Transformation", as used herein, refers to a process in which a cell's genotype is changed as a result of the cellular uptake of exogenous DNA or RNA, and, for example, the transformed cell expresses a recombinant form of a vertebrate hh polypeptide or, where anti-sense expression occurs from the transferred gene, the expression of a naturally-occurring form of the vertebrate hh protein is disrupted.
S 30 As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of preferred vector is an episome, a nucleic acid capable of extra-chromosomal replication. Preferred vectors are those capable of autonomous replication and/expression of nucleic acids to which they are linked. Vectors capable of directing the expression of genes to which they are operatively linked are referred to herein as "expression vectors". In general, expression vectors of utility in recombinant DNA techniques are often in the form of "plasmids" which refer generally to 17 circular double stranded DNA loops which, in their vector form are not bound to the chromosome. In the present specification, "plasmid" and "vector" are used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors which serve equivalent functions and which become known in the art subsequently hereto.
"Transcriptional regulatory sequence" is a generic term used throughout the specification to refer to DNA sequences, such as initiation signals, enhancers, and promoters, which induce or control transcription of protein coding sequences with which they are operably linked. In preferred embodiments, transcription of one of the recombinant vertebrate hedgehog genes is under the control of a promoter sequence (or other transcriptional regulatory sequence) which controls the expression of the recombinant gene in a cell-type in which expression is intended. It will also be understood that the recombinant gene can be under the control of transcriptional regulatory sequences which are the same or which are different from those sequences which control transcription of the naturallyoccurring forms of hedgehog proteins.
As used herein, the term "tissue-specific promoter" means a DNA sequence that serves as a promoter, regulates expression of a selected DNA sequence operably linked to the promoter, and which effects expression of the selected DNA sequence in specific cells of a tissue, such as cells of neural origin, e.g. neuronal cells. The term also covers so-called "leaky" promoters, which regulate expression of a selected DNA primarily in one tissue, but cause expression in other tissues as well.
As used herein, a "transgenic animal" is any animal, preferably a non-human mammal, bird or an amphibian, in which one or more of the cells of the animal contain heterologous nucleic acid introduced by way of human intervention, such as by transgenic 25 techniques well known in the art. The nucleic acid is introduced into the cell, directly or indirectly by introduction into a precursor of the cell, by way of deliberate genetic manipulation, such as by microinjection or by infection with a recombinant virus. The term genetic manipulation does not include classical cross-breeding, or in vitro fertilization, but rather is directed to the introduction of a recombinant DNA molecule. This molecule may be 30 integrated within a chromosome, or it may be extrachromosomally replicating DNA. In the typical transgenic animals described herein, the transgene causes cells to express a recombinant form of one of the vertebrate hh proteins, e.g. either agonistic or antagonistic forms. However, transgenic animals in which the recombinant vertebrate hh gene is silent are also contemplated, as for example, the FLP or CRE recombinase dependent constructs described below. The "non-human animals" of the invention include vertebrates such as rodents, non-human primates, sheep, dog, cow, chickens, amphibians, reptiles, etc. Preferred non-human animals are selected from the rodent family including rat and mouse, most /9 preferably mouse, though transgenic amphibians, such as members of the Xenopus genus, and transgenic chickens can also provide important tools for understanding and identifying agents which can affect, for example, embryogenesis and tissue formation. The term "chimeric animal" is used herein to refer to animals in which the recombinant gene is found, or in which the recombinant is expressed in some but not all cells of the animal. The term "tissuespecific chimeric animal" indicates that one of the recombinant vertebrate hh genes is present and/or expressed in some tissues but not others.
As used herein, the term "transgene" means a nucleic acid sequence (encoding, e.g., one of the vertebrate hh polypeptides), which is partly or entirely heterologous, foreign, to the transgenic animal or cell into which it is introduced, or, is homologous to an endogenous gene of the transgenic animal or cell into which it is introduced, but which is designed to be inserted, or is inserted, into the animal's genome in such a way as to alter the genome of the cell into which it is inserted it is inserted at a location which differs from that of the natural gene or its insertion results in a knockout). A transgene can include one or more transcriptional regulatory sequences and any other nucleic acid, such as introns, that may be necessary for optimal expression of a selected nucleic acid.
As is well known, genes for a particular polypeptide may exist in single or multiple copies within the genome of an individual. Such duplicate genes may be identical or may have certain modifications, including nucleotide substitutions, additions or deletions, which 20 all still code for polypeptides having substantially the same activity. The term "DNA sequence encoding a vertebrate hh polypeptide" may thus refer to one or more genes within a particular individual. Moreover, certain differences in nucleotide sequences may exist between individual organisms, which are called alleles. Such allelic differences may or may not result in differences in amino acid sequence of the encoded polypeptide yet still encode a 25 protein with the same biological activity.
"Homology" refers to sequence similarity between two peptides or between two nucleic acid molecules. Homology can 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 base or amino acid, then the molecules are 30 homologous at that position. A degree of homology 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 percent identity, with one of the vertebrate hh sequences of the present invention.
"Cells," "host cells" or "recombinant host cells" are terms used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in /9 succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
A "chimeric protein" or "fusion protein" is a fusion of a first amino acid sequence encoding one of the subject vertebrate hh polypeptides with a second amino acid sequence defining a domain foreign to and not substantially homologous with any domain of one of the vertebrate hh proteins. 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. In general, a fusion protein can be represented by the general formula Xhh-Y, wherein hh represents a portion of the protein which is derived from one of the vertebrate hh proteins, and X and Y are independently absent or represent amino acid sequences which are not related to one of the vertebrate hh sequences in an organism, including naturally occurring mutants.
As used herein, the terms "transforming growth factor-beta" and "TGF-p" denote a family of structurally related paracrine polypeptides found ubiquitously in vertebrates, and prototypic of a large family of metazoan growth, differentiation, and morphogenesis factors (see, for review, Massaque et al. (1990) Ann Rev Cell Biol 6:597-641; and Sporn et al. (1992) i J Cell Biol 119:1017-1021). Included in this family are the "bone morphogenetic proteins" 20 or "BMPs", which refers to proteins isolated from bone, and fragments thereof and synthetic peptides which are capable of inducing bone deposition alone or when combined with appropriate cofactors. Preparation of BMPs, such as BMP-1, and is described in, for example, PCT publication WO 88/00205. Wozney (1989) Growth Fact Res 1:267-280 describes additional BMP proteins closely related to BMP-2, and which have been designated 25 BMP-5, and PCT publications W089/09787 and W089/09788 describe a protein called now known to be BMP-7. Other BMPs are known in the art.
The term "isolated" as also used herein with respect to nucleic acids, such as DNA or *RNA, refers to molecules separated from other DNAs, or RNAs, respectively, that are present in the natural source of the macromolecule. For example, an isolated nucleic acid encoding 30 one of the subject vertebrate hh polypeptides preferably includes no more than 10 kilobases (kb) of nucleic acid sequence which naturally immediately flanks the vertebrate hh gene in genomic DNA, more preferably no more than 5kb of such naturally occurring flanking sequences, and most preferably less than 1.5kb of such naturally occurring flanking sequence.
The term isolated as used herein also refers to a nucleic acid or peptide that is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized.
.2 0 Moreover, an "isolated nucleic acid" is meant to include nucleic acid fragments which are not naturally occurring as fragments and would not be found in the natural state.
As described below, one aspect of the invention pertains to isolated nucleic acids comprising the nucleotide sequences encoding vertebrate hh homologues, and/or equivalents of such nucleic acids. The term nucleic acid as used herein is intended to include fragments as equivalents. The term equivalent is understood to include nucleotide sequences encoding functionally equivalent hh polypeptides or functionally equivalent peptides having an activity of a vertebrate hh protein such as described herein. Equivalent nucleotide sequences will include sequences that differ by one or more nucleotide substitutions, additions or deletions, such as allelic variants; and will, therefore, include sequences that differ from the nucleotide sequence of the vertebrate hh cDNAs shown in SEQ ID Nos:1-7 due to the degeneracy of the genetic code. Equivalents will also include nucleotide sequences that hybridize under stringent conditions equivalent to about 20-27 0 C below the melting temperature (Tm) of the DNA duplex formed in about 1M salt) to the nucleotide sequences represented in SEQ ID Nos: 1-7. In one embodiment, equivalents will further include nucleic acid sequences derived from and evolutionarily related to, a nucleotide sequences shown in any of SEQ ID Nos:1-7.
Moreover, it will be generally appreciated that, under certain circumstances, it may be advantageous to provide homologs of one of the subject hedgehog polypeptides which function in a limited capacity as one of either an hh agonist or an hh antagonist, in order to 20 promote or inhibit only a subset of the biological activities of the naturally-occurring form of the protein. Thus, specific biological effects can be elicited by treatment with a homolog of limited function, and with fewer side effects relative to treatment with agonists or antagonists which are directed to all of the biological activities of naturally occurring forms of hedgehog proteins.
Homologs of one of the subject hedgehog proteins can be generated by mutagenesis, such as by discrete point mutation(s), or by truncation. For instance, mutation can give rise i to homologs which retain substantially the same, or merely a subset, of the biological activity of the hh polypeptide from which it was derived. Alternatively, antagonistic forms of the protein can be generated which are able to inhibit the function of the naturally occurring form 30 of the protein, such as by competitively binding to an hh receptor.
Polypeptides referred to herein as having an activity of a vertebrate hh protein are defined as peptides that have an amino acid sequence corresponding to all or a portion of the amino acid sequences of a vertebrate hh proteins shown in any of 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 and which have at least one biological activity of a vertebrate hh protein. Examples of such biological activity of a vertebrate hh protein include the ability to induce (or otherwise
Z/
modulate) formation and differentiation of the head, limbs, lungs, central nervous system (CNS), or mesodermal patterning of developing vertebrate embryos. In preferred embodiments, the biological activity can comprise an ability to regulate neurogenesis, such as a motor neuron inducing activity, a neuronal differentiation inducing activity, or a neuronal survival promoting activity. Hedgehog proteins of the present invention can also have biological activities which include an ability to regulate organogensis, such as through the ability to influence limb patterning, by, for example, skeletogenic activity. The biological activity associated with the hedgehog proteins of the present invention can also include the ability to induce stem cell or germ cell differentiation, including the ability to induce differentiation of chondrocytes or an involvement in spermatogenesis. Hedgehog proteins of the present invention can also be characterized in terms of biological activities which include: an ability to modulate proliferation, survival and/or differentiation of mesodermally-derived tissue, such as tissue derived from dorsal mesoderm; the ability to modulate proliferation, survival and/or differentiation of ectodermally-derived tissue, such as tissue derived from the neural tube, neural crest, or head mesenchyme; the ability to modulate proliferation, survival and/or differentiation of endodermally-derived tissue, such as tissue derived from the primitive gut Moreover, as described in the Examples below, the subject hedgehog proteins have the ability to induce expression of secondary signaling molecules, such as members of the Transforming Growth Factor p (TGFp) family, including bone morphogenic proteins, e.g.
20 BMP-2 and LAiP-4, as well hs members of the fibroblast growth factor (FGF) family, such as Fgf-4. Other biological activities of the subject hedgehog proteins are described herein or will be reasonably apparent to those skilled in the art. According to the present invention, a polypeptide has biological activity if it is a specific agonist or antagonist of a naturallyoccurring form of a vertebrate hedgehog protein.
Preferred nucleic acids encode a vertebrate hedgehog polypeptide comprising an amino acid sequence at least 60% homologous, more preferably 70% homologous and most preferably 80% homologous 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 0 preferably at least about 95%, and most preferably at least about 98-99% homology with an amino acid sequence represented in one of SEQ ID Nos:8-14 are also within the scope of the invention. In one embodiment, the nucleic acid is a cDNA encoding a peptide having at least i. one activity of the subject vertebrate hh polypeptide. Preferably, the nucleic acid includes all or a portion of the nucleotide sequence corresponding to the coding region of SEQ ID Nos: 7.
Another aspect of the invention provides a nucleic acid which hybridizes under high or low stringency conditions to a nucleic acid represented by one of SEQ ID Nos:l-7.
Appropriate stringency conditions which promote DNA hybridization, for example, 6.0 x L2.
sodium chloride/sodium citrate (SSC) at about 45 0 C, followed by a wash of 2.0 x SSC at 0 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. For example, the salt concentration in the wash step can be selected from a low stringency of about 2.0 x SSC at 50 0 C to a high stringency of about 0.2 x SSC at 50 0 C. In addition, the temperature in the wash step can be increased from low stringency conditions at room temperature, about 22 0 C, to high stringency conditions at about 65 0
C.
Nucleic acids, having a sequence that differs from the nucleotide sequences shown in one of SEQ ID No:l, SEQ ID No:2, SEQ ID No:3, SEQ ID No:4, SEQ ID No:5, SEQ ID No:6 or SEQ ID No:7 due to degeneracy in the genetic code are also within the scope of the invention. Such nucleic acids encode functionally equivalent peptides a peptide having a biological activity of a vertebrate hh polypeptide) but differ in sequence from the sequence shown in the sequence listing due to degeneracy in the genetic code. For example, a number of amino acids are designated by more than one triplet. Codons that specify the same amino acid, or synonyms (for example, CAU and CAC each encode histidine) may result in "silent" mutations which do not affect the amino acid sequence of a vertebrate hh polypeptide.
However, it is expected that DNA sequence polymorphisms that do lead to changes in the amino acid sequences of the subject hh polypeptides will exist among vertebrates. One skilled in the art will appreciate that these variations in one or more nucleotides (up to about 20 3-5% of the nucleotides) of the nucleic acids encoding polypeptides having an activity of a vertebrate hh polypeptide may exist among individuals of a given species due to natural allelic variation.
Fragments of the nucleic acids encoding an active portion of the vertebrate hedgehog proteins are also within the scope of the invention. As used herein, a hedgehog gene 25 fragment refers to a nucleic acid having fewer nucleotides than the nucleotide sequence encoding the entire amino acid sequence of a vertebrate hh protein 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, yet which (preferably) encodes a peptide which retains some biological activity of the full length protein, e.g. the fragment retains the ability to induce formation and differentiation of the head, limbs, lungs, central nervous system (CNS), or mesodermal patterning of developing vertebrate embryo. Nucleic acid fragments within the scope of the present invention include those capable of hybridizing under high or low stringency conditions with nucleic acids from other species for use in screening protocols to detect other hedgehog homologs, as well as those capable of hybridizing with nucleic acids from human specimens for use in detecting the presence of a nucleic acid encoding a hedgehog protein, including alternate isoforms, e.g. mRNA splicing variants. Nucleic acids within the scope of the invention may also contain linker sequences, modified restriction endonuclease sites and 23 other sequences useful for molecular cloning, expression or purification of recombinant forms of the subject hh polypeptides.
As indicated by the examples set out below, hedgehog protein-encoding nucleic acids can be obtained from mRNA present in any of a number of eukaryotic cells. It should also be possible to obtain nucleic acids encoding vertebrate hh polypeptides of the present invention from genomic DNA obtained from both adults and embryos. For example, a gene encoding a hh protein can be cloned from either a cDNA or a genomic library in accordance with protocols described herein, as well as those generally known to persons skilled 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 vertebrate hh protein can also be cloned using established polymerase chain reaction techniques in accordance with the nucleotide sequence information provided by the invention. The nucleic acid of the invention can be DNA or RNA. A preferred nucleic acid is a cDNA represented by a sequence selected from the group consisting of SEQ ID Nos:l-7.
Another aspect of the invention relates to the use of the isolated nucleic acid in "antisense" therapy. As used herein, "antisense" therapy refers to administration or in situ generation of oligonucleotide probes or their derivatives which specifically hybridizes (e.g.
20 binds) under cellular conditions, with the cellular mRNA and/or genomic DNA encoding one or more of the subject hedgehog proteins so as to 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. In general, "antisense" therapy refers to S 25 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 cellular mRNA which encodes a vertebrate hh protein. Alternatively, 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 vertebrate hh gene. Such oligonucleotide probes are preferably modified oligonucleotide which are resistant to endogenous nucleases, e.g. exonucleases and/or endonucleases, and is therefore stable in vivo. Exemplary 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 annrnah,'. tn -2Y 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.
Accordingly, the modified oligomers of the invention are useful in therapeutic, diagnostic, and research contexts. In therapeutic applications, the oligomers are utilized in a manner appropriate for antisense therapy in general. For such therapy, the oligomers of the invention can be formulated for a variety of loads of administration, including systemic and topical or localized administration. Techniques and formulations generally may be found in Remmington's Pharmaceutical Sciences, Meade Publishing Co., Easton, PA. For systemic administration, injection is preferred, including intramuscular, intravenous, intraperitoneal, and subcutaneous for injection, the oligomers of the invention can be formulated in liquid solutions, preferably in physiologically compatible buffers such as Hank's solution or Ringer's solution. In addition, the oligomers may be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms are also included.
Systemic administration can also be by transmucosal or transdermal means, or the compounds can be administered orally. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration bile salts and fusidic acid derivatives. In addition, detergents may be used to 20 facilitate permeation. Transmucosal administration may be through nasal sprays or using suppositories. For oral administration, the oligomers are formulated into conventional oral administration forms such as capsules, tablets, and tonics. For topical administration, the oligomers of the invention are formulated into ointments, salves, gels, or creams as generally known in the art.
i: 25 In addition to use in therapy, the oligomers of the invention may be used as diagnostic reagents to detect the presence or absence of the target DNA or RNA sequences to which they specifically bind. Such diagnostic tests are described in further detail below.
ma Likewise, the antisense constructs of the present invention, by antagonizing the normal biological activity of one of the hedgehog proteins, can be used in the manipulation 30 of tissue, e.g. tissue differentiation, both in vivo and in ex vivo tissue cultures.
Also, the anti-sense techniques microinjection of antisense molecules, or transfection with plasmids whose transcripts are anti-sense with regard to an hh mRNA or gene sequence) can be used to investigate role of hh in developmental events, as well as the normal cellular function of hh in adult tissue. Such techniques can be utilized in cell culture, but can also be used in the creation of transgenic animals.
2 This invention also provides expression vectors containing a nucleic acid encoding a vertebrate hh 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. Regulatory sequences are art-recognized and are selected to direct expression of the subject vertebrate hh proteins. Accordingly, the term 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). For instance, 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 vertebrate hh polypeptides of this invention. 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 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 X the control regions for fd coat protein, the promoter for 3-phosphoglycerate kinase or other glycolytic enzymes, the promoters of acid phosphatase, Pho5, the promoters of the yeast ac-mating factors, the polyhedron promoter of the baculovirus system and other sequences known to control the 20 expression of genes of prokaryotic or eukaryotic cells or their viruses, and various combinations thereof. It should be understood that 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. In one embodiment, the expression vector includes a recombinant gene encoding a peptide having an agonistic activity of a subject hedgehog polypeptide, or alternatively, encoding a peptide which is an antagonistic form of the hh protein. Such expression vectors can be used to transfect cells and thereby produce polypeptides, including fusion proteins, encoded by nucleic acids as described herein.
30 Moreover, 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 one of the subject vertebrate hedgehog proteins. Thus, another aspect of the invention features expression vectors for in vivo or in vitro transfection and expression of a vertebrate hh polypeptide in particular cell types so as to reconstitute the function of, or alternatively, abrogate the function of hedgehog-induced signaling in a tissue in which the naturally-occurring form of the protein is misexpressed; or to deliver a form of the protein which alters differentiation of tissue, or which inhibits neoplastic transformation.
Expression constructs of the subject vertebrate hh polypeptide, and mutants thereof, 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 subject gene 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.
antibody conjugated), polylysine conjugates, gramacidin S, artificial viral envelopes or other such intracellular carriers, as well as direct injection of the gene construct or CaPO 4 precipitation carried out in vivo. It will be appreciated that because 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. Furthermore, it will be recognized that 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. Infection of cells with a viral vector has the advantage that a 20 large proportion of the targeted cells can receive the nucleic acid. Additionally, molecules encoded within the viral vector, 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, 25 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. (1990) Blood 76:271). Thus, recombinant retrovirus can be constructed in which part of the retroviral coding sequence (gag, pol, env) has been replaced by nucleic acid encoding one of the subject proteins rendering 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 27) 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. Examples of suitable retroviruses include pLJ, pZIP, pWE and pEM which are well known to those skilled in the art. Examples of suitable packaging virus lines for preparing both ecotropic and amphotropic retroviral systems include WCrip, iCre, y2 and WAm. Retroviruses have been used to introduce a variety of genes into many different cell types, including neuronal 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. USA 85:3014-3018; Armentano et al. (1990) Proc. Natl. Acad Sci. USA 87:6141-6145; Huber et al. (1991) Proc. Natl. Acad- Sci. USA 88:8039-8043; Ferry et al. (1991) Proc. Natl. Acad. Sci. USA 88:8377-8381; Chowdhury et al. (1991) Science 254:1802-1805; van Beusechem et al. (1992) Proc. Natl.
Acad Sci. USA 89:7640-7644; Kay et al. (1992) Human Gene Therapy 3:641-647; Dai et al.
(1992) Proc. Natl. Acad. Sci. USA 89:10892-10895; Hwu et al. (1993) Immunol. 150:4104- 4115; U.S. Patent No. 4,868,116; U.S. Patent No. 4,980,286; PCT Application
WO
89/07136; PCT Application WO 89/02468; PCT Application WO 89/05345; and PCT Application WO 92/07573).
Furthermore, it has been shown that it is possible to limit the infection spectrum of retroviruses and consequently of retroviral-based vectors, by modifying the viral packaging 20 proteins on the surface of the viral particle (see, for example PCT publications W093/25234 [and W094/06920). For instance, 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 Chem 266:14143-14146).
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 single-chain antibody/env fusion proteins). This technique, while useful to limit or otherwise direct the infection to certain tissue types, can also be used to convert an 30 ecotropic vector in to an amphotropic vector.
Moreover, use of retroviral gene delivery can be further enhanced by the use of tissueor cell-specific transcriptional regulatory sequences which control expression of the hh gene s of the retroviral vector.
Another viral gene delivery system useful in the present invention utilizes adenovirusderived 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) BioTechniaou.. f6i-cl 28' Rosenfeld et al. (1991) Science 252:431-434; and Rosenfeld et al. (1992) Cell 68:143-155.
Suitable adenoviral vectors derived from the adenovirus strain Ad type 5 d1324 or other strains of adenovirus Ad2, Ad3, Ad7 etc.) 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 airway epithelium (Rosenfeld et al.
(1992) cited supra), endothelial cells (Lemarchand et al. (1992) Proc. Natl. Acad. Sci. USA 89:6482-6486), hepatocytes (Herz and Gerard (1993) Proc. Natl. Acad. Sci. USA 90:2812- 2816) and muscle cells (Quantin et al. (1992) Proc. Natl. Acad. Sci. USA 89:2581-2584).
Furthermore, 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.
Additionally, 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 retroviral DNA). Moreover, 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). Most replication-defective 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, Jones et al.
0: 20 (1979) Cell 16:683; Berkner et 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 ElA promoter, the major late promoter (MLP) and associated leader sequences, the E3 promoter, or exogenously added promoter sequences.
Yet another viral vector system useful for delivery of one of the subject vertebrate hh genes is the adeno-associated virus (AAV). Adeno-associated virus is a naturally occurring defective virus that requires another virus, such as an adenovirus or a herpes virus, as a helper virus for efficient replication and a productive life cycle. (For a review see Muzyczka et al.
Curr. Topics in Micro. and Immunol. (1992) 158:97-129). It is also one of the few viruses 30 that may integrate its DNA into non-dividing cells, and exhibits a high frequency of stable integration (see for example Flotte et al. (1992) Am. J. Respir. Cell. Mol. Biol. 7:349-356; Samulski et al. (1989) J. Virol. 63:3822-3828; and McLaughlin et al. (1989) J. Virol.
62:1963-1973). Vectors containing as little as 300 base pairs of AAV can be packaged and can integrate. Space for exogenous DNA is limited to about 4.5 kb. An AAV vector such as that described in Tratschin et al. (1985) Mol. Cell. Biol. 5:3251-3260 can be used to introduce DNA into cells. A variety of nucleic acids have been introduced into different cell types using AAV vectors (see for example Hermonat et al. (1984) Proc. Natl. Acad Sci. USA 81:6466-6470; Tratschin et al. (1985) Mol. Cell. Biol. 4:2072-2081; Wondisford et al. (1988) Mol Endocrinol. 2:32-39; Tratschin et al. (1984) J. Virol 51:611-619; and Flotte et al.
(1993)J. Biol. Chem. 268:3781-3790).
In addition to viral transfer methods, such as those illustrated above, non-viral methods can also be employed to cause expression of a subject 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. In preferred embodiments, non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject hh polypeptide gene by the targeted cell.
Exemplary gene delivery systems of this type include liposomal derived systems, poly-lysine conjugates, and artificial viral envelopes.
In clinical settings, 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. For instance, 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. In other embodiments, initial delivery of the recombinant gene is more limited with introduction into the animal being quite localized. For example, the gene delivery vehicle can be introduced by catheter (see U.S. Patent 5,328,470) or by stereotactic injection Chen et al. (1994) PNAS 91: 3054-3057). A vertebrate hh gene, such as any one of the clones represented in the group consisting of SEQ ID NO:1-7, can be delivered in a gene therapy construct by electroporation using techniques described, for example, by Dev et al. ((1994) Cancer Treat Rev 20:105-115).
25 The pharmaceutical preparation of the gene therapy construct can consist essentially co* of the gene delivery system in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is .imbedded. Alternatively, where the complete gene delivery system can be produced intact from recombinant cells, e.g. retroviral vectors, the pharmaceutical preparation can comprise one or more cells which produce the gene delivery system.
Another aspect of the present invention concerns recombinant forms of the hedgehog proteins. Recombinant polypeptides preferred by the present invention, in addition to native hedgehog proteins, are at least 60% homologous, more preferably 70% homologous and most preferably 80% homologous with an amino acid sequence represented by any of SEQ ID Nos:8-14. Polypeptides which possess an activity of a hedgehog protein either agonistic or antagonistic), and which are at least 90%, more preferably at least 95%, and most preferably at least about 98-99% homologous with a sequence selected from the group consisting of SEQ ID Nos:8-14 are also within the scope of the invention.
The term "recombinant protein" refers to a polypeptide of the present invention which is produced by recombinant DNA techniques, wherein generally, DNA encoding a vertebrate hh polypeptide is inserted into a suitable expression vector which is in turn used to transform a host cell to produce the heterologous protein. Moreover, 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 present invention further pertains to recombinant forms of one of the subject hedgehog polypeptides which are encoded by genes derived from a vertebrate organism, particularly a mammal a human), and which have amino acid sequences evolutionarily related to the hedgehog proteins represented in SEQ ID Nos:8-14. Such recombinant hh polypeptides preferably are capable of functioning in one of either role of an agonist or antagonist of at least one biological activity of a wild-type ("authentic") hedgehog protein of the appended sequence listing. The term "evolutionarily related to", with respect to amino acid sequences of vertebrate hedgehog proteins, refers to both polypeptides having amino acid sequences which have arisen naturally, and also to mutational variants of vertebrate hh polypeptides which are derived, for example, by combinatorial mutagenesis. Such evolutionarily derived hedgehog proteins polypeptides preferred by the present invention are at least 60% homologous, more preferably 70% homologous and most preferably homologous with the amino acid sequence selected from the group consisting of SEQ ID Nos:8-14. Polypeptides having at least about 90%, more preferably at least about 95%, and 25 most preferably at least about 98-99% homology with a sequence selected from the group consisting of SEQ ID Nos:8-14 are also within the scope of the invention.
S.
The present invention further pertains to methods of producing the subject hedgehog polypeptides. For example, 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 vertebrate hh polypeptide. Alternatively, the peptide may be retained cytoplasmically by removing the signal peptide sequence from the recombinant hh gene and the cells harvested, lysed and the protein isolated. A cell culture includes host cells, media 35 and other byproducts. Suitable media for cell culture are well known in the art. The recombinant hh polypeptide can be isolated from cell culture medium, host cells, or both using techniques known in the art for purifying proteins including ion-exchanee 3/ chromatography, gel filtration chromatography, ultrafiltration, electrophoresis, and immunoaffinity purification with antibodies specific for such peptide. In a preferred embodiment, the recombinant hh polypeptide is a fusion protein containing a domain which facilitates its purification, such as an hh/GST fusion protein.
This invention also pertains to a host cell transfected to express a recombinant form of the subject hedgehog polypeptides. The host cell may be any prokaryotic or eukaryotic cell.
Thus, a nucleotide sequence derived from the cloning of vertebrate hedgehog proteins, encoding all or a selected portion of the full-length protein, can be used to produce a recombinant form of a vertebrate hh polypeptide via microbial or eukaryotic cellular processes. Ligating the polynucleotide sequence into a gene construct, such as an expression vector, and transforming or transfecting into hosts, either eukaryotic (yeast, avian, insect or mammalian) or prokaryotic (bacterial cells), are standard procedures used in producing other well-known proteins, e.g. insulin, interferons, human growth hormone, IL-1, IL-2, and the like. Similar procedures, or modifications thereof, can be employed to prepare recombinant hedgehog polypeptides by microbial means or tissue-culture technology in accord with the subject invention.
The recombinant hedgehog genes can be produced by ligating nucleic acid encoding an hh protein, jr 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 hh polypeptides include plasmids and other vectors. For instance, 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.
A number of vectors exist for the expression of recombinant proteins in yeast. For 25 instance, 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 the presence of the pBR322 ori, and in S. cerevisiae due to the replication 30 determinant of the yeast 2 micron plasmid. In addition, drug resistance markers such as ampicillin can be used. In an illustrative embodiment, an hh 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:1-7.
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, 32 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. Some of these vectors are modified with sequences from bacterial plasmids, such as pBR322, to facilitate replication and drug resistance selection in both prokaryotic and eukaryotic cells. Alternatively, 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. The various methods employed in the preparation of the plasmids and transformation of host organisms are well known in the art. For other suitable expression systems for both prokaryotic and eukaryotic cells, as well as general recombinant procedures, see Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1989) Chapters 16 and 17.
In some instances, it may be desirable to express the recombinant hedgehog polypeptide by the use of a baculovirus expression system. Examples of such baculovirus expression systems include pVL-derived vectors (such as pVL1392, pVL1393 and pVL941), pAcUW-derived vectors (such as pAcUW1), and pBlueBac-derived vectors (such as the B-gal containing pBlueBac III).
When it is desirable to express only a portion of an hh protein, such as a form lacking a portion of the N-terminus, i.e. a truncation mutant which lacks the signal peptide, it may be necessary to add a start codon (ATG) to the oligonucleotide fragment containing the desired sequence to be expressed. It is well known in the art that a methionine at the N-terminal position can be enzymatically cleaved by the use of the enzyme methionine aminopeptidase (MAP). MAP has been cloned from E. coli (Ben-Bassat et al. (1987) J. Bacteriol. 169:751- S 757) and Salmonella typhimurium and its in vitro activity has been demonstrated on 25 recombinant proteins (Miller et al. (1987) PNAS 84:2718-1722). Therefore, removal of an Nterminal methionine, if desired, can be achieved either in vivo by expressing hedgehogderived polypeptides in a host which produces MAP E. coli or CM89 or S. cerevisiae), or in vitro by use of purified MAP procedure of Miller et al., supra).
Alternatively, the coding sequences for the polypeptide can be incorporated as a part of a fusion gene including a nucleotide sequence encoding a different polypeptide. This type Sof expression system can be useful under conditions where it is desirable to produce an immunogenic fragment of a hedgehog protein. For example, the VP6 capsid protein of rotavirus can be used as an immunologic carrier protein for portions of the hh polypeptide, either in the monomeric form or in the form of a viral particle. The nucleic acid sequences corresponding to the portion of a subject hedgehog protein to which antibodies are to be raised can be incorporated into a fusion gene construct which includes coding sequences for a late vaccinia virus structural protein to produce a set of recombinant viruses expressing 33 fusion proteins comprising hh epitopes as part of the virion. It has been demonstrated with the use of immunogenic fusion proteins utilizing the Hepatitis B surface antigen fusion proteins that recombinant Hepatitis B virions can be utilized in this role as well. Similarly, chimeric constructs coding for fusion proteins containing a portion of an hh protein and the poliovirus capsid protein can be created to enhance immunogenicity of the set of polypeptide antigens (see, for example, EP Publication No: 0259149; and Evans et al. (1989) Nature 339:385; Huang et al. (1988) J Virol. 62:3855; and Schlienger et al. (1992) J Virol. 66:2).
The Multiple Antigen Peptide system for peptide-based immunization can also be utilized to generate an immunogen, wherein a desired portion of an hh polypeptide is obtained directly from organo-chemical synthesis of the peptide onto an oligomeric branching lysine core (see, for example, Posnett et al. (1988) JBC 263:1719 and Nardelli et al. (1992) J lImmunol. 148:914). Antigenic determinants of hh proteins can also be expressed and presented by bacterial cells.
In addition to utilizing fusion proteins to enhance immunogenicity, it is widely appreciated that fusion proteins can also facilitate the expression of proteins, and accordingly, can be used in the expression of the vertebrate hh polypeptides of the present invention. For example, hedgehog polypeptides can be generated as glutathione-S-transferase (GST-fusion) proteins. Such 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. John Wiley Sons, 1991)). In another embodiment, 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 Ssequence which naturally occurs at the N-terminus of the hh protein of the pro-form, in order to permit purification of the poly(His)-hh protein by affinity chromatography using a Ni 2 metal resin. The purification leader sequence can then be subsequently removed by treatment with enterokinase see Hochuli et al. (1987) J. Chromatography 411:177; and Janknecht et al. PNAS 88:8972).
Techniques for making fusion genes are known to those skilled in the art. Essentially, the joining of various DNA fragments coding for different polypeptide sequences is i 30 performed in accordance with conventional techniques, employing blunt-ended or staggerended 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. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, 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 391 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 hh derivatives by forming covalent or aggregate conjugates with other chemical moieties, such as glycosyl groups, 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.
For instance, 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. For example, 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 Korblihtt et al. (1985) EMBO 4:1755-9) can be added to the hh 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 present invention also makes available isolated hedgehog polypeptides which are isolated from, or otherwise substantially free of other cellular and extracellular proteins, especially morphogenic proteins or other extracellular or cell surface associated proteins which may normally be associated with the hedgehog polypeptide. The term "substantially free of other cellular or extracellular proteins" (also referred to herein as "contaminating proteins") or "substantially pure or purified preparations" are defined as encompassing preparations of hh polypeptides having less than 20% (by dry weight) contaminating protein, and preferably having less than 5% contaminating protein. Functional forms of the subject polypeptides can be prepared, for the first time, as purified preparations by using a cloned gene as described herein. By "purified", it is meant, when referring to a peptide or DNA or RNA sequence, that the indicated molecule is present in the substantial absence of other biological macromolecules, such as other proteins. The term "purified" as used herein preferably means at least 80% by dry weight, more preferably in the range of 95-99% by weight, and most preferably at least 99.8% by weight, of biological macromolecules of the same type present (but water, buffers, and other small molecules, especially molecules having a molecular weight of less than 5000, can be present). The term "pure" as used herein Spreferably has the same numerical limits as "purified" immediately above. "Isolated" and "purified" do not encompass either natural materials in their native state or natural materials that have been separated into components in an acrylamide gel) but not obtained either as pure lacking contaminating proteins, or chromatography reagents such as denaturing agents and polymers, e.g. acrylamide or agarose) substances or solutions. In preferred embodiments, purified hedgehog preparations will lack any contaminating proteins from the same animal from that hedgehog is normally produced, as can be accomplished by recombinant expression of, for example, a human hedgehog protein in a non-human cell.
As described above for recombinant polypeptides, isolated hh 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 (see, for instance, Examples 6 and 9).
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. In addition, fragments can be chemically synthesized using techniques known in the art such as conventional Merrifield solid phase f-Moc or t-Boc chemistry. For example, 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 "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 posttranslationally modified in a manner different than the authentic protein. Exemplary 25 derivatives of vertebrate hedgehog proteins include polypeptides which lack N-glycosylation sites to produce an unglycosylated protein), or which lack N-terminal and/or C-terminal sequences.
Modification of the structure of the subject vertebrate hh polypeptides can be for such purposes as enhancing therapeutic or prophylactic efficacy, or stability ex vivo shelf life 30 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.
For example, it is reasonable to expect that an isolated replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, a threonine with a serine, or a similar 36 replacement of an amino acid with a structurally related amino acid isosteric and/or isoelectric mutations) will not have a major effect on the biological activity of the resulting molecule. Conservative replacements are those that take place within a family of amino acids that are related in their side chains. Genetically encoded amino acids are can be divided into four families: acidic aspartate, glutamate; basic lysine, arginine, histidine; (3) nonpolar alanine, valine, .leucine, isoleucine, proline, phenylalanine, methionine, tryptophan; and uncharged polar glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine. Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids. In similar fashion, the amino acid repertoire can be grouped as (1) acidic aspartate, glutamate; basic lysine, arginine histidine, aliphatic glycine, alanine, valine, leucine, isoleucine, serine, threonine, with serine and threonine optionally be grouped separately as aliphatic-hydroxyl; aromatic phenylalanine, tyrosine, tryptophan; amide asparagine, glutamine; and sulfur -containing cysteine and methionine.
(see, for example, Biochemistry, 2nd ed., Ed. by L. Stryer, WH Freeman and 1981).
Whether a change in the amino acid sequence of a peptide results in a functional hedgehog homolog functional in the sense that it acts to mimic or antagonize the wild-type form) 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.
This invention further contemplates a method for generating sets of combinatorial mutants of the subject hedgehog proteins as well as truncation mutants, and is especially useful for identifying potential variant sequences 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 hh homologs which can act as either agonists or antagonist, or alternatively, possess novel activities all together. To illustrate, hedgehog homologs can be engineered by the present method to provide more efficient binding to a cognate receptor, yet still retain at least a portion of an activity associated with hh. Thus, combinatorially-derived homologs can be generated to have an increased potency relative to a naturally occurring form of the protein. Likewise, 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 Sagonists. Moreover, manipulation of certain domains of hh by the present method can S 35 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.
37 In one aspect of this method, the amino acid sequences for a population of hedgehog homologs or other related proteins are aligned, preferably to promote the highest homology possible. Such a population of variants can include, for example, hh 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. In a preferred embodiment, the variegated library of hedgehog variants is generated by combinatorial mutagenesis at the nucleic acid level, and is encoded by a variegated gene library. For instance, a mixture of synthetic oligonucleotides can be enzymatically ligated into gene sequences such that the degenerate set of potential hh sequences are expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins for phage display) containing the set of hh sequences therein.
As illustrated in Figure 5A, to analyze the sequences of a population of variants, 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. In order to maintain the highest homology in alignment of sequences, deletions in the sequence of a variant relative to the reference sequence can be represented by an amino acid space or while insertional mutations in the variant relative to the reference sequence can be disregarded and left out of the sequence of the variant when aligned. For instance, Figure includes the alignment of several cloned forms of hh from different species. Analysis of the alignment of the hh clones shown in Figure 5A can give rise to the generation of a degenerate library of polypeptides comprising potential hh sequences.
In an illustrative embodiment, alignment of exon 1/2 encoded sequences the Nterminal approximately 165 residues of the mature protein) of each of the Shh clones produces a degenerate set of Shh polypeptides represented by the general formula: C-G-P-G-R-G-X A-E-K-T-L-G-A-S-G-R-Y-E-G-K-I-X(3)-R-N-S-E-R-F-K-E-L-T-P-N-Y-N- P-D-I-I-F-K-D-E-E-N-T-G-A-D-R-L-M-T-Q-R-C-K-D-K-L-N-X(4)
-L-A-I-
30 L-H-Y-E-G-R-A-V-D-I-T-T-S-D-R-D-X(8)-S-K-Y-G A-V-E-A-G-F-D-W-V-Y-Y-E-S-K-A-H-I-H-C-S-V-K-A-E (SEQ ID No: wherein each of the degenerate positions can be an amino acid which occurs in that position in one of the human, mouse, chicken or zebrafish Shh clones, or, to expand the 35 library, each X can also be selected from amongst amino acid residue which would be conservative substitutions for the amino acids which appear naturally in each of those positions. For instance, Xaa(l) represents Gly, Ala, Val, Leu, Ile, Phe, Tyr or Trp Xaa(2) represents Arg, His or Lys; Xaa(3) represents Gly, Ala, Val, Leu, Ile, Ser or Thr; Xaa(4) represents Gly, Ala, Val, Leu. Ile, 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; and Xaa(i 0) represents Gly, Ala, Val, Leu, le, Ser or Thr. In an even more expansive library, each X can be selected from any amino acid.
In similar fashion, alignment of each of the human, mouse, chicken and zebrafish hedgehog clones (Figure 5B), can provide a degenerate polypeptide sequence represented by the general formula: C-G-P-G-R-G-X(1) -X -P-K-X -L-X P-L-X(9) -Y-K-Q-F-X(10) -P-X (11) -X(12) -X(13) -E-X(14) -T-L-G-A-S-G- -X(16) -E-G-X(17) -X(18) -X(19) -R-X (20) -S-E-R-F-X (21) -X(22) I-F-K-D-E-E-N -X (23) -G-A-D-R-L-M-T-X (24) -R-C- K-X (25) -X(26) -X(27) -N-X(28) -L-A-I-S-V-M-N-X (29) -W-P-G-V-X(30) L-R-V-T-E-G-X (31) -D-E-D-G-H-H-X (32) -X (33) -X(34) -S-L-H-Y-E-G-R- A-X(35) -D-I-T-T-S-D-R-D-X(36) -X(37) -K-Y-G-X (38) -L-X(39) -R-L-A- -X(41) -H-X (42) -H-X(43) -S-V-K-X(44) (SEQ ID No: 41), wherein, as above, each of the degenerate positions can be an amino acid which occurs in a corresponding position in one of the wild-type clones, and may also include amino acid residue which would be conservatve substitutions, or each X can be any amino acid residue.
In an exemplary embodiment, Xaa(l) represents Gly, Ala, Val, Leu, Ile, Pro, Phe or Tyr; Xaa(2) represents Gly, Ala, Val, Leu or le; Xaa(3) represents Gly, Ala, Val, Leu, le, Lys, His or Arg; Xaa(4) represents Lys, Arg or His; Xaa(5) represents Phe, Tip, Tyr or an amino acid gap; Xaa(6) represents Gly, Ala, Val, Leu, le or an amino acid gap; Xaa(7) represents 25 Asn, Gin, His, Arg or Lys; Xaa(8) represents Gly, Ala, Val, Leu, le, Ser or Thr; Xaa(9) represents Gly, Ala, Val, Leu, Ile, Ser or Thr; Xaa(l0) represents Gly, Ala, Val, Leu, le, Ser or Thr; Xaa(i 1) represents Ser, Thr, Gin or Asn; Xaa(1 2) represents Met, Cys, Gly, Ala, Val, Leu, Ile, Ser or Thr; Xaa(13) represents Gly, Ala, Val, Leu, Ile or Pro; Xaa(14) represents Arg, His or Lys; Xaa(1 5) represents Gly, Ala, Val, Leu, le, Pro, Arg, His or Lys; Xaa(1 6) 30 represents Gly, Ala, Val, Leu, Ile, Phe or Tyr; Xaa(l 7) represents Mrg, His or Lys; Xaa(1 8) represents Gly, Ala, Val, Leu, Ile, Ser or Thr; Xaa( 19) represents Thr or Scr; represents .Giy, Ala, Val, Leu, le, Asn or Gin; Xaa(21) represents Mrg, His or Lys; Xaa(22) represents Asp or Giu; Xaa(23) represents Ser or Thr; Xaa(24) represents Glu, Asp, Gin or Asn; Xaa(25) represents Glu or Asp; Xaa(26) represents Mrg, His or Lys; Xaa(27) represents Gly, Ala, Val, Leu or le; Xaa(28) represents Gly, Ala, Val, Leu, le, Thr or Ser; Xaa(29) represents Met, Cys, Gin, Asn, Mrg, Lys or His; Xaa(30) represents Mrg, His or Lys; Xaa(3 1) represents Tip, Phe, Tyr, Mrg, His or Lys; Xaa(32) represents Gly, Ala, Val, Leu, le, Ser, Thr, Tyr or Phe; Xaa(33) represents Gin, Asn, Asp or Glu; Xaa(34) represents Asp or Glu; represents Gly, Ala, Val, Leu, or le; Xaa(36) represents Mrg, His or Lys; Xaa(37) represents Asn, Gin, Thr or Ser; Xaa(38) represents Gly, Ala, Val, Leu, le, Ser, Thr, Met or 37 Cys; Xaa(39) represents Gly, Ala, Val, Leu, lie, Thr or Ser; Xaa(40) represents Arg, His or Lys; Xaa(41) represents Asn, Gin, Gly, Ala, Val, Leu or Ile; Xaa(42) represents Gly, Ala, Val, Leu or Ile; Xaa(43) represents Gly, Ala, Val, Leu, lie, Ser, Thr or Cys; Xaa(44) represents Gly, Ala, Val, Leu, lie, Thr or Ser; and Xaa(45) represents Asp or Glu.
There are many ways by which the library of potential hh 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 hh 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. AG Walton, Amsterdam: Elsevier pp273- 289; Itakura et al. (1984) Annu. Rev. Biochem. 53:323; Itakura et al. (1984) Science 198:1056; Ike et al. (1983) Nucleic Acid Res. 11:477. Such techniques have been employed in the directed evolution of other proteins (see, for example, Scott et al. (1990) Science 249:386-390; Roberts et al. (1992) PNAS 89:2429-2433; Devlin et al. (1990) Science 249: 404-406; Cwirla et al. (1990) PNAS 87: 6378-6382; as well as U.S. Patents Nos. 5,223,409, 5,198,346, and 5,096,815).
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.
In one embodiment, the combinatorial library is designed to be secreted the polypeptides of the library all include a signal sequence but no transmembrane or S"cytoplasmic domains), and is used to transfect a eukaryotic cell that can be co-cultured with embryonic cells. A functional hedgehog protein secreted by the cells expressing the combinatorial library will diffuse to neighboring embryonic cells and induce a particular biological response, such as to illustrate, neuronal differentiation. Using antibodies directed to epitopes of particular neuronal cells Islet-1 or Pax-1), the pattern of detection of neuronal induction will resemble a gradient function, and will allow the isolation (generally after several repetitive rounds of selection) of cells producing active hedgehog homologs.
Likewise, hh antagonists can be selected in similar fashion by the ability of the cell producing a functional antagonist to protect neighboring cells from the effect of wild-type hedgehog added to the culture media.
To illustrate, target cells are cultured in 24-well microtitre plates. Other eukaryotic cells are transfected with the combinatorial hh gene library and cultured in cell culture inserts 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 hh 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, the inserts are removed and the effect of the variant hedgehog proteins on the target cells determined. For example, where the target cell is a neural crest cell and the activity desired from the hh homolog is the induction of neuronal differentiation, then fluorescently-labeled antibodies specific for Islet-1 or other neuronal markers can be used to score for induction in the target cells as indicative of a functional hh in that well. Cells from the inserts corresponding to wells which score positive for activity can be split and recultured on several inserts, the process being repeated until the active clones are identified.
In yet another screening assay, 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 an hedgehog receptor or a ligand which binds the hedgehog protein) via this gene product is detected in a "panning assay". Such S panning steps can be carried out on cells cultured from embryos. For instance, 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. (1992) TIBS 18:136-140). In a similar fashion, fluorescently labeled molecules which bind hh can be used to score for potentially functional hh homologs. Cells can be visually inspected and separated under a fluorescence microscope, or, where the morphology of the' cell permits, separated by a fluorescenceactivated cell sorter.
In an alternate embodiment, the gene library is expressed as a fusion protein on the surface of a viral particle. For instance, in the filamentous phage system, foreign peptide sequences can be expressed on the surface of infectious phage, thereby conferring two significant benefits. First, since these phage can be applied to affinity matrices at very high concentrations, large number of phage can be screened at one time. Second, since each infectious phage displays the combinatorial gene product on its surface, if a particular phage is recovered from an affinity matrix in low yield, the phage can be amplified by another round of infection. The group of almost identical E.coli filamentous phages M13, 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).
In an illustrative embodiment, the recombinant phage antibody system (RPAS, Pharamacia Catalog number 27-9400-01) can be easily modified for use in expressing and screening hh combinatorial libraries. For instance, the pCANTAB 5 phagemid of the RPAS kit contains the gene which encodes the phage gill coat protein. The hh combinatorial gene library can be cloned into the phagemid adjacent to the gIII signal sequence such that it will be expressed as a gI fusion protein. After ligation, the phagemid is used to transform competent E. coli TG1 cells. Transformed cells are subsequently infected with M13K07 helper phage to rescue the phagemid and its candidate hh gene insert. The resulting recombinant phage contain phagemid DNA encoding a specific candidate hh, and display one or more copies of the corresponding fusion coat protein. The phage-displayed candidate hedgehog proteins which are capable of binding an hh receptor are selected or enriched by panning. For instance, the phage library can be applied to cultured embryonic cells 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 gIIl coat protein, they will retain S their ability to infect E. coli. Thus, successive rounds of reinfection of E. coli, and panning will greatly enrich for hh homologs, which can then be screened for further biological 25 activities in order to differentiate agonists and antagonists. Moreover, differential panning, with two or more different hh-responsive cells, can facilitate isolation of hedgehog homologs of selectively narrower biological activity relative to the wild-type protein.
The invention also provides for reduction of the vertebrate hh protein to generate mimetics, e.g. peptide or non-peptide agents, which are able to disrupt binding of a vertebrate hh polypeptide of the present invention with an hh receptor. Thus, such mutagenic S techniques as described above are also useful to map the determinants of the hedgehog S proteins which participate in protein-protein interactions involved in, for example, binding of the subject vertebrate hh polypeptide to other extracellular matrix components. To illustrate, the critical residues of a subject hh polypeptide or hh ligand which are involved in molecular 35 recognition of an hh receptor can be determined and used to generate hedgehog-derived peptidomimetics which competitively inhibit binding of the authentic hedgehog protein with that moiety. By employing, for example, scanning mutagenesis to map the amino acid V2 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. For instance, nonhydrolyzable peptide analogs of such residues can be generated using benzodiazepine see Freidinger et al. in Peptides: Chemistry and Biology, G.R. Marshall ed., ESCOM Publisher: Leiden, Netherlands, 1988), azepine see Huffman et al. in Peptides: Chemistry and Biology, G.R. Marshall ed., ESCOM Publisher: Leiden, Netherlands, 1988), substituted gama lactam rings (Garvey et al. in Peptides: Chemistry and Biology, G.R.
Marshall ed., ESCOM Publisher: Leiden, Netherlands, 1988), keto-methylene pseudopeptides (Ewenson et al. (1986) J Med Chem 29:295; and Ewenson et al. in Peptides: Structure and Function (Proceedings of the 9th American Peptide Symposium) Pierce Chemical Co.
Rockland, IL, 1985), p-turn dipeptide cores (Nagai et al. (1985) Tetrahedron Lett 26:647; and Sato et al. (1986) J Chem Soc Perkin Trans 1:1231), and p-aminoalcohols (Gordon et al.
(1985) Biochem Biophys Res Commun126:419; and Dann et al. (1986) Biochem Biophys Res Commun 134:71).
Another aspect of the invention pertains to an antibody specifically reactive with a vertebrate hedgehog protein. For example, by using immunogens derived from hedgehog protein, e.g. based on the cDNA sequences, anti-protein/anti-peptide antisera or monoclonal antibodies can be made by standard protocols (See, for example, Antibodies: A Laboratory Manual ed. by Harlow and Lane (Cold Spring Harbor Press: 1988)). A mammal, such as a mouse, a hamster or rabbit can be immunized with an immunogenic form of the peptide S a vertebrate hh polypeptide or an antigenic fragment which is capable of eliciting an antibody response). Techniques for conferring immunogenicity on a protein or peptide include conjugation to carriers or other techniques well known in the art. An immunogenic portion of a hedgehog protein can be administered in the presence of adjuvant. The progress of immunization can be monitored by detection of antibody titers in plasma or serum. Standard ELISA or other immunoassays can be used with the immunogen as antigen to assess the levels of antibodies. In a preferred embodiment, the subject antibodies are immunospecific for antigenic determinants of a hedgehog protein of a vertebrate organism, such as a mammal, e.g. antigenic determinants of a protein represented by SEQ ID Nos:8-14 or a closely related homolog at least 85% homologous, preferably at least 90% homologous, and more preferably at least 95% homologous). In yet a further preferred embodiment of the present invention, in order to provide, for example, antibodies which are immuno-selective for discrete hedgehog homologs, e.g. Shh versus Dhh versus Ihh, the anti-hh polypeptide antibodies do not substantially cross react does not react specifically) with a protein which is, for example, less than 85% homologous to any of SEQ ID Nos:8-14; less than homologous with one of SEQ ID Nos:8-14; less than 98-99% homologous with 43 one of SEQ ID Nos:8-14. By "not substantially cross react", it is meant that the antibody has a binding affinity for a non-homologous protein which is at least one order of magnitude, more preferably at least 2 orders of magnitude, and even more preferably at least 3 orders of magnitude less than the binding affinity of the antibody for one or more of the proteins of SEQ ID Nos:8-14.
Following immunization of an animal with an antigenic preparation of a hedgehog protein, anti-hh antisera can be obtained and, if desired, polyclonal anti-hh antibodies isolated from the serum. To produce monoclonal antibodies, antibody-producing cells (lymphocytes) can be harvested from an immunized animal and fused by standard somatic cell fusion procedures with immortalizing cells such as myeloma cells to yield hybridoma cells. Such techniques are well known in the art, an include, for example, the hybridoma technique (originally developed by Kohler and Milstein, (1975) Nature, 256: 495-497), the human B cell hybridoma technique (Kozbar et al., (1983) Immunology Today, 4: 72), and the EBVhybridoma technique to produce human monoclonal antibodies (Cole et al., (1985) Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc. pp. 77-96). Hybridoma cells can be screened immunochemically for production of antibodies specifically reactive with a vertebrate hh polypeptide of the present invention and monoclonal antibodies isolated from a culture comprising such hybridoma cells.
The term antibody as used herein is intended to include fragments thereof which are also specifically reactive with one of the subject vertebrate hh polypeptides. Antibodies can be fragmented using conventional techniques and the fragments screened for utility in the same manner as described above for whole antibodies. For example, F(ab) 2 fragments can S be generated by treating antibody with pepsin. The resulting F(ab) 2 fragment can be treated 5 to reduce disulfide bridges to produce Fab fragments. The antibody of the present invention 25 is further intended to include bispecific and chimeric molecules having affinity for a hedgehog protein conferred by at least one CDR region of the antibody.
Both monoclonal and polyclonal antibodies (Ab) directed against authentic hedgehog polypeptides, or hedgehog variants, and antibody fragments such as Fab and F(ab) 2 can be used to block the action of one or more hedgehog proteins and allow the study of the role of 30 these proteins in, for example, embryogenesis and/or maintenance of differential tissue. For S.example, purified monoclonal Abs can be injected directly into the limb buds of chick or mouse embryos. It is demonstrated in the examples below that hh is expressed in the limb buds of, for example, day 10.5 embryos. Thus, the use of anti-hh Abs during this developmental stage can allow assessment of the effect of hh on the formation of limbs in vivo. In a similar approach, hybridomas producing anti-hh monoclonal Abs, or biodegradable gels in which anti-hh Abs are suspended, can be implanted at a site proximal or within the area at which hh action is intended to be blocked. Experiments of this nature can aid in deciphering the role of this and other factors that may be involved in limb patterning and tissue formation.
Antibodies which specifically bind hedgehog epitopes can also be used in immunohistochemical staining of tissue samples in order to evaluate the abundance and pattern of expression of each of the subject hh polypeptides. Anti-hedgehog antibodies can be used diagnostically in immuno-precipitation and immuno-blotting to detect and evaluate hedgehog protein levels in tissue as part of a clinical testing procedure. For instance, such measurements can be useful- in predictive valuations of the onset or progression of neurological disorders, such as those marked by denervation-like or disuse-like symptoms.
Likewise, the ability to monitor hh levels in an individual can allow determination of the efficacy of a given treatment regimen for an individual afflicted with such a disorder. The level of hh polypeptides may be measured in bodily fluid, such as in samples of cerebral spinal fluid or amniotic fluid, or can be measured in tissue, such as produced by biopsy.
Diagnostic assays using anti-hh antibodies can include, for example, immunoassays designed to aid in early diagnosis of a neurodegenerative disorder, particularly ones which are manifest at birth. Diagnostic assays using anti-hh polypeptide antibodies can also include immunoassays designed to aid in early diagnosis and phenotyping of a differentiative disorder, as well as neoplastic or hyperplastic disorders.
Another application of anti-hh antibodies of the present invention is in the immunological screening of cDNA libraries constructed in expression vectors such as Xgtl 1, Xgtl8-23, XZAP, and XORF8. Messenger libraries of this type, having coding sequences inserted in the correct reading frame and orientation, can produce fusion proteins. For instance, Agtll will produce fusion proteins whose amino termini consist of B-galactosidase amino acid sequences and whose carboxy termini consist of a foreign polypeptide. Antigenic 25 epitopes of an hh protein, e.g. other orthologs of a particular hedgehog protein or other *homologs from the same species, can then be detected with antibodies, as, for example, reacting nitrocellulose filters lifted from infected plates with anti-hh antibodies. Positive phage detected by this assay can then be isolated from the infected plate. Thus, the presence of hedgehog homologs can be detected and cloned from other animals, as can alternate isoforms (including splicing variants) fr6m humans.
Moreover, the nucleotide sequences determined from the cloning of hh genes from S"vertebrate organisms will further allow for the generation of probes and primers designed for .use in identifying and/or cloning hedgehog homologs in other cell types, e.g. from other tissues, as well as hh homologs from other vertebrate organisms. For instance, the present invention also provides a probe/primer comprising a substantially purified oligonucleotide, which oligonucleotide comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least 10 consecutive nucleotides of sense or anti-sense sequence selected from the group consisting of SEQ ID No:l, SEQ ID No:2, SEQ ID No:3, SEQ ID No:4, SEQ ID No:5, SEQ ID No:6 and SEQ ID No:7, or naturally occurring mutants thereof.
For instance, primers based on the nucleic acid represented in SEQ ID Nos:1-7 can be used in PCR reactions to clone hedgehog homologs. Likewise, probes based on the subject hedgehog sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins. In preferred embodiments, the probe further comprises a label group attached thereto and able to be detected, e.g. the label group is selected from the group consisting of radioisotopes, fluorescent compounds, enzymes, and enzyme co-factors.
Such probes can also be used as a part of a diagnostic test kit for identifying cells or tissue which misexpress a hedgehog protein, such as by measuring a level of a hedgehog encoding nucleic acid in a sample of cells from a patient; e.g. detecting hh mRNA levels or determining whether a genomic hh gene has been mutated or deleted.
To illustrate, nucleotide probes can be generated from the subject hedgehog genes which facilitate histological screening of intact tissue and tissue samples for the presence (or absence) of hedgehog-encoding transcripts. Similar to the diagnostic uses of anti-hedgehog antibodies, the use of probes directed to hh messages, or to genomic hh sequences, can be used for both predictive and therapeutic evaluation of allelic mutations which might be manifest in, for example, neoplastic or hyperplastic disorders unwanted cell growth) or abnormal differentiation of tissue. Used in conjunction with immunoassays as described above, the oligonucleotide probes can help facilitate the determination of the molecular basis for a developmental disorder which may involve some abnormality associated with expression (or lack thereof) of a hedgehog protein. For instance, variation in polypeptide synthesis can be differentiated from a mutation in a coding sequence.
Accordingly, the present method provides a method for determining if a subject is at S. 25 risk for a disorder characterized by aberrant control of differentiation or unwanted cell proliferation. For instance, the subject assay can be used in the screening and diagnosis of genetic and acquired disorders which involve alteration in one or more of the hedgehog genes. In preferred embodiments, the subject method can be generally characterized as comprising: detecting, in a tissue sample of the subject a human patient), the presence or absence of a genetic lesion characterized by at least one of a mutation of a gene encoding a hedgehog protein or (ii) the mis-expression of a hedgehog gene. To illustrate, such genetic lesions can be detected by ascertaining the existence of at least one of a deletion of one or more nucleotides from a hedgehog gene, (ii) an addition of one or more .nucleotides to a hedgehog gene, (iii) a substitution of one or more nucleotides of a hedgehog gene, (iv) a gross chromosomal rearrangement of a hedgehog gene, a gross alteration in the level of a messenger RNA transcript of an hh gene, (vi) the presence of a non-wild type splicing pattern of a messenger RNA transcript of a vertebrate hh gene, and (vii) a non-wild type level of a hedgehog protein. In one aspect of the invention there is provided a probe/primer comprising an oligonucleotide containing a region of nucleotide sequence which is capable of hybridizing to a sense or antisense sequence selected from the group consisting of SEQ ID Nos:1-7, or naturally occurring mutants thereof, or 5' or 3' flanking sequences or intronic sequences naturally associated with a vertebrate hh gene. The probe is exposed to nucleic acid of a tissue sample; and the hybridization of the probe to the sample nucleic acid is detected. In certain embodiments, detection of the lesion comprises utilizing the probe/primer in a polymerase chain reaction (PCR) (see, U.S. Patent No: 4,683,195 and 4,683,202) or, alternatively, in a ligation chain reaction (LCR) (see, Landegran et al. (1988) Science, 241:1077-1080; and NaKazawa et al. (1944) PNAS 91:360-364) the later of which can be particularly useful for detecting point mutations in hedgehog genes.
Alternatively, immunoassays can be employed to determine the level of hh proteins, either soluble or membrane bound.
Yet another diagnostic screen employs a source of hedgehog protein directly. As described herein, hedgehog proteins of the present invention are involved in the induction of differentiation. Accordingly, the pathology of certain differentiative and/or proliferative disorders can be marked by loss of hedgehog sensitivity by the afflicted tissue.
Consequently, the response of a tissue or cell sample to an inductive amount of a hedgehog protein can be used to detect and characterize certain cellular transformations and degenerative conditions. For instance, tissue/cell samples from a patient can be treated with a hedgehog agonist and the response of the tissue to the treatment determined. Response can be qualified and/or quantified, for example, on the basis of phenotypic change as result of hedgehog induction. For example, expression of gene products induced by hedgehog treatment can be scored for by immunoassay. The patched protein, for example, is 25 upregulated in drosophila in response to Dros-HH, and, in light of the findings herein, a presumed vertebrate homolog will similarly be upregulated. Thus, detection of patched expression on the cells of the patient sample can permit detection of tissue that is not hedgehog-responsive. Likewise, scoring for other phenotypic markers provides a means for determining the response to hedgehog.
Furthermore, by making available purified and recombinant hedgehog polypeptides, the present invention facilitates the development of assays which can be used to screen for drugs, including hedgehog homologs, which are either agonists or antagonists of the normal cellular function of the subject hedgehog polypeptides, or of their role in the pathogenesis of cellular differentiation and/or proliferation and disorders related thereto. In one embodiment, 35 the assay evaluates the ability of a compound to modulate binding between a hedgehog polypeptide and a hedgehog receptor. A variety of assay formats will suffice and, in light of the present inventions, will be comprehended by skilled artisan.
9/7 In many drug screening programs which test libraries of compounds and natural extracts, high throughput assays are desirable in order to maximize the number of compounds surveyed in a given period of time. Assays which are performed in cell-free systems, such as may be derived with purified or semi-purified proteins, are often preferred as "primary" screens in that they can be generated to permit rapid development and relatively easy detection of an alteration in a molecular target which is mediated by a test compound.
Moreover, the effects of cellular toxicity and/or bioavailability of the test compound can be generally ignored in the in vitro system, the assay instead being focused primarily on the effect of the drug on the molecular target as may be manifest in an alteration of binding affinity with receptor proteins. Accordingly, in an exemplary screening assay of the present invention, the compound of interest is contacted with a hedgehog receptor polypeptide which is ordinarily capable of binding a hedgehog protein. To the mixture of the compound and receptor is then added a composition containing a hedgehog polypeptide. Detection and quantification of receptor/hedgehog complexes provides a means for determining the 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. Moreover, 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 a composition containing the receptor protein, and the formation of receptor/hedgehog complex is quantitated in the absence of the test compound.
In an illustrative embodiment, the polypeptide utilized as a hedgehog receptor can be generated from the drosophila patched protein or a vertebrate homolog thereof. In light of the ability of, for example, Shh to activate Dros-HH pathways in transgenic drosophila (see 25 Example it may be concluded that vertebrate hedgehog proteins are capable of binding to Drosophila HH receptors. Accordingly, an exemplary screening assay includes a suitable portion of the patched protein (SEQ ID No. 42), such as one or both of the substantial extracellular domains residues Lys-93 to His-426 and Arg-700 to Arg-966). For instance, the patched protein can be provided in soluble form, as for example a preparation of 30 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), or can be provided as part of a liposomal preparation or expressed on the surface of a cell.
Complex formation between the hedgehog polypeptide and a hedgehog receptor may 35 be detected by a variety of techniques. For instance, modulation of the formation of complexes can be quantitated using, for example, detectably labeled proteins such as radiolabelled, fluorescently labeled, or enzymatically labeled hedgehog polypeptides, by immunoassay, or by chromatographic detection.
Typically, it will be desirable to immobilize either the hedgehog receptor or the hedgehog polypeptide to facilitate separation of receptor/hedgehog complexes from uncomplexed forms of one of the proteins, as well as to accommodate automation of the assay. In one embodiment, a fusion protein can be provided which adds a domain that allows the protein to be bound to a matrix. For example, 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. an 35 S-labeled hedgehog polypeptide, and the test compound and incubated under conditions conducive to complex formation, e.g. at physiological conditions for salt and pH, though slightly more stringent conditions may be desired. Following incubation, the beads are washed to remove any unbound hedgehog polypeptide, and the matrix bead-bound radiolabel determined directly beads placed in scintillant), or in the supernatant after the receptor/hedgehog complexes are dissociated.
Alternatively, the complexes can 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.
Other techniques for immobilizing proteins on matrices are also available for use in the subject assay. For instance, soluble portions of the hedgehog receptor protein can be immobilized utilizing conjugation of biotin and streptavidin. For instance, biotinylated -reeeepter--moleeules-can-be-prepared- from- biotin-NHS -(N-hydroxy-succinimide) using techniques well known in the art biotinylation kit, Pierce Chemicals, Rockford, IL), and S: immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
25 Alternatively, 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. As above, preparations of a hedgehog polypeptide and a test compound are incubated in the receptor-presenting wells of the plate, and the amount of receptor/hedgehog complex trapped in the well can be quantitated. Exemplary methods for detecting such complexes, in addition to those described above for the GST-immobilized 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. In the instance of 35 the latter, the enzyme can be chemically conjugated or provided as a fusion protein with the hedgehog polypeptide. To illustrate, the hedgehog polypeptide can be chemically crosslinked 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. Likewise, a fusion protein comprising the hedgehog polypeptide and glutathione-S-transferase can be provided, and complex formation quantitated by detecting the GST activity using 1-chloro-2,4-dinitrobenzene (Habig et al (1974) JBiol Chem 249:7130).
For processes which rely on immunodetection for quantitating one of the proteins trapped in the complex, antibodies against the protein, such as the anti-hedgehog antibodies described herein, can be used. Alternatively, 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 from commercial sources). For instance, 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 see Ellison et al. (1991) J Biol Chem 266:21150-21157) which includes a 10-residue sequence from c-myc, as well as the pFLAG system (International Biotechnologies, Inc.) or the pEZZ-protein A system (Pharamacia, NJ).
Where the desired portion of the hh receptor (or other hedgehog binding molecule) cannot be provided in soluble form, liposomal vesicles can be used to provide manipulatable and isolatable sources of the receptor. For example, both authentic and recombinant forms of 20 the patched protein can be reconstituted in artificial lipid vesicles 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).
In addition to cell-free assays, such as described above, the readily available source of 25 vertebrate hedgehog proteins provided by the present invention also facilitates the generation of cell-based assays for identifying small molecule agonists/antagonists and the like.
Analogous to the cell-based assays described above for screening combinatorial libraries, cells which are sensitive to hedgehog induction can be contacted with a hedgehog protein and a test agent of interest, with the assay scoring for modulation in hedgehog inductive responses by the target cell in the presence and absence of the test agent. As with the cellfree assays, agents which produce a statistically significant change in hedgehog activities (either inhibition or potentiation) can be identified. In an illustrative embodiment, motor neuron progenitor cells, such as from neural plate explants, can be used as target cells.
Treatment of such explanted cells with, for example, Shh causes the cells to differentiate into motor neurons. By detecting the co-expression of the LIM homeodomain protein Islet-1 (Thor et al. (1991) Neuron 7:881-889; Ericson et al. (1992) Science 256:1555-1560) and the immunoglobulin-like protein SCI (Tanaka et al. (1984) Dev Biol 106:26-37), the ability of a candidate agent to potentiate or inhibit Shh induction of motor neuron differentiation can be measured. The hedgehog protein can be provided as a purified source, or in the form of cells/tissue which express the protein and which are co-cultured with the target cells.
In yet another embodiment, the method of the present invention can be used to isolate and clone hedgehog receptors. For example, purified hedgehog proteins of the present invention can be employed to precipitate hedgehog receptor proteins from cell fractions prepared from cells which are responsive to a hedgehog protein. For instance, purified hedgehog protein can be derivatized with biotin (using, for instance, NHS-Biotin, Pierce Chemical catalog no. 21420G), and the biotinylated protein utilized to saturate membrane bound hh receptors. The hedgehog bound receptors can subsequently be adsorbed or immobilized on streptavidin. If desired, the hedgehog-receptor complex can be cross-linked with a chemical cross-linking agent. In such as manner, hh receptors can be purified, preferably to near homogeneity. The isolated hh receptor can then be partially digested with, for example, trypsin, and the resulting peptides separated by reverse-phase chromatography.
The chromatography fragments are then analyzed by Edman degradation to obtain single sequences for two or more of the proteolytic fragments. From the chemically determined amino acid sequence for each of these tryptic fragments, a set of oligonucleotide primers can be designed for PCR. These primers can be used to screen both genomic and cDNA libraries.
Similar strategies for cloning receptors have been employed, for example, to obtain the recombinant gene for somatostatin receptors (Eppler et al. (1992) J Biol Chem 267:15603- 15612).
Other techniques for identifying hedgehog receptors by expression cloning will be evident in light of the present disclosure. For instance, purified hh polypeptides can be immobilized in wells of micro titre plates and contacted with, for example, COS cells transfected with a cDNA library from tissue expected to be responsive to hedgehog induction). From this panning assay, cells which express hedgehog receptor molecules can be isolated on the basis of binding to the immobilized hedgehog protein. Another cloning system, described in PCT publications WO 92/06220 of Flanagan and Leder, involves the use of an expression cloning system whereby a hedgehog receptor is stored on the basis of 30 binding to a hedgehog/alkaline phosphatase fusion protein (see also Cheng et al. (1994) Cell 79:157-168) Another aspect of the present invention relates to a method of inducing and/or maintaining a differentiated state, enhancing survival, and/or promoting proliferation of a cell responsive to a vertebrate hedgehog protein, by contacting the cells with an hh agonist or an hh antagonist as the circumstances may warrant. For instance, it is contemplated by the invention that, in light of the present finding of an apparently broad involvement of hedgehog proteins in the formation of ordered spatial arrangements of differentiated tissues in -57 vertebrates, the subject method could be used to generate and/or maintain an array of different vertebrate tissue both in vitro and in vivo. The hh agent, whether inductive or antiinductive, can be, as appropriate, any of the preparations described above, including isolated polypeptides, gene therapy constructs, antisense molecules, peptidomimetics or agents identified in the drug assays provided herein. Moreover, it is contemplated that, based on the observation of activity of the vertebrate hedgehog proteins in Drosophila, hh agents, for purposes of therapeutic and diagnostic uses, can include the Dros-HH protein and homologs thereof. Moreover, the source of hedgehog protein can be, in addition to purified protein or recombinant cells, cells or tissue explants which naturally produce one or more hedgehog proteins. For instance, as described in Example 2, neural tube explants from embryos, particularly floorplate tissue, can provide a source for Shh polypeptide, which source can be implanted in a patient or otherwise provided, as appropriate, for induction or maintenance of differentiation.
For example, the present method is applicable to cell culture techniques. In vitro neuronal culture systems have proved to be fundamental and indispensable tools for the study of neural development, as well as the identification of neurotrophic factors such as nerve growth factor (NGF), ciliary trophic factors (CNTF), and brain derived neurotrophic factor (BDNF). Once a neuronal cell has become terminally-differentiated it typically will not change to another terminally differentiated cell-type. However, neuronal cells can 20 nevertheless readily lose their differentiated state. This is commonly observed when they are Sgrown in culture from adult tissue, and when they form a blastema during regeneration. The S* present method provides a means for ensuring an adequately restrictive environment in order to maintain neuronal cells at various stages of differentiation, and can be employed, for instance, in cell cultures designed to test the specific activities of other trophic factors. In S 25 such embodiments of the subject method, the cultured cells can be contacted with an hh polypeptide, or an agent identified in the assays described above, in order to induce neuronal differentiation of a stem cell), or to maintain the integrity of a culture of terminallydifferentiated neuronal cells by preventing loss of differentiation. The source of hedgehog protein in the culture can be derived from, for example, a purified or semi-purified protein S 30 composition added directly to the cell culture media, or alternatively, supported and/or released from a polymeric device which supports the growth of various neuronal cells and which has been doped with the protein. The source of the hedgehog protein can also be a cell that is co-cultured with the intended neuronal cell and which produces a recombinantor wild-type hedgehog protein. Alternatively, the source can be the neuronal cell itself which has been engineered to produce a recombinant hedgehog protein. In an exemplary embodiment, a naive neuronal cell a stem cell) is treated with an hh agonist in order to induce differentiation of the cells into, for example, sensory neurons or, alternatively, motomeurons. Such neuronal cultures can be used as convenient assay systems as well as 52 sources ofimplantable cells for therapeutic treatments. For example, hh polypeptides may be useful in establishing and maintaining the olfactory neuron cultures described in U.S. Patent 5,318,907 and the like.
According to the present invention, large numbers of non-tumorigenic neural progenitor cells can be perpetuated in vitro and induced to differentiate by contact with hedgehog proteins. Generally, a method is provided comprising the steps of isolating neural progenitor cells from an animal, perpetuating these cells in vitro or in vivo, preferably in the presence of growth factors, and differentiating these cells into particular neural phenotypes, neurons and glia, by contacting the cells with a hedgehog agonist.
Progenitor cells are thought to be under a tonic inhibitory influence which maintains the progenitors in a suppressed state until their differentiation is required. However, recent techniques have been provided which permit these cells to be proliferated, and unlike neurons which are terminally differentiated and therefore non-dividing, they can be produced in unlimited number and are highly suitable for transplantation into heterologous and autologous hosts with neurodegenerative diseases.
By "progenitor" it is meant an oligopotent or multipotent stem cell which is able to divide without limit and, under specific conditions, can produce daughter cells which terminally differentiate such as into neurons and glia. These cells can be used for transplantation into a heterologous or autologous host. By heterologous is meant a host other than the animal from which the progenitor cells were originally derived. By autologous is meant the identical host from which the cells were originally derived.
i Cells can be obtained from embryonic, post-natal, juvenile or adult neural tissue from S. any animal. By any animal is meant any multicellular animal which contains nervous tissue.
More particularly, is meant any fish, reptile, bird, amphibian or mammal and the like. The 25 most preferable donors are mammals, especially mice and humans.
In the case of a heterologous donor animal, the animal may be euthanized, and the brain and specific area of interest removed using a sterile procedure. Brain areas of particular interest include any area from which progenitor cells can be obtained which will serve to restore function to a degenerated area of the host's brain. These regions include areas of the central nervous system (CNS) including the cerebral cortex, cerebellum, midbrain, brainstem, spinal cord and ventricular tissue, and areas of the peripheral nervous system (PNS) including the carotid body and the adrenal medulla. More particularly, these areas include regions in the basal ganglia, preferably the striatum which consists of the caudate and putamen, or various cell groups such as the globus pallidus, the subthalamic nucleus, the nucleus basalis which is found to be degenerated in Alzheimer's Disease patients, or the substantia nigra pars compacta which is found to be degenerated in Parkinson's Disease patients.
53 Human heterologous neural progenitor cells may be derived from fetal tissue obtained from elective abortion, or from a post-natal, juvenile or adult organ donor. Autologous neural tissue can be obtained by biopsy, or from patients undergoing neurosurgery in which neural tissue is removed, in particular during epilepsy surgery, and more particularly during temporal lobectomies and hippocampalectomies.
Cells can be obtained from donor tissue by dissociation of individual cells from the connecting extracellular matrix of the tissue. Dissociation can be obtained using any known procedure, including treatment with enzymes such as trypsin, collagenase and the like, or by using physical methods of dissociation such as with a blunt instrument. Dissociation of fetal cells can be carried out in tissue culture medium, while a preferable medium for dissociation of juvenile and adult cells is artificial cerebral spinal fluid (aCSF). Regular aCSF contains 124 mM NaCI, 5 mM KC1, 1.3 mM MgCI 2 2 mM CaC1 2 26 mM NaHCO 3 and 10 mM Dglucose. Low Ca 2 aCSF contains the same ingredients except for MgCI 2 at a concentration of 3.2 mM and CaCI 2 at a concentration of 0.1 mM.
.Dissociated cells can be placed into any known culture medium capable of supporting cell growth, including MEM, DMEM, RPMI, F-12, and the like, containing supplements which are required for cellular metabolism such as glutamine and other amino acids, vitamins, minerals and useful proteins such as transferrin and the like. Medium may also contain antibiotics to prevent contamination with yeast, bacteria and fungi such as penicillin, 20 streptomycin, gentamicin and the like. In some cases, the medium may contain serum derived from bovine, equine, chicken and the like. A particularly preferable medium for cells Sis a mixture of DMEM and F-12.
Conditions for culturing should be close to physiological conditions. The pH of the culture media should be close to physiological pH, preferably between pH 6-8, more preferably close to pH 7, even more particularly about pH 7.4. Cells should be cultured at a temperature close to physiological temperature, preferably between 30 0 C-40*C, more preferably between 32*C-38°C, and most preferably between 35 0 C-37 0
C.
Cells can be grown in suspension or on a fixed substrate, but proliferation of the progenitors is preferably done in suspension to generate large numbers of cells by formation of "neurospheres" (see, for example, Reynolds et al. (1992) Science 255:1070-1709; and PCT Publications W093/01275, W094/09119, W094/10292, and W094/16718). In the case of propagating (or splitting) suspension cells, flasks are shaken well and the neurospheres allowed to settle on the bottom comer of the flask. The spheres are then transferred to a ml centrifuge tube and centrifuged at low speed. The medium is aspirated, the cells resuspended in a small amount of medium with growth factor, and the cells mechanically dissociated and resuspended in separate aliquots of media.
Cell suspensions in culture medium are supplemented with any growth factor which allows for the proliferation of progenitor cells and seeded in any receptacle capable of sustaining cells, though as set out above, preferably in culture flasks or roller bottles. Cells typically proliferate within 3-4 days in a 37 0 C incubator, and proliferation can be reinitiated at any time after that by dissociation of the cells and resuspension in fresh medium containing growth factors.
In the absence of substrate, cells lift off the floor of the flask and continue to proliferate in suspension forming a hollow sphere of undifferentiated cells. After approximately 3-10 days in vitro, the proliferating clusters (neurospheres) are fed every 2-7 days, and more particularly every 2-4 days by gentle centrifugation and resuspension in medium containing growth factor.
After 6-7 days in vitro, individual cells in the neurospheres can be separated by physical dissociation of the neurospheres with a blunt instrument, more particularly by triturating the neurospheres with a pipette. Single cells from the dissociated neurospheres are suspended in culture medium containing growth factors, and differentiation of the cells can be induced by plating (or resuspending) the cells in the presence of a hedgehog agonist, and (optionally) any other factor capable of sustaining differentiation, such as bFGF and the like.
To further illustrate other uses of hedgehog agonists and antagonists, it is noted that S intracerebral grafting has emerged as an additional approach to central nervous system therapies. For example, one approach to repairing damaged brain tissues involves the transplantation of cells from fetal or neonatal animals into the adult brain (Dunnett et al.
(1987) J Exp Biol 123:265-289; and Freund et al. (1985) J Neurosci 5:603-616). Fetal neurons from a variety of brain regions can be successfully incorporated into the adult brain, and such grafts can alleviate behavioral defects. For example, movement disorder induced by lesions of dopaminergic projections to the basal ganglia can be prevented by grafts of embryonic dopaminergic neurons. Complex cognitive functions that are impaired after lesions of the neocortex can also be partially restored by grafts of embryonic cortical cells.
The use of hedgehog proteins or mimetics, such as Shh or Dhh, in the culture can prevent loss of differentiation, or where fetal tissue is used, especially neuronal stem cells, can be S 30 used to induce differentiation.
Stem cells useful in the present invention are generally known. For example, several neural crest cells have been identified, some of which are multipotent and likely represent uncommitted neural crest cells, and others of which can generate only one type of cell, such as sensory neurons, and likely represent committed progenitor cells. The role of hedgehog proteins employed in the present method to culture such stem cells can be to induce differentiation of the uncommitted progenitor and thereby give rise to a committed progenitor cell, or to cause further restriction of the developmental fate of a committed progenitor cell towards becoming a terminally-differentiated neuronal cell. For example, the present method can be used in vitro to induce and/or maintain the differentiation of neural crest cells into glial cells, schwann cells, chromaffin cells, cholinergic sympathetic or parasympathetic neurons, as well as peptidergic and serotonergic neurons. The hedgehog protein can be used alone, or can be used in combination with other neurotrophic factors which act to more particularly enhance a particular differentiation fate of the neuronal progenitor cell. In the later instance, an hh polypeptide might be viewed as ensuring that the treated cell has achieved a particular phenotypic state such that the cell is poised along a certain developmental pathway so as to be properly induced upon contact with a secondary neurotrophic factor. In similar fashion, even relatively undifferentiated stem cells or primitive neuroblasts can be maintained in culture and caused to differentiate by treatment with hedgehog agonists. Exemplary primitive cell cultures comprise cells harvested from the neural plate or neural tube of an embryo even before much overt differentiation has occurred.
In addition to the implantation of cells cultured in the presence of a functional hedgehog activity and other in vitro uses described above, yet another aspect of the present invention concerns the therapeutic application of a hedgehog protein or mimetic to enhance survival of neurons and other neuronal cells in both the central nervous system and the peripheral nervous system. The ability of hedgehog protein to regulate neuronal 20 differentiation during development of the nervous system and also presumably in the adult state indicates that certain of the hedgehog proteins can be reasonably expected to facilitate control of adult neurons with regard to maintenance, functional performance, and aging of normal cells; repair and regeneration processes in chemically or mechanically lesioned cells; and prevention of degeneration and premature death which result from loss of differentiation in certain pathological conditions. In light of this understanding, the present invention specifically contemplates applications of the subject method to the treatment of (prevention and/or reduction of the severity of) neurological conditions deriving from: acute, subacute, or chronic injury to the nervous system, including traumatic injury, chemical injury, vasal injury and deficits (such as the ischemia resulting from stroke), together with 30 infectious/inflammatory and tumor-induced injury; (ii) aging of the nervous system including Alzheimer's disease; (iii) chronic neurodegenerative diseases of the nervous system, including Parkinson's disease, Huntington's chorea, amylotrophic lateral sclerosis and "the like, as well as spinocerebellar degenerations; and (iv) chronic immunological diseases of the nervous system or affecting the nervous system, including multiple sclerosis.
Many neurological disorders are associated with degeneration of discrete populations of neuronal elements and may be treatable with a therapeutic regimen which includes a hedgehog agonist. For example, Alzheimer's disease is associated with deficits in several neurotransmitter systems, both those that project to the neocortex and those that reside with the cortex. For instance, the nucleus basalis in patients with Alzheimer's disease have been observed to have a profound loss of neurons compared to age-matched controls.
Although Alzheimer's disease is by far the most common form of dementia, several other disorders can produce dementia. Several of these are degenerative diseases characterized by the death of neurons in various parts of the central nervous system, especially the cerebral cortex. However, some forms of dementia are associated with degeneration of the thalmus or the white matter underlying the cerebral cortex. Here, the cognitive dysfunction results from the isolation of cortical areas by the degeneration of efferents and afferents. Huntington's disease involves the degeneration of intrastraital and cortical cholinergic neurons and GABAergic neurons. Pick's disease is a severe neuronal degeneration in the neocortex of the frontal and anterior temporal lobes, sometimes accompanied by death of neurons in the striatum. Treatment of patients suffering from such degenerative conditions can include the application of hedgehog polypeptides, or agents which mimic their effects, in order to control, for example, differentiation and apoptotic events which give rise to loss of neurons to enhance survival of existing neurons) as well as promote differentiation and repopulation by progenitor cells in the area affected. In preferred embodiments, a source of a hedgehog agent is stereotactically provided within or proximate the area of degeneration. In addition to degenerative-induced dementias, a pharmaceutical preparation of one or more of the subject hedgehog proteins can be applied opportunely in the treatment of neurodegenerative disorders which have manifestations of tremors and involuntary movements. Parkinson's disease, for example, primarily affects subcortical structures and is characterized by degeneration of the nigrostriatal pathway, raphe nuclei, locus cereleus, and 5 the motor nucleus of vagus. Ballism is typically associated with damage to the subthalmic 25 nucleus, often due to acute vascular accident. Also included are neurogenic and myopathic diseases which ultimately affect the somatic division of the peripheral nervous system and are manifest as neuromuscular disorders. Examples include chronic atrophies such as amyotrophic lateral sclerosis, Guillain-Barre syndrome and chronic peripheral neuropathy, as well as other diseases which can be manifest as progressive bulbar palsies or spinal muscular 30 atrophies. The present method is amenable to the treatment of disorders of the cerebellum which result in hypotonia or ataxia, such as those lesions in the cerebellum which produce disorders in the limbs ipsilateral to the lesion. For instance, a preparation of a hedgehog homolog can be used to treat a restricted form of cerebellar cortical degeneration involving S'the anterior lobes (vermis and leg areas) such as is common in alcoholic patients.
In an illustrative embodiment, the subject method is used to treat amyotrophic lateral sclerosis. ALS is a name given to a complex of disorders that comprise upper and lower motor neurons. Patients may present with progressive spinal muscular atrophy, progressive bulbar palsy, primary lateral sclerosis, or a combination of these conditions. The major 57 pathological abnormality is characterized by a selective and progressive degeneration of the lower motor neurons in the spinal cord and the upper motor neurons in the cerebral cortex.
The therapeutic application of a hedgehog agonist, particularly Dhh, can be used alone, or in conjunction with other neurotrophic factors such as CNTF, BDNF or NGF to prevent and/or reverse motor neuron degeneration in ALS patients.
Hedgehog proteins of the present invention can also be used in the treatment of autonomic disorders of the peripheral nervous system, which include disorders affecting the innervation of smooth muscle and endocrine tissue (such as glandular tissue). For instance, the subject method can be used to treat tachycardia or atrial cardiac arrythmias which may arise from a degenerative condition of the nerves innervating the striated muscle of the heart.
Furthermore, a potential role for certain of the hedgehog proteins, which is apparent from the appended examples, mainly the data of respecting hedgehog expression in sensory and motor neurons of the head and trunk (including limb buds), concerns the role of hedgehog proteins in development and maintenance of dendritic processes of axonal neurons.
Potential roles for hedgehog proteins consequently include guidance for axonal projections and the ability to promote differentiation and/or maintenance of the innervating cells to their axonal processes. Accordingly, compositions comprising hedgehog agonists or other hedgehog agents described herein, may be employed to support, or alternatively antagonize the survival and reprojection of several types of ganglionic neurons sympathetic and sensory 20 neurons as well as motor neurons. In particular, such therapeutic compositions may be useful in treatments designed to rescue, for example, various neurons from lesion-induced death as S: well as guiding reprojection of these neurons after such damage. Such diseases include, but are not limited to, CNS trauma infarction, infection (such as viral infection with varicellazoster), metabolic disease, nutritional deficiency, toxic agents (such as cisplatin treatment).
Moreover, certain of the hedgehog agents (such as antagonistic form) may be useful in the selective ablation of sensory neurons, for example, in the treatment of chronic pain syndromes.
As appropriate, hedgehog agents can be used in nerve prostheses for the repair of central and peripheral nerve damage. In. particular, where a crushed or severed axon is a.
S 30 intubulated by use of a prosthetic device, hedgehog polypeptides can be added to the prosthetic device to increase the rate of growth and regeneration of the dendridic processes.
Exemplary nerve guidance channels are described in U.S. patents 5,092,871 and 4,955,892.
Accordingly, a severed axonal process can be directed toward the nerve ending from which it was severed by a prosthesis nerve guide which contains, e.g. a semi-solid formulation containing hedgehog polypeptide or mimetic, or which is derivatized along the inner walls with a hedgehog protein.
In another embodiment, the subject method can be used in the treatment of neoplastic or hyperplastic transformations such as may occur in the central nervous system. For instance, certain of the hedgehog proteins (or hh agonists) which induce differentiation of neuronal cells can be utilized to cause such transformed cells to become either post-mitotic or apoptotic. Treatment with a hedgehog agent may facilitate disruption of autocrine loops, such as TGF-P or PDGF autostimulatory loops, which are believed to be involved in the neoplastic transformation of several neuronal tumors. Hedgehog agonists may, therefore, be of use in the treatment of, for example, malignant gliomas, medulloblastomas, neuroectodermal tumors, and ependymonas.
Yet another aspect of the present invention concerns the application of the discovery that hedgehog proteins are morphogenic signals involved in other vertebrate organogenic pathways in addition to neuronal differentiation as described above, having apparent roles in other endodermal patterning, as well as both mesodermal and endodermal differentiation processes. As described in the Examples below, Shh clearly plays a role in proper limb growth and patterning by initiating expression of signaling molecules, including Bmp-2 in the mesoderm and Fgf-4 in the ectoderm. Thus, it is contemplated by the invention that compositions comprising hedgehog proteins can also be utilized for both cell culture and therapeutic methods involving generation and maintenance of non-neuronal tissue.
In one embodiment, the present invention makes use of the discovery that hedgehog 20 proteins, such as Shh, are apparently involved in controlling the development of stem cells responsible for formation of the digestive tract, liver, lungs, and other organs which derive from the primitive gut. As described in the Examples below, Shh serves as an inductive signal from the endoderm to the mesoderm, which is critical to gut morphogenesis.
Therefore, for example, hedgehog agonists can be employed in the development and maintenance of an artificial liver which can have multiple metabolic functions of a normal liver. In an exemplary embodiment, hedgehog agonists can be used to induce differentiation of digestive tube stem cells to form hepatocyte cultures which can be used to populate extracellular matrices, or which can be encapsulated in biocompatible polymers, to form both implantable and extracorporeal artificial livers.
30 In another embodiment, therapeutic compositions of hedgehog agonists can be utilized in conjunction with transplantation of such artificial livers, as well as embryonic liver S"structures, to promote intraperitoneal implantation, vascularization, and in vivo differentiation and maintenance of the engrafted liver tissue.
In yet another embodiment, hedgehog agonists can be employed therapeutically to regulate such organs after physical, chemical or pathological insult. For instance, therapeutic compositions comprising hedgehog agonists can be utilized in liver repair subsequent to a partial hepatectomy. Similarly, therapeutic compositions containing hedgehog agonists can be used to promote regeneration of lung tissue in the treatment of emphysema.
In still another embodiment of the present invention, compositions comprising hedgehog agonists can be used in the in vitro generation of skeletal tissue, such as from skeletogenic stem cells, as well as the in vivo treatment of skeletal tissue deficiencies. The present invention particularly contemplates the use of hedgehog agonists which maintain a skeletogenic activity, such as an ability to induce chondrogenesis and/or osteogenesis. By "skeletal tissue deficiency", it is meant a deficiency in bone or other skeletal connective tissue at any site where it is desired to restore the bone or connective tissue, no matter how the deficiency originated, e.g. whether as a result of surgical intervention, removal of tumor, ulceration, implant, fracture, or other traumatic or degenerative conditions.
For instance, the present invention makes available effective therapeutic methods and compositions for restoring cartilage function to a connective tissue. Such methods are useful in, for example, the repair of defects or lesions in cartilage tissue which is the result of degenerative wear such as, that which results in arthritis, as well as other mechanical derangements which may be caused by trauma to the tissue, such as a displacement of torn meniscus tissue, meniscectomy, a laxation of a joint by a torn ligament, malignment ofjoints, bone fracture, or by hereditary disease. The present reparative method is also useful for remodeling cartilage matrix, such as in plastic or reconstructive surgery, as well as 20 periodontal surgery. The present method may also be applied to improving a previous reparative procedure, for example, following surgical repair of a meniscus, ligament, or cartilage. Furthermore, it may prevent the onset or exacerbation of degenerative disease if applied early enough after trauma.
In one embodiment of the present invention, the subject method comprises treating the afflicted connective tissue with a therapeutically sufficient amount of a hedgehog agonist, particularly an Ihh agonist, to generate a cartilage repair response in the connective tissue by stimulating the differentiation and/or proliferation of chondrocytes embedded in the tissue.
Induction of chondrocytes by treatment with a hedgehog agonist can subsequently result in the synthesis of new cartilage matrix by the treated cells. Such connective tissues as articular cartilage, interarticular cartilage (menisci), costal cartilage (connecting the true ribs and the sternum), ligaments, and tendons are particularly amenable to treatment in reconstructive and/or regenerative therapies using the subject method. As used herein, regenerative therapies include treatment of degenerative states which have progressed to the point of which impairment of the tissue is obviously manifest, as well as preventive treatments of tissue where degeneration is in its earliest stages or imminent. The subject method can further be used to prevent the spread of mineralisation into fibrotic tissue by maintaining a constant production of new cartilage.
(0 In an illustrative embodiment, the subject method can be used to treat cartilage of a diarthroidal joint, such as a knee, an ankle, an elbow, a hip, a wrist, a knuckle of either a finger or toe, or a temperomandibular joint. The treatment can be directed to the meniscus of the joint, to the articular cartilage of the joint, or both. To further illustrate, the subject method can be used to treat a degenerative disorder of a knee, such as which might be the result of traumatic injury a sports injury or excessive wear) or osteoarthritis. An injection of a hedgehog agonist into the joint with, for instance, an arthroscopic needle, can be used to treat the afflicted cartilage. In some instances, the injected agent can be in the form of a hydrogel or other slow release vehicle described above in order to permit a more extended and regular contact of the agent with the treated tissue.
The present invention further contemplates the use of the subject method in the field of cartilage transplantation and prosthetic device therapies. To date, the growth of new cartilage from either transplantation of autologous or allogenic cartilage has been largely unsuccessful. Problems arise, for instance, because the characteristics of cartilage and fibrocartilage varies between different tissue: such as between articular, meniscal cartilage, ligaments, and tendons, between the two ends of the same ligament or tendon, and between the superficial and deep parts of the tissue. The zonal arrangement of these tissues may reflect a gradual change in mechanical properties, and failure occurs when implanted tissue, which has not differentiated under those conditions, lacks the ability to appropriately respond.
For instance, when meniscal cartilage is used to repair anterior cruciate ligaments, the tissue undergoes a metaplasia to pure fibrous tissue. By promoting chondrogenesis, the subject method can be used to particularly addresses this problem, by causing the implanted cells to become more adaptive to the new environment and effectively resemble hypertrophic chondrocytes of an earlier developmental stage of the tissue. Thus, the action of 25 chondrogensis in the implanted tissue, as provided by the subject method, and the mechanical forces on the actively remodeling tissue can synergize to produce an improved implant more suitable for the new function to which it is to be put.
In similar fashion, the subject method can be applied to enhancing both the generation of prosthetic cartilage devices and to their implantation. The need for improved treatment 30 has motivated research aimed at creating new cartilage that is based on collagenglycosaminoglycan templates (Stone et al. (1990) Clin Orthop Relat Red 252:129), isolated chondrocytes (Grande et al. (1989) J Orthop Res 7:208; and Takigawa et al. (1987) Bone Miner 2:449), and chondrocytes attached to natural or synthetic polymers (Walitani et al.
(1989) J Bone Jt Surg 71B:74; Vacanti et al. (1991) Plast Reconstr Surg 88:753; von Schroeder et al. (1991) JBiomed Mater Res 25:329; Freed et al. (1993) JBiomed Mater Res 27:11; and the Vacanti et al. U.S. Patent No. 5,041,138). For example, chondrocytes can be grown in culture on biodegradable, biocompatible highly porous scaffolds formed from polymers such as polyglycolic acid, polylactic acid, agarose gel, or other polymers which degrade over time as function of hydrolysis of the polymer backbone into innocuous monomers. The matrices are designed to allow adequate nutrient and gas exchange to the cells until engraftment occurs. The cells can be cultured in vitro until adequate cell volume and density has developed for the cells to be implanted. One advantage of the matrices is that they can be cast or molded into a desired shape on an individual basis, so that the final product closely resembles the patient's own ear or nose (by way of example), or flexible matrices can be used which allow for manipulation at the time of implantation, as in a joint.
In one embodiment of the subject method, the implants are contacted with a hedgehog agonist during the culturing process, such as an Ihh agonist, in order to induce and/or maintain differentiated chondrocytes in the culture in order as to further stimulate cartilage matrix production within the implant. In such a manner, the cultured cells can be caused to maintain a phenotype typical of a chondrogenic cell hypertrophic), and hence continue the population of the matrix and production of.cartilage tissue.
In another embodiment, the implanted device is treated with a hedgehog agonist in order to actively remodel the implanted matrix and to make it more suitable for its intended function. As set out above with respect to tissue transplants, the artificial transplants suffer from the same deficiency of not being derived in a setting which is comparable to the actual mechanical environment in which the matrix is implanted. The activation of the chondrocytes in the matrix by the subject method can allow the implant to acquire characteristics similar to the tissue for which it is intended to replace.
In yet another embodiment, the subject method is used to enhance attachment of prosthetic devices. To illustrate, the subject method can be used in the implantation of a S. periodontal prosthesis, wherein the treatment of the surrounding connective tissue stimulates formation of periodontal ligament about the prosthesis, as well as inhibits formation of fibrotic tissue proximate the prosthetic device.
In still further embodiments, the subject method can be employed for the generation of bone (osteogenesis) at a site in the animal where such skeletal tissue is deficient. Indian hedgehog is particularly associated with the hypertrophic chondrocytes that are ultimately replaced by osteoblasts. For instance, administration of a hedgehog agent of the present invention can be employed as part of a method for treating bone loss in a subject, e.g. to prevent and/or reverse osteoporosis and other osteopenic disorders, as well as to regulate bone growth and maturation. For example, preparations comprising hedgehog agonists can be employed, for example, to induce endochondral ossification, at least so far as to facilitate the formation of cartilaginous tissue precursors to form the "model" for ossification.
Therapeutic compositions of hedgehog agonists can be supplemented, if required, with other 62 osteoinductive factors, such as bone growth factors TGF-3 factors, such as the bone morphogenetic factors BMP-2 and BMP-4, as well as activin), and may also include, or be administered in combination with, an inhibitor of bone resorption such as estrogen, bisphosphonate, sodium fluoride, calcitonin, or tamoxifen, or related compounds. However, it will be appreciated that hedgehog proteins, such as Ihh and Shh are likely to be upstream of BMPs, e.g. hh treatment will have the advantage of initiating endogenous expression of BMPs along with other factors.
In yet another embodiment of the present invention, a hedgehog antagonist can be used to inhibit spermatogenesis. Thus, in light of the present finding that hedgehog proteins are involved in the differentiation and/or proliferation and maintenance of testicular germ cells, hedgehog antagonist can be utilized to block the action of a naturally-occurring hedgehog protein. In a preferred embodiment, the hedgehog antagonist inhibits the biological activity of Dhh with respect to spermatogenesis, by competitively binding hedgehog receptors in the testis. In similar fashion, hedgehog agonists and antagonists are potentially useful for modulating normal ovarian function.
The source of hedgehog polypeptides, whether for cell culture or for in vivo application, can be in the form of a purified protein composition, or can eb from a cell expressing either a recombinant or endogenous form of the polypeptide, such as embryonic tissue floor plate tissue explants). Moreover, is addition to those forms of the vertebrate hedgehog polypeptides described herein, the present invention further contemplates the use of the drosophila hedgehog (Dros-HH) protein to induce cells and tissue of vertebrate organisms.
In the instance of protein compositions, the hedgehog protein, or a pharmaceutically acceptable salt thereof, may be conveniently formulated for administration with a biologically acceptable medium, such as water, buffered saline, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like) or suitable mixtures thereof. The optimum **concentration of the active ingredient(s) in the chosen medium can be determined empirically, according to procedures well known to medicinal chemists. As used herein, "biologically acceptable medium" includes any and all solvents, dispersion media, and the 30 like which may be appropriate for the desired route of administration of the pharmaceutical preparation. The use of such media for pharmaceutically active substances is known in the art. Except insofar as any conventional media or agent is incompatible with the activity of the hedgehog protein, its use in the pharmaceutical preparation of the invention is contemplated. Suitable vehicles and their formulation inclusive of other proteins are described, for example, in the book Remington's Pharmaceutical Sciences (Remington's Pharmaceutical Sciences. Mack Publishing Company, Easton, Pa., USA 1985). These vehicles include injectable "deposit formulations". Based on the above, such pharmaceutical (o3 formulations include, although not exclusively, solutions or freeze-dried powders of a hedgehog homolog (such as a Shh, Dhh or Mhh) in association with one or more pharmaceutically acceptable vehicles or diluents, and contained in buffered media at a suitable pH and isosmotic with physiological fluids. For illustrative purposes only and without being limited by the same, possible compositions or formulations which may be prepared in the form of solutions for the treatment of nervous system disorders with a hedgehog protein are given in U.S. Patent No. 5,218,094. In the case of freeze-dried preparations, supporting excipients such as, but not exclusively, mannitol or glycine may be used and appropriate buffered solutions of the desired volume will be provided so as to obtain adequate isotonic buffered solutions of the desired pH. Similar solutions may also be used for the pharmaceutical compositions of hh in isotonic solutions of the desired volume and include, but not exclusively, the use of buffered saline solutions with phosphate or citrate at suitable concentrations so as to obtain at all times isotonic pharmaceutical preparations of the desired pH, (for example, neutral pH).
Methods of introduction of exogenous hh at the site of treatment include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, oral, intranasal and topical. In addition, it may be desirable to introduce the pharmaceutical compositions of the invention into the central nervous system by any suitable route, including intraventricular and intrathecal injection. Intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
Methods of introduction may also be provided by rechargeable or biodegradable devices. Various slow release polymeric devices have been developed and tested in vivo in recent years for the controlled delivery of drugs, including proteinacious biopharmaceuticals.
A variety of biocompatible polymers (including hydrogels), including both biodegradable and non-degradable polymers, can be used to form an implant for the sustained release of an hh at a particular target site. Such embodiments of the present invention can be used for the delivery of an exogenously purified hedgehog protein, which has been incorporated in the polymeric device, or for the delivery of hedgehog produced by a cell encapsulated in the polymeric device.
30 An essential feature of certain embodiments of the implant can be the linear release of the hh, which can be achieved through the manipulation of the polymer composition and form. By choice of monomer composition or polymerization technique, the amount of water, porosity and consequent permeability characteristics can be controlled. The selection of the shape, size, polymer, and method for implantation can be determined on an individual basis according to the disorder to be treated and the individual patient response. The generation of such implants is generally known in the art. See, for example, Concise Encyclopedia of Medical Dental Materials, ed. by David Williams (MIT Press: Cambridge, MA, 1990); and the Sabel et al. U.S. Patent No. 4,883,666. In another embodiment of an implant, a source of cells producing a hedgehog protein, or a solution of hydogel matrix containing purified hh, is encapsulated in implantable hollow fibers. Such fibers can be pre-spun and subsequently loaded with the hedgehog source (Aebischer et al. U.S. Patent No. 4,892,538; Aebischer et al. U.S. Patent No. 5,106,627; Hoffman et al. (1990) Expt. Neurobiol. 110:39- 44; Jaeger et al. (1990) Prog. Brain Res. 82:41-46; and Aebischer et al. (1991) J. Biomech.
Eng. 113:178-183), or can be co-extruded with a polymer which acts to form a polymeric coat about the hh source (Lim U.S. Patent No. 4,391,909; Sefton U.S. Patent No. 4,353,888; Sugamori et al. (1989) Trans. Am. Artif Intern. Organs 35:791-799; Sefton et al. (1987) Biotehnol. Bioeng. 29:1135-1143; and Aebischer et al. (1991) Biomaterials 12:50-55).
In yet another embodiment of the present invention, the pharmaceutical hedgehog protein can be administered as part of a combinatorial therapy with other agents. For example, the combinatorial therapy can include a hedgehog protein with at least one trophic factor. Exemplary trophic factors include nerve growth factor, cilliary neurotrophic growth factor, schwanoma-derived growth factor, glial growth factor, stiatal-derived neuronotrophic factor, platelet-derived growth factor, and :;catter factor (HGF-SF). Antimitogenic agents can also be used, for example, when proliferation of surrounding glial cells or astrocytes is undesirable in the regeneration of nerve cells. Examples of such antimitotic agents include cytosine, arabinoside, 5-fluorouracil, hydrcxyurea, and methotrexate.
20 Another aspect of the invention features transgenic non-human animals which express Sa heterologous hedgehog gene of the present invention, or which have had one or more genomic hedgehog genes disrupted in at least one of the tissue or cell-types of the animal.
Accordingly, the invention features an animal model for developmental diseases, which animal has hedgehog allele which is mis-expressed. For example, a mouse can be bred which has one or more hh alleles deleted or otherwise rendered inactive. Such a mouse model can then be used to study disorders arising from mis-expressed hedgehog genes, as well as for evaluating potential therapies for similar disorders.
Another aspect of the present invention concerns transgenic animals which are comprised of cells (of that animal) which contain a transgene of the present invention and 30 which preferably (though optionally) express an exogenous hedgehog protein in one or more cells in the animal. A hedgehog transgene can encode the wild-type form of the protein, or can encode homologs thereof, including both agonists and antagonists, as well as antisense constructs. In preferred embodiments, the expression of the transgene is restricted to specific subsets of cells, tissues or developmental stages utilizing, for example, cis-acting sequences that control expression in the desired pattern. In the present invention, such mosaic expression of a hedgehog protein can be essential for many forms of lineage analysis and can additionally provide a means to assess the effects of, for example, lack of hedgehog expression which might grossly alter development in small patches of tissue within an otherwise normal embryo. Toward this end, tissue-specific regulatory sequences and conditional regulatory sequences can be used to control expression of the transgene in certain spatial pattems. Moreover, temporal patterns of expression can be provided by, for example, conditional recombination systems or prokaryotic transcriptional regulatory sequences.
Genetic techniques which allow for the expression of transgenes can be regulated via site-specific genetic manipulation in vivo are known to those skilled in the art. For instance, genetic systems are available which allow for the regulated expression of a recombinase that catalyzes the genetic recombination a target sequence. As used herein, the phrase "target sequence" refers to a nucleotide sequence that is genetically recombined by a recombinase.
The target sequence is flanked by recombinase recognition sequences and is generally either excised or inverted in cells expressing recombinase activity. Recombinase catalyzed recombination events can be designed such that recombination of the target sequence results in either the activation or repression of expression of one of the subject hedgehog proteins.
For example, excision of a target sequence which interferes with the expression of a recombinant hh gene, such as one which encodes an antagonistic homolog or an antisense transcript, can be designed to activate expression of that gene. This interference with expression of the protein can result from a variety of mechanisms, such as spatial separation of the hh gene from the promoter element or an internal stop codon. Moreover, the transgene can be made wherein the coding sequence of the gene is flanked by recombinase recognition sequences and is initially transfected into cells in a 3' to 5' orientation with respect to the promoter element. In such an instance, inversion of the target sequence will reorient the subject gene by placing the 5' end of the coding sequence in an orientation with respect to the promoter element which allow for promoter driven transcriptional activation.
25 In an illustrative embodiment, either the crelloxP recombinase system of bacteriophage P1 (Lakso et al. (1992) PNAS 89:6232-6236; Orban et al. (1992) PNAS 89:6861-6865) or the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al.
(1991) Science 251:1351-1355; PCT publication WO 92/15694) can be used to generate in vivo site-specific genetic recombination systems. Cre recombinase catalyzes the site-specific S 30 recombination of an intervening target sequence located between loxP sequences. loxP sequences are 34 base pair nucleotide repeat sequences to which the Cre recombinase binds and are required for Cre recombinase mediated genetic recombination. The orientation of loxP sequences determines whether the intervening target sequence is excised or inverted when Cre recombinase is present (Abremski et al. (1984) J. Biol. Chem. 259:1509-1514); catalyzing the excision of the target sequence when the loxP sequences are oriented as direct repeats and catalyzes inversion of the target sequence when loxP sequences are oriented as inverted repeats.
Accordingly, genetic recombination of the target sequence is dependent on expression of the Cre recombinase. Expression of the recombinase can be regulated by promoter elements which are subject to regulatory control, tissue-specific, developmental stage-specific, inducible or repressible by externally added agents. This regulated control will result in genetic recombination of the target sequence only in cells where recombinase expression is mediated by the promoter element. Thus, the activation expression of a recombinant hedgehog protein can be regulated via control of recombinase expression.
Use of the crelloxP recombinase system to regulate expression of a recombinant hh protein requires the construction of a transgenic animal containing transgenes encoding both the Cre recombinase and the subject protein. Animals containing both the Cre recombinase and a recombinant hedgehog gene can be provided through the construction of "double" transgenic animals. A convenient method for providing such animals is to mate two transgenic animals each containing a transgene, an hh gene and recombinase gene.
One advantage derived from initially constructing transgenic animals containing a hedgehog transgene in a recombinase-mediated expressible format derives from the likelihood that the subject protein, whether agonistic or antagonistic, can be deleterious upon expression in the transgenic animal. In such an instance, a founder population, in which the subject transgene is silent in all tissues, can be propagated and maintained. Individuals of this founder population can be crossed with animals expressing the recombinase in, for example, one or more tissues andcor a desired temporal pattern. Thus, the creation of a founder population in which, for example, an antagonistic hh transgene is silent will allow the study of progeny from that founder in which disruption of hedgehog mediated induction in a particular tissue or at certain developmental stages would result in, for example, a lethal phenotype.
25 Similar conditional transgenes can be provided using prokaryotic promoter sequences which require prokaryotic proteins to be simultaneous expressed in order to facilitate expression of the hedgehog transgene. Exemplary promoters and the corresponding transactivating prokaryotic proteins are given in U.S. Patent No. 4,833,080.
Moreover, expression of the conditional transgenes can be induced by gene therapy- 30 like methods whelin a gene encoding the trans-activating protein, e.g. a recombinase or a prokaryotic protein, is delivered to the tissue and caused to be expressed, such as in a celltype specific manner. By this method, a hedgehog transgene could remain silent into adulthood until "turned on" by the introduction of the trans-activator.
In an exemplary embodiment, the "transgenic non-human animals" of the invention are produced by introducing transgenes into the germline of the non-human animal.
Embryonic target cells at various developmental stages can be used to introduce transgenes.
Different methods are used depending on the stage of development of the embryonic target cell. The zygote is the best target for micro-injection. In the mouse, the male pronucleus reaches the size of approximately 20 micrometers in diameter which allows reproducible injection of 1-2pl of DNA solution. The use of zygotes as a target for gene transfer has a major advantage in that in most cases the injected DNA will be incorporated into the host gene before the first cleavage (Brinster et al. (1985) PNAS 82:4438-4442). As a consequence, all cells of the transgenic non-human animal will carry the incorporated transgene. This will in general also be reflected in the efficient transmission of the transgene to offspring of the founder since 50% of the germ cells will harbor the transgene. Microinjection of zygotes is the preferred method for incorporating transgenes in practicing the invention.
Retroviral infection can also be used to introduce hedgehog transgenes into a nonhuman animal. The developing non-human embryo can be cultured in vitro to the blastocyst stage. During this time, the blastomeres can be targets for retroviral infection (Jaenich, R.
(1976) PNAS 73:1260-1264). Efficient infection of the blastomeres is obtained by enzymatic treatment to remove the zona pellucida (Manipulating the Mouse Embryo, Hogan eds. (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1986). The viral vector system used to introduce the transgene is typically a replication-defective retrovirus carrying the transgene S(Jahner et al. (1985) PNAS 82:6927-6931; Van der Putten et al. (1985) PNAS 82:6148-6152).
Transfection is easily and efficiently obtained by culturing the blastomeres on a monolayer of virus-producing cells (Van der Putten, supra; Stewart ei al. (1987) EMBO J. 6:383-388).
Alternatively, infection can be performed at a later stage. Virus or virus-producing cells can be injected into the blastocoele (Jahner et al. (1982) Nature 298:623-628). Most of the Sfounders will be mosaic for the transgene since incorporation occurs only in a subset of the cells which formed the transgenic non-hurnan animal. Further, the founder may contain 25 various retroviral insertions of the transgene at different positions in the genome which generally will segregate in the offspring. In addition, it is also possible to introduce transgenes into the germ line by intrauterine retroviral infection of the midgestation embryo (Jahner et al. (1982) supra).
A third type of target cell for transgene introduction is the embryonic stem cell (ES).
ES cells are obtained from pre-implantation embryos cultured in vitro and fused with embryos (Evans et al. (1981) Nature 292:154-156; Bradley et al. (1984) Nature 309:255-258; 0:060* Gossler et al. (1986) PNAS 83: 9065-9069; and Robertson et al. (1986) Nature 322:445-448).
Transgenes can be efficiently introduced into the ES cells by DNA transfection or by retrovirus-mediated transduction. Such transformed ES cells can thereafter be combined with 35 blastocysts from a non-human animal. The ES cells thereafter colonize the embryo and contribute to the germ line of the resulting chimeric animal. For review see Jaenisch, R.
(1988) Science 240:1468-1474.
Methods of making hedgehog knock-out or disruption transgenic animals are also generally known. See, for example, Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1986). Recombinase dependent knockouts can also be generated, e.g. by homologous recombination to insert recombinase target sequences flanking portions of an endogenous hh gene, such that tissue specific and/or temporal control of inactivation of a hedgehog allele can be controlled as above.
Exemplification The invention, now being generally described, will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain aspects and embodiments of the present invention and are not intended to limit the invention.
Example 1 Cloning and Expression of Chick Sonic Hedgehog Experimental Procedures Using degenerate PCR primers, vHH50 (SEQ ID No:18), vHH30 (SEQ ID No:19) and vHH3I (SEQ ID No:20) corresponding to a sequence conserved between Drosophila hedgehog (Dros-HH)(SEQ ID No:34) (Lee, J.J. et al. (1992) Cell 71: 33-50; Mohler, J. et al., 20 (1992) Development 115: 957-971) and mouse Indian hedgehog (Ihh) (SEQ ID No:10), a 220 base pair (bp) fragment was amplified from chicken genomic DNA. From 15 isolates, two distinct sequences were cloned, pCHA (SEQ ID No:35) and pCHB (SEQ ID No:36), each highly homologous to mouse lhh (Figure A probe made from isolate pCHA did not detect expression in embryonic tissues. Isolate pCHB, however, detected a 4 kb message in RNA prepared from embryonic head, trunk, or limb bud RNA. This cloned PCR fragment was therefore used as a probe to screen an unamplified cDNA library prepared from SHamburger Hamilton stage 22 (Hamburger, W. et al., (1951) J. Morph. 88: 49-92) limb bud RNA as described below.
A single 1.6 kilobase (kb) cDNA clone, pHH-2, was selected for characterization and 30 was used in all subsequent analyses. The gene encoding for this cDNA was named Sonic Hedgehog (after the Sega computer game cartoon character). Sequencing of the entire cDNA confirmed the presence of a single long open reading frame potentially encoding for a protein of 425 amino acids The clone extends 220 bp upstream of the predicted initiator methionine and approximately 70 bp beyond the stop codon. No consensus polyadenylation signal could be identified in the 3' untranslated region. A second potential initiator methionine occurs at amino acid residue 4. The putative translation initiation signals surrounding both methionines are predicted to be equally efficient (Kozak, (1987) Nuc.
Acids Res. 15: 8125-8132). When the pHH-2 Sonic cDNA is used to probe a northern blot of stage 24 embryonic chick RNA, a single mRNA species of approximately 4 kb is detected in both limb and trunk tissue. The message size was predicted by comparing it to the position of 18S and 28S ribosomal RNA. Hybridized mRNA was visualized after a two day exposure to a phosphoscreen. Because the Sonic cDNA clone pHH-2 is only 1.6 kb, it is likely to be missing approximately 2.4 kb of untranslated sequence.
PCR Cloning All standard cloning techniques were performed according to Ausubel et. al. (1989), and all enzymes were obtained from Boehringer Mannheim Biochemicals. Degenerate oligonucleotides corresponding to amino acid residues 161 to 237 of the Drosophila hedgehog protein (SEQ ID No:34) (Lee, J.J. et. al., (1992) Cell 71: 33-50) were synthesized.
These degenerate oligonucleotides, vHH50 (SEQ ID No: 18), vHH30 (SEQ ID No: 19), and vHH31 (SEQ ID No:20) also contained Eco RI, Cla I, and Xba I sites, respectively, on their ends to facilitate subcloning. The nucleotide sequence of these oligos is given below: 5'-GGAATTCCCAG(CA)GITG(CT)AA(AG)GA(AG)(CA)(AG)I(GCT)IAA-3' 5'-TCATCGATGGACCCA(GA)TC(GA)AAICCIGC(TC)TC-3' vHH3I: 5'-GCTCTAGAGCTCIACIGCIA(GA)IC(GT)IGC-3' where I represents inosine. Nested PCR was performed by first amplifying chicken genomic DNA using the vHH50 and vHH30 primer pair and then further amplifying that product using the vHH50 and vHH3I primer pair. In each case the reaction conditions were: initial denaturation at 930 C for 2.5 min., followed by 30 cycles of 940 C for 45 s, 500 C for 1 min., 25 720 C for 1, and a final incubation of 72° C for 5 min. The 220 bp PCR product was subcloned into pGEM7zf (Promega). Two unique clones, pCHA (SEQ ID No:35) and pCHB (SEQ ID No:36) were identified.
DNA Sequence Analysis Nucleotide sequences were determined by the dideoxy chain termination method 30 (Sanger, F. et al., (1977) Proc. Natl. Acad. Sci. USA 74: 5463-5467) using Sequenase v2.0 T7 DNA polymerase (US Biochemicals). 5' and 3' nested deletions ofpHH-2 were generated by using the nucleases Exo III and SI (Erase a Base, Promega) and individual subclones sequenced. DNA and amino acid sequences were analyzed using both GCG (Devereux, J. et al., (1984) Nuc. Acids Res. 12: 387-394) and DNAstar software. Searches for related 76> sequences were done through the BLAST network service (Altschul, S.F. et al., (1990) J.
Mol. Biol. 215: 403-410) provided by the National Center for Biotechnology Information.
Southern Blot Analysis Five p.g of chick genomic DNA was digested with Eco RI and/or Barn HI, fractionated on a 1% agarose gel, and transferred to a nylon membrane (Genescreen, New England Nuclear). The filters were probed with 3 2 P-labeled hha or hhb at 42 0 C in hybridization buffer BSA, 500 mM NaHPO 4 7% SDS, 1 mM EDTA, pH 7.2; Church, G.M. et al., (1984) Proc. Natl. Acad. Sci. USA 81: 1991-1995). The blots were washed at 630 C once in 0.5% bovine serum albumin, 50 mM NaHPO4 (pH 5% SDS, 1 mM EDTA and twice in 40 mM NaHPO4 (pH 1% SDS, ImM EDTA, and visualized on Kodak film.
Isolation Of Chicken Sonic cDNA Clones A stage 22 limb bud cDNA library was constructed in Xgtl0 using Eco RI/NotI linkers. Unamplified phage plaques (106) were transferred to nylon filters (Colony/Plaque screen, NEN) and screened with a 3 2 P-labelled pooled inserts from PCR clones pCHA (SEQ ID No:35) and pCHB (SEQ ID No:36). Hybridization was performed at 420 C in formamide 2X SSC, 10% dextran sulfate, 1% SDS and washing as described in the Southern Blot procedure. Eight positive plaques were identified, purified and their cDNA inserts excised with EcoRI and subcloned into pBluescript SK+ (Stratagene). All eight had approximately 1.7 kb inserts with identical restriction patterns. One, pHH-2, was chosen for sequencing and used in all further manipulations.
Preparation Of Digoxigenin-Labeled Riboprobes Plasmid pHH-2 was linearized with Hind III and transcribed with T3 RNA polymerase (for antisense probes) or with Barn HI and transcribed with T7 RNA polymerase according to the manufacturers instructions for the preparation of non-radioactive digoxigenin transcripts. Following the transcription reaction, RNA was precipitated, and resuspended in RNAse-free water.
Whole Mount In Situ Hybridization Whole-mount in situ hybridization was performed using protocols modified from Parr, B.A. et al.. (1993) Development 119: 247-261; Sasaki, H. et al. (1993) Development 118: 47-59; Rosen, B. et al. (1993) Trends Genet. 9: 162-167. Embryos from incubated fertile White Leghorn eggs (Spafas) were removed from the egg and extra-embryonic membranes dissected in calcium/magnesium-free phosphate-buffered saline (PBS) at room temperature. Unless otherwise noted, all washes are for five minutes at room temperature.
71 Embryos were fixed overnight at 4°C with 4% paraformaldehyde in PBS, washed twice with PBT (PBS with 0.1% Tween-20) at 4 0 C, and dehydrated through an ascending methanol series in PBT 50%, 75%, 2 X 100% methanol). Embryos were stored at -20 0 C until further use.
Both pre-limb bud and limb bud stage embryos were rehydrated through an descending methanol series followed by two washes in PBT. Limb bud stage embryos were bleached in 6% hydrogen peroxide in PBT, washed three times with PBT, permeabilized with proteinase K (Boehringer, 2 (pg/ml) for 15 minutes, washed with 2 mg/ml glycine in PBT for minutes, and twice with PBT. Pre-limb bud stage embryos were permealibized (without prior incubation with hydrogen peroxide) by three 30 minute washes in RIPA buffer (150 mM NaCI, 1% NP-40, 0.5% Deoxycholate, 0.1% SDS, ImM EDTA, 50 mM Tris-HCI, pH In all subsequent steps, pre-limb bud and limb bud stage embryos were treated equivalently. Embryos were fixed with 4% paraformaldehyde/0.2% gluteraldehyde in PBT, washed four times with PBT, once with pre-hybridization buffer (50% formamide, 5 X SSC, 1% SDS, 50 pg/ml total yeast RNA, 50 tg/ml heparin, pH and incubated with fresh prehybridization buffer for one hour at 70 0 C. The pre-hybridization buffer was then replaced with hybridization buffer (pre-hybridization buffer with digoxigenin labeled riboprobe at 1 pg/ml) and incubated overnight at 70 0
C.
Following hybridization, embryos were washed 3 X 30 minutes at 70 0 C with solution 1 (50% formamide, 5 X SSC, 1% SDS, pH 3 X 30 minutes at 70 0 C with solution 3 formamide, 2 X SSC, pH and three times at room temperature with TBS (Trisbuffered saline with 2 mM levamisole) containing 0.1% Tween-20. Non-specific binding of antibody was prevented by preblocking embryos in TBS/0.1% Tween-20 containing heat-inactivated sheep serum for 2.5 hours at room temperature and by pre-incubating anti- 25 digoxigenin Fab alkaline-phosphatase conjugate (Boehringer) in TBS/0.1% containing heat inactivated 1% sheep serum and approximately 0.3% heat inactivated chick embryo powder. After an overnight incubation at 4°C with the pre-adsorbed antibody in TBS/0.1% Tween-20 containing 1% sheep serum, embryos were washed 3 X 5 minutes at room temperature with TBS/0.1% Tween-20, 5 X 1.5 hour room temperature washes with TBS/1% Tween-20, and overnight with TBS/1% Tween-20 at 4 0 C. The buffer was exchanged by washing 3 X 10 minutes with NTMT (100mM NaC1, 100 mM Tris-HC1, mM MgC12, 0.1% Tween-20, 2 mM levamisole). The antibody detection reaction was performed by incubating embryos with detection solution (NTMT with 0.25 mg/ml NBT and 0.13 mg/ml X-Phos). In general, pre-limb bud stage embryos were incubated for 5-15 hours 35 and limb bud stage embryos 1-5 hours. After the detection reaction was deemed complete, embryos were washed twice with NTMT, once with PBT (pH postfixed with 4% paraformaldehyde/0.1% gluteraldehyde in PBT, and washed several times with PBT. In 72 some cases embryos were cleared through a series of 30%, 50%, 70%, and 80% glycerol in PBT. Whole embryos were photographed under transmitted light using a Nikon zoom stereo microscope with Kodak Ektar 100 ASA film. Selected embryos were processed for frozen sections by dehydration in 30% sucrose in PBS followed by embedding in gelatin and freezing. 25 p.m cryostat sections were collected on superfrost plus slides (Fisher), rehydrated in PBS, and mounted with gelvatol. Sections were photographed with Nomarski optics using a Zeiss Axiophot microscope and Kodak Ektar 25 ASA film.
(ii) Sequence Homolgy Comparison Between Chicken Sonic hh And Dros-HH And Other Vertebrate Sonic hh Proteins The deduced Sonic amino acid sequence (SEQ ID No:8) is shown and compared to the Drosophila hedgehog protein (SEQ ID No:34) in Figure 2. Over the entire open reading frame the two proteins are 48% homologous at the amino acids level. The predicted Drosophila protein extends 62 aa beyond that of Sonic at its amino terminus. This Nterminal extension precedes the putative signal peptide (residues 1-26) of the fly protein (SEQ ID No:34), and has been postulated to be removed during processing of the secreted form of Drosophila hedgehog (Lee, J.J. et al., (1992) Cell 71: 33-50). The sequence of residues 1-26 of the Sonic protein (SEQ ID No:8) matches well with consensus sequences for eukaryotic signal peptides (Landry, S.J. et al., (1993) Trends. Biochem. Sci. 16: 159-163) and is therefore likely to serve that function for Sonic. Furthermore, Figure 3 shows a hydropathy plot (Kyte, J. et-al., (1982) J. Mol. Biol.157: 133-148) indicating that residues 1-26 of the Sonic protein (SEQ ID No:8) exhibit a high hydrophobic moment in accord with identified S: eukaryotic signal peptides. Cleavage of the putative signal sequence should occur C-terminal to residue 26 according to the predictive method of von Henjie, G. (1986) Nucl. Acid. Res.
I o 11: 1986. A single potential N-linked glycosylation site is located at amino acid residue 282 25 of the Sonic protein (SEQ ID No:8). The predicted Sonic protein does not contain any other strong consensus motifs, and is not homologous to any other proteins outside of the Hedgehog family.
The mouse (SEQ ID No: 11) and zebrafish (SEQ ID No:12) homologs of Sonic have also been isolated. A comparison of these and the Drosophila sequence is shown schematically in Figure 4. All of the vertebrate proteins have a similar predicted structure: a putative signal peptide at their amino terminus, followed by an extraordinarily similar 182 amino acid region (99% identity in chicken versus mouse and 95% identity in chicken versus zebrafish) and a less well conserved carboxy-terminal region.
(iii) At Least Three Hedgehog Homologues Are Present In The Chicken Genome Since two distinct PCR products encoding for chicken hedgehogs were amplified from genomic DNA, the total number of genes in the chicken hedgehog family needed to be 73 estimated. The two PCR clones pCHA (SEQ ID No:35) and pCHB (SEQ ID No:36) were used to probe a genomic Southern blot under moderately stringent conditions as described in the above Experimental Procedures. The blot was generated by digesting 5 pg of chick chromosomal DNA with EcoRI and BamHI alone and together. Each probe reacted most strongly with a distinct restriction fragment. For example, the blot probed with pCHA, shows three bands in each of the Barn HI lanes, one strong at 6.6 kb and two weak at 3.4 and 2.7 kb. The blot probed with pCHB, shows the 2.7 kb band as the most intense, while the 3.4 and 6.6 kb bands are weaker. A similar variation of intensities can also be seen in the Barn HI/Eco RI and EcoRI lanes. Exposure times were 72 hr. This data indicates that each probe recognizes a distinct chicken hedgehog gene, and that a third as yet uncharacterized chicken hedgehog homolog exists in the chicken genome.
(iv) Northern Analysis Defining Sites Of Sonic Transcription Northern analysis was performed which confirmed that Sonic is expressed during chick development. The spatial and temporal expression of Sonic in the chick embryo from gastrulation to early organogenesis was determined by whole mount in situ hybridization using a riboprobe corresponding to the full-length Sonic cDNA (SEQ ID No: 1).
total RNA isolated from stage 24 chick leg buds or bodies (without heads or limbs) was fractionated on a 0.8% agarose formaldehyde gel and transferred to a nylon membrane (Hybond N, Amersham). The blot was probed with the 1.6 kb EcoRI insert from pHH-2. Random-primed a3 2 P-labelled insert was hybridized at 42 0 C hybridization buffer BSA, 500mM NaHPO 4 7% SDS, 1 mM EDTA, pH 7.2) and washed at 630 C once in 0.5% bovine serum albumin, 50 mM NaHPO4 (pH 5% SDS, 1 mM EDTA and once in 40 mM NaHPO4 (pH 1% SDS, ImM EDTA. The image was visualized using a phosphoimager (Molecular Dynamics) and photographed directly from the video monitor.
25 Expression Of Sonic During Mid-Gastrulation Sonic message is detected in. the gastrulating blastoderm at early stage 4, the earliest stage analyzed. Staining is localized to the anterior end of the primitive streak in a region corresponding to Hensen's node. As gastrulation proceeds, the primitive streak elongates to its maximal cranial-caudal extent, after which Hensen's node regresses caudally and the 30 primitive streak shortens. At an early point of node regression, Sonic mRNA can be detected at the node and in midline cells anterior to the node. By late stage 5, when the node has migrated approximately one-third of the length of the fully elongated primitive streak, prominent Sonic expression is seen at the node and in the midline of the embryo, reaching its anterior limit at the developing head process. Sections at a cranial level show that Sonic mRNA is confined to invaginated axial mesendoderm, tissue which contributes to foregut and notochord. More caudally, but still anterior to Hensen's node, staining of axial 79 mesoderm is absent and Sonic expression is confined to the epiblast. At the node itself, high levels of Sonic message are observed in an asymmetric distribution extending to the left of and posterior to the primitive pit. This asymmetric distribution is consistently observed (6/6 embryos from stages 5-7) and is always located to the left of the primitive pit. At the node, and just posterior to the node, Sonic expression is restricted to the epiblast and is not observed in either mesoderm or endoderm. The expression of Sonic in the dorsal epiblast layer without expression in underlying axial mesoderm contrasts markedly with later stages where Sonic expression in underlying mesoderm always precedes midline neural tube expression.
(vi) Expression Of Sonic During Head Fold Stages During the formation and differentiation of the head process, Sonic mRNA is detected in midline cells of the neural tube, the foregut, and throughout most of the axial mesoderm.
At stage 7, Sonic message is readily detected asymmetrically at the node and in ventral midline cells anterior to the node. The rostral limit of Sonic expression extends to the anterior-most portions of the embryo where it is expressed in the foregut and prechordal mesoderm (Adelmann, (1932) Am. J. Anat. 31, 55-101). At stage 8, expression of Sonic persists along the entire ventral midline anterior to Hensen's node, while the node region itself no longer expresses Sonic. Transverse sections at different axial levels reveal that at stage 8 Sonic is coexpressed in the notochord and the overlying ventromedial neuroectoderm from anterior to Hensen's node to the posterior foregut. The levels of Sonic message are not uniform in the neural tube: highest levels are found at the presumptive mid- :and hindbrain regions with progressively lower levels anterior and posterior. The increasing I graded expression in the neural tube from Hensen's node to the rostral brain may reflect the developmental age of the neuroectoderm as differentiation proceeds from posterior to 25 anterior. At the anterior-most end of the embryo, expression is observed in midline cells of the dorsal and ventral foregut as well as in prechordal mesoderm. Although the prechordal mesoderm is in intimate contact with the overlying ectoderm, the latter is devoid of Sonic expression.
(vii) Expression Of Sonic During Early CNS Differentiation 30 At stages 10 through 14, Sonic expression is detected in the notochord, ventral neural **tube (including the floor plate), and gut precursors. By stage 10, there is a marked expansion of the cephalic neuroectoderm, giving rise to the fore- mid- and hind-brain. At stage Sonic mRNA is abundantly expressed in the ventral midline of the hindbrain and posterior midbrain. This expression expands laterally in the anterior midbrain and posterior forebrain.
Expression does not extend to the rostral forebrain at this or later stages. Sections reveal that Sonic is expressed in the notochord, the prechordal mesoderm, and the anterior midline of the foregut. Expression in the neuroepithelium extends from the forebrain caudally. In the posterior-most regions of the embryo which express Sonic, staining is found only in the notochord and not in the overlying neurectoderm. This contrasts with earlier expression in which the posterior domains of Sonic expression contain cells are located in the dorsal epiblast, but not in underlying mesoderm or endoderm. Midgut precursors at the level of the anterior intestinal portal also show weak Sonic expression.
At stage 14, expression continues in all three germ layers. The epithelium of the.
closing midgut expresses Sonic along with portions of the pharyngeal endoderm and anterior foregut. Ectoderm lateral and posterior to the tail bud also exhibits weak expression. At this stage, Sonic is also expressed along entire length of the notochord which now extends rostrally only to the midbrain region and no longer contacts the neuroepithelium at the anterior end of the embryo. Expression in head mesenchyme anterior to the notochord is no longer observed. In the neural tube Sonic is found along the ventral midline of the fore- midand hindbrain and posteriorly in the spinal cord In the forebrain, expression is expanded laterally relative to the hindbrain. At midgut levels, expression of Sonic in the neural tube appears:to extend beyond the floor plate into more lateral regions. As observed at stage Sonic at stage 14 is found in'the notochord, but ilot in the ventral neural tube in posteriormost regions of the embryo. When neuroectodermal expression is first observed in the posterior embryo, it is located in midline cells which appear to be in contact with the notochord. At later stages, expression continues in areas which show expression at stage 14, namely the CNS, gut epithelium including the allantoic stalk, and axial mesoderm.
(viii) Sonic Is Expressed In Posterior Limb Bud Mesenchyme The limb buds initially form as local thickenings of the lateral plate mesoderm. As distal outgrowth occurs during stage 17, Sonic expression becomes apparent in posterior S 25 regions of both the forelimb and the hindlimb. Sections through a stage 21 embryo at the level of the forelimbs reveal that expression of So;nic in limb buds is limited to mesenchymal tissue. A more detailed expression profile of Sonic during limb development is discussed below in Example 3. Briefly, as the limb bud grows out, expression of Sonic narrows along the anterior-posterior axis to become a thin stripe along the posterior margin closely apposed S 30 to the ectoderm. Expression is not found at more proximal regions of the bud. High levels of Sonic expression are maintained until around stage 25/26 when staining becomes weaker.
Expression of Sonic is no longer observed in wing buds or leg buds after stage 28.
7 6 Example 2 Mouse Sonic Hedgehog Is Implicated in the Regulation of CNS and Limb Polarity Experimental Procedures Isolation OfHedgehog Phage Clones The initial screen for mammalian hh genes was performed, as above, using a 700bp PCR fragment encompassing exons 1 and 2 of the Dros-HH gene. Approximately one million plaques of a 129/Sv Lambda Fix II genomic library (Stratagene) were hybridized with an ct 3 2 P-dATP labeled probe at low stringency 55°C in 6xSSC, 0.5%SDS, 5 x Denhardt's; final wash at 60°C in 0.5 x SSC, 0.1% SDS for Five cross hybridizing :phage plaques corresponding to the Dhh gene were purified. Restriction enzyme analysis indicated that all clones were overlapping. Selected restriction enzyme digests were then performed to map and subclone one of these. Subclones in pGEM (Promega) or Bluescript (Stratagene) which cross-hybridized with the Dros-HH fragment where sequenced using an ABI automatic DNA sequencer.
Mouse Ihh and Shh were identified by low stringency hybridization (as described above) with a chick Shh cDNA clone to one million plaques of an:8.5 day Xgtl0 mouse embryo cDNA library (Fahrner, K. et.al., (1987) EMBO J. 6: 1265-1271). Phage plaques containing a 1.8kb 1.h and 0.64 and 2.8kb Shh inserts were identified. Inserts were excised and subcloned into Bluescript (Stratagene) for dideoxy chain termination sequencing using modified T7 DNA polymerase (USB). The larger Shh clone contained a partially processed cDNA in which intron splicing at the exon 1/2 junction had not occurred.
To screen for additional Ihh and Shh cDNA clones, an 8.5 day XZAPII cDNA library was probed at high stringency (at 65 0 C in 6xSSC, 0.5% SDS, 5 x Denhardt's; final wash at C in 0.lxSSC, 0.1% SDS for 30') with the Ihh and Shh mouse cDNA clones. No additional 25 Ihh clones were identified. However several 2.6kb, apparently full length, Shh clones were isolated. The DNA sequence of the additional 5' coding region not present in the original 0.64 and 2.8kb Shh clones was obtained by analysis of one of the 2.6kb inserts.
Northern Blot Analysis Expression of Shh was investigated by RNA blot analysis using 20 pg of total RNA 30 from adult brain, spleen, kidney, liver, lung, 16.5dpc brain, liver and lung; 9.5dpc to 17.5dpc whole embryo; 9.5dpc forebrain, midbrain and 10.5dpc brain. RNA samples were electrophoretically separated on a 1.2% agarose gel, transferred and u.v. crosslinked to Genescreen (DuPont) and probed with 2X10 6 cpm/ml of an a 3 2 P-dATP labeled mouse Shh probe (2.8kb insert from Xgt 10 screen). Hybridization was performed at 42°C in formamide 5x Denhardt's, 5xSSPE, 0.1%SDS, 6.5% dextran, 200pg/ml salmon sperm DNA.
77 Final wash was at 55°C in O.1xSSC, 0.1%SDS. The blot was exposed for 6 days in the presence of an intensifying screen.
In Situ Hybridization, P-Galactosidase Staining And Histological Analysis Embryos from 7.25 to 14.5dpc were analyzed for either Shh or HNF-3P expression by whole mount in situ hybridization to digoxygenin labeled RNA probes as described in Wilkinson, (1992) In situ Hybridization: A Practical Approach. Oxford; Parr et al., (1993) Development 119:247-261. The mouse Shh probe was either a 2.8kb or 0.6kb RNA transcript generated by T7 (2.8kb) or T3 (0.6kb) transcription of XbaI and HindIII digests of Bluescript (Stratagene) subclones of the original Shh cDNA inserts. The HNF-33 probe was generated by HindIII linearization of a HNF-3p cDNA clone (Sasaki, H. et al., (1993) Development 118: 47-59) and T7 polymerase transcription of 1.6kb transcript. Embryos were photographed on an Olympus-SZH photomicroscope using Kodak Ektachrome EPY 64T color slide film.
Sections through wild type and WEXP2-CShh transgenic embryos were prepared and hybridized with 3 5 S-UIP labeled RNA probes (Wilkinson, D.G. et al., (1987) Development 99: 493-500). Sections were photographed as described in McMahon, A.P. et al., (1992) Cell 69: 581-595.
P Staining of WEXP2-lacZ embryos with pwas performed according to Whiting, J. et al., (1991) Genes Dev. 5: 2048-2059. General histological analysis of wildtype and 20 WEXP2-CShh transgenic embryos was performed on paraffin sections of Bouin's fixed embryos counterstained with hematoxylin and eosin. Histological procedures were as .described by Kaufman, M.H. (1992) The Atlas of Mouse Development, London: Academic Press. Sections were photographed on a Leitz Aristoplan compound microscope using S. Kodak EPY 64T color slide film.
DNA Constructs For Transgenics Genomic Wnt-l fragments were obtained by screening a XGEM12 (Promega) 129/Sv mouse genomic library with a 375 bp Mlul-BglII fragment derived from the fourth exon of the murine Wnt-l gene. One of the clones (W1-15.1) was used in this study.
As an initial step towards the generation of the pWEXP2 expression vector, W1-15.1 was digested to completion with restriction enzymes AatIl and ClaI, and a 2774 bp AatIl- ClaI fragment isolated. This fragment was ligated into AatII and ClaI cut pGEM-7Zf vector (Promega), generating pWl-18. This plasmid was digested with HindII and ligated to annealed oligonucleotides lad (SEQ ID No:21) and lac2 (SEQ ID No:22) generating pW1- 18S* which has a modified polylinker downstream of the ClaI restriction site. This construct (pWl-18S*) was digested with Clal and Bgll and ligated with both the 2.5 kb 3' Clal BglI 7 exon-intron region and 5.5 kb 3' BglII -BglII Wnt-1 enhancer, generating pWRES4. This construct contains a 10.5 kb genomic region which starts upstream of the Wnt-1 translation initiation codon (at an AatII site approximately 1.0kb from the ATG) and extends to a Bglll site 5.5 kb downstream of the Wnt-1 polyadenylation signal. This plasmid also contains a 250 bp region of the neomycin phosphotransferase (neo) gene inserted in inverse orientation in the 3' transcribed but untranslated region. Finally, to generate the WEXP2 expression vector, a 2 kb Sfi I fragment was amplified from pWRES4 using Sf-1 (SEQ ID No:23) and Sf-2 (SEQ ID No:24) oligonucleotides. This amplified fragment was digested with Sfi I and inserted into Sfi I linearized pWRES4, generating pWEXP2. This destroys the Wnt-1 translation initiation codon, and replaces it by a polylinker containing Nru I, Eco RV, Sac II, and Bst BI restriction sites, which are unique in pWEXP2.
The WEXP2 lacZ construct was obtained by inserting an end-filled Bgl II Xho I lacZ fragment isolated from the pSDKlacZpA vector in the Nru I cut pWEXP2 expression vector. Similarly, the WEXP2 CShh construct was obtained by inserting an end-filled XbaI cDNA fragment containing the full Chick Shh coding sequence (SEQ ID No: 1) into the Nru I cut WEXP2 expression vector.
Oligonucleotide sequences are as follows: lad: 5'-AGCTGTCGACGCGGCCGCTACGTAGGTTACCGACGTCAAGCTTAGATCTC-3' lac2: 5'-AGCTGAGATCTAAGCTTGACGTCGGTAACCTACGTAGCGGCCGCGTCGAC-3' Sf-1: 5'-GATCGGCCAGGCAGGCCTCGCGATATCGTCACCGCGGTATTCGAA-3' Sf-2: 5'-AGTGCCAGTCGGGGCCCCCAGGGCCGCGCC-3' Production And Genotyping Of Transgenic Embryos Transgenic mouse embryos were generated by microinjection of linear DNA fragments into the male pronucleus of B6CBAFl/J (C57BL/6J X CBA/J) zygotes. CD-1 or B6CBAFl/J females were used as recipients for injected embryos. Go mice embryos were collected at 9.5, 10.5, and 11.5 dpc, photographed using an Olympus SZH stereophotomicroscope on Kodak EPY-64T color slide film, then processed as described earlier.
WEXP2-lacZ and WEXP2-CShh transgenic embryos were identified by PCR analysis of proteinase-K digests of yolk sacs. Briefly, yolk sacs were carefully dissected free from 30 maternal and embryonic tissues, avoiding cross-contamination between littermates, then washed once in PBS. After overnight incubation at 55C in 50 pl of PCR proteinase-K S.:i digestion buffer (McMahon, A.P. et al., (1990) Cell 62: 1073-1085). 1 pl of heat-inactivated digest was subjected to polymerase chain reaction (PCR) in a 20 pl volume for 40 cycles as follows: 94*C for 30 seconds, 55C for 30 seconds, 72°C for 1 minute, with the reaction ingredients described previously (McMahon, A.P. et al., (1990) Cell 62: 1073-1085)). In the case of the WEXP2 lacZ transgenic embryos, oligonucleotides 137 (SEQ ID No:25) and 7q 138 (SEQ ID No:26) amplify a 352 bp lacZ specific product. In the case of the WEXP2- CShh embryos, oligonucleotides WPR2 (Wnt-1-specific) (SEQ ID No:27) and 924 (Chick Shh-specific) (SEQ ID No:28) amplify a 345 bp fragment spanning the insertion junction of the Chick-Shh cDNA in the WEXP2 expression vector. Table 2 summarizes the results of WEXP2-C-Shh transgenic studies.
Oligonucleotide sequences are as follows: 137: 5'-TACCACAGCGGATGGTTCGG-3' 138: 5' -GTGGTGGTTATGCCGATCGC-3' WPR2: 5'-TAAGAGGCCTATAAGAGGCGG-3' 924: 5'-AAGTCAGCCCAGAGGAGACT-3' (ii) Mouse hh Genes The combined screening of mouse genomic and 8.5 day post coitum (dpc) cDNA libraries identified three mammalian hh counterparts (Figure 5A) which herein will be referred to as Desert, Indian and Sonic hedgehog (Dhh, Ihh and Shh, respectively).
Sequences encoding Dhh (SEQ ID No:2) were determined from analysis of clones identified by low stringency screening of a mouse genomic library. DNA sequencing of one of five overlapping lambda phage clones identified three homologous regions encoding a single open reading frame interrupted by introns in identical position to those of the Dros-HH gene (Figure 5A). Splicing across the exon 1/2 boundary was confirmed by polymerase chain reaction (PCR) amplification of first strand cDNA generated from adult testicular RNA. The partial sequence of Ihh (SEQ ID No:3) and the complete sequence of Shh (SEQ ID No:4) coding regions were determined from the analysis of overlapping cDNA clones isolated from 8.5 dpc cDNA libraries. The longest Shh clone, 2.6kb, appears to be full length when compared with the Shh transcript present in embryonic RNAs. The 1.8kb partial length Ihh 25 cDNA is complete at the 3' end, as evidenced by the presence of a polyadenylation consensus sequence and short poly A tail.
Alignment of the predicted Dros-HH protein sequence (SEQ ID No:34) with those of the mouse Dhh (SEQ ID No:9), Ihh (SEQ ID No:10) and Shh (SEQ ID No: 11), and chick Shh (SEQ ID No:8) and zebrafish Shh (SEQ ID No:12), reveals several interesting features of the 30 :hh-family (Figure 5A). All the vertebrate hh-proteins contain an amino terminal hydrophobic region of approximately 20 amino acids immediately downstream of the initiation methionine. Although the properties of these new hh proteins have not been investigated, it is likely that this region constitutes a signal peptide and vertebrate hhs are secreted proteins.
Signal peptide cleavage is predicted to occur (von Heijne, (1986) Nucleic Acids Research 14: 4683-4690) just before an absolutely conserved six amino acid stretch, CGPGRG (SEQ ID No:29) (corresponding to residues 85-90)(Figure 5A), in all hh proteins. This generates 96 processed mouse Dhh (SEQ ID No:9) and Shh (SEQ ID No: 11) proteins of 41 and 44 kd, respectively. Interestingly, Dros-HH (SEQ ID No:34) is predicted to contain a substantial amino terminal extension beyond the hydrophobic domain suggesting that the Drosophila protein enters the secretory pathway by a type II secretory mechanism. This would generate a transmembrane tethered protein which would require subsequent cleavage to release a 43 kd secreted form of the protein. In vitro analysis of Dros-HH is consistent with this interpretation (Lee, J.J. et al., (1992) Cell 71: 33-50). However, there also appears to be transitional initiation at a second methionine (position 51 of SEQ ID No:34) just upstream of the hydrophobic region (Lee, J.J. et al., (1992) Cell 71: 33-50), suggesting that Dros-HH, like its vertebrate counterparts, may also be secreted by recognition of a conventional amino terminal signal peptide sequence.
Data base searches for protein sequences related to vertebrate hh's failed to identify any significant homologies, excepting Dros-HH. In addition, searching the "PROSITE" data bank of protein motifs did not reveal any peptide motifs which are conserved in the different hh proteins. Thus, the hhs represent a novel family of putative cell signaling molecules.
One feature of the amino acid alignment is the high conservation of hh sequences.
Vertebrate hhs share 47 to 51% amino acid identity with Dros-HH throughout the predicted processed polypeptide sequence (Figure Dhh has a slightly higher identity than that of Ihh and Shh suggesting that Dhh may be the orthologue of Dros-HH. Conservation is highest in the amino terminal half of the proteins, indeed, from position 85 (immediately after the predicted shared cleavage site) to 249, 62% of the amino acids are completely invariant amongst the Drosophila and vertebrate proteins. Comparison of mouse Dhh, Ihh and Shh where their sequences overlap in this more conserved region, indicates that Ihh and Shh are more closely related (90% amino acid identity; residues 85 to 266) than with the Dhh 25 sequence (80% amino acid identity; residues 85 to 266). Thus, Ihh and Shh presumably resulted from a more recent gene duplication event.
Comparison of cross species identity amongst Shh proteins reveals an even more striking sequence conservation. Throughout the entire predicted processed sequence mouse and chick Shh share 84% of amino acid residues (Figure However, in the amino terminal 30 half (positions 85 to 266) mouse and chick are 99% and mouse and zebrafish 94% identical in an 180 amino acid stretch. Conservation falls off rapidly after position 266 (Figure S. 0: SEQ ID No:40 shows the consensus sequence in the amino terminal half of all vertebrate Shh genes (human, mouse, chicken and zebrafish) identified to date. SEQ ID No:41 shows the consensus sequence in the amino terminal half of vertebrate hedgehog genes (Shh, Ihh, and Dhh) identified to date in different species (mouse, chicken, human and zebrafish).
In summary, hh family members are likely secreted proteins consisting of a highly conserved amino terminal and more divergent carboxyl terminal halves. The extreme interspecies conservation of the vertebrate Shh protein points to likely conservation of Shh function across vertebrate species.
(iii) Expression of Mouse Shh at the Axial Midline Expression of Shh in the mouse was examined in order to explore the role of mouse Shh (SEQ ID No: 11) in vertebrate development. Northern blots of embryonic and adult RNA samples were probed with a radiolabelled mouse Shh cDNA probe. An Shh transcript of approximately 2.6kb was detected in 9.5dpc whole embryo RNA, and 9.5 and 10.5dpc brain RNA fractions. No expression was detected in total RNA samples from later embryonic stages. Of the late fetal and adult tissue RNAs examined Shh expression was only detected in 16.5dpc and adult lung.
To better define the precise temporal and spatial expression of Shh an extensive series of whole mount and serial section in situ hybridizations were performed using digoxygenin and 3 5 S-radiolabelled RNA probes, respectively, to mouse embryo samples from 7.25dpc (mid streak egg cylinder stage of gastrulation) to 13.5dpc. No Shh expression is detected at mid-gastrulation stages (7.25dpc) prior to the appearance of the node, the mouse counterpart of the amphibian organizer and chick Hensen's node. When the primitive streak is fully extended and the midline mesoderm of the head process is emerging from the node (7.5 to 20 7.75dpc), Shh is expressed exclusively in the head process. At late head fold stages, Shh is expressed in the node and midline mesoderm of the head process extending anteriorly under the presumptive brain. Just prior to somite formation, Shh extends to the anterior limit of the midline mesoderm, underlying the presumptive midbrain. As somites are formed, the embryonic axis extends caudally. The notochord, which represents the caudal extension of the head process, also expresses Shh, and expression is maintained in the node.
Interestingly, by 8 somites (8.5dpc) strong Shh expression appears in the CNS.
Expression is initiated at the ventral midline of the midbrain, above the rostral limit of the head process. By 10 somites CNS expression in the midline extends rostrally in the forebrain and caudally into the hindbrain and rostral spinal cord. Expression is restricted in the hindbrain to the presumptive floorplate, whereas midbrain expression extends ventrolaterally. In the forebrain, there is no morphological floor plate, however ventral Shh expression here is continuous with the midbrain. By 15 somites ventral CNS expression is continuous from the rostral limit of the diencephalon to the presumptive spinal cord in somitic regions. Over the next 18 to 24 hrs, to the 25-29 somite stage, CNS expression intensifies and forebrain expression extends rostral to the optic stalks. In contrast to all other CNS regions, in the rostral half of the diencephalon, Shh is not expressed at the ventral 82 midline but in two strips immediately lateral to this area which merge again in the floor of the forebrain at its rostral limit. Expression of Shh in both the notochord and floorplate is retained until at least 13.5dpc.
Several groups have recently reported the cloning and expression of vertebrate members of a family of transcription factors, related to the Drosophilaforkhead gene. One of these, HNF-3 shows several similarities in expression to Shh (Sasaki, H. et al., (1993) Development 118: 47-59) suggesting that HNF-3p may be a potential regulator of Shh. To investigate this possibility, direct comparison of HNF-3p and Shh expression was undertaken. HNF-3p transcripts are first detected in the node (as previously reported by Sasaki, H. et al., (1993) supra), prior to the emergence of the head process and before Shh is expressed. From the node, expression proceeds anteriorly in the head process, similar to Shh expression. Activation of HNF-3p within the CNS is first observed at 2-3 somites, in the presumptive mid and hindbrain, prior to the onset of Shh expression. By 5 somites, expression in the midbrain broadens ventro-laterally, extends anteriorly into the forebrain and caudally in the presumptive floor plate down much of the neuraxis in the somitic region.
Strong expression is maintained at this time in the node and notochord. However, by somites expression in the head process is lost and by 25-29 somites notochordal expression is only present in the most extreme caudal notochord. In contrast to the transient expression of HNF-3p in the midline mesoderm, expression in the floor plate is stably retained until at least 11.5dpc. Thus, there are several spatial similarities between the expression of HNF-3p and Shh in both the midline mesoderm and ventral CNS and it is likely that both genes are expressed in the same cells. However, in both regions, HNF-3p expression precedes that of Shh. The main differences are in the transient expression of HNF-3p in the head process and notochord and Shh expression in the forebrain. Whereas HNF-3p and Shh share a similar 25 broad ventral and ventral lateral midbrain and caudal diencephalic expression, only Shh extends more rostrally into the forebrain. In general, these results are consistent with a model in which initial activation of Shh expression may be regulated by HNF-3p.
The similarity in Shh and HNF-30 expression domains is also apparent in the definitive endoderm which also lies at the midline. Broad HNF-3P expression in the foregut 30 pocket is apparent at 5 somites as previously reported by Sasaki, H. et al., (1993) supra. Shh is also expressed in the endoderm, immediately beneath the forebrain. Both genes are active in the rostral and caudal endoderm from 8 to 11 somites. Whereas HNF-3p is uniformly S". expressed, Shh expression is initially restricted to two ventro-lateral strips of cells. Ventral restricted expression of Shh is retained in the most caudal region of the presumptive gut until at least 9.5dpc whereas HNF-30 is uniformly expressed along the dorso-ventral axis. Both genes are expressed in the pharyngeal ectoderm at 9.5dpc and expression is maintained in the gut until at least 11.5dpc. Moreover, expression of Shh in the embryonic and adult lung RNA T3 suggests that endodermal expression of Shh may continue in, at least some endoderm derived organs.
(iv) Expression OfShh In The Limb Expression of Shh is not confined to midline structures. By 30-35 somites (9.75dpc), expression is detected in a small group of posterior cells in the forelimb bud. The forelimb buds form as mesenchymal outpocketings on the flanks, opposite somites 8 to 12, at approximately the 17 to 20 somite stage. Shh expression is not detectable in the forelimbs until about 30-35 somites, over 12 hours after the initial appearance of the limbs. Expression is exclusively posterior and restricted to mesenchymal cells. By 10.5dpc, both the fore and hindlimbs have elongated substantially from the body flank. At this time Shh is strongly expressed in the posterior, distal aspect of both limbs in close association with the overlying ectoderm. Analysis of sections at this stage detects Shh expression in an approximately six .cell wide strip of posterior mesenchymal cells. In the forelimb, Shh expression ceases by 11.5dpc. However, posterior, distal expression is still detected in the hindlimb. No limb expression is detected beyond 12.5dpc.
Ectopic Expression Of Shh Grafting studies carried out riacipally in the chick demonstrate that cell signals derived from the notochord and floor plate pattern the ventral aspect of the CNS (as described above). In the limb, a transient signal produced by a group of posterior cells in both limb S 20 buds, the zone of polarizing activity (ZPA), is thought to regulate patterning across the anterior-posterior axis. Thus, the sequence of Shh, which predicts a secreted protein and the S expression profile in midline mesoderm, the floor plate and in the limb, suggest that Shh signaling may mediate pattern regulation in the ventral CNS and limb.
To determine whether Shh may regulate ventral development in the early mammalian 25 CNS, a Wnt-l enhancer was used to alter its normal domain of expression. Wnt-l shows a dynamic pattern of expression which is initiated in the presumptive midbrain just prior to somite formation. As the neural folds elevate and fuse to enclose the neural tube, Wnt-1 expression in the midbrain becomes restricted to a tight circle, just anterior of the midbrain, the ventral midbrain and the dorsal midline of the diencephalon, midbrain, myelencephalon and spinal cord (Wilkinson, D.G. et al., (1987) Cell 50: 79-88; McMahon, A.P. et al., (1992) Cell 69: 581-595; Parr, B.A. et al., (1993) Development 119: 247-261).
It was determined that essentially normal expression of lacZ reporter constructs within the Wnt-l expression domain is dependent upon a 5.5kb enhancer region which lies downstream of the Wnt-l polyadenylation sequence. A construct was generated for ectopic expression of cDNA clones in the Wnt-l domain and tested in transgenics using a lacZ reporter (pWEXP-lacZ; Figure Two of the four G o transgenic embryos showed readily detectable P-galactosidase activity, and in both expression occurred throughout the normal Wnt-1 expression domain. More extensive studies with a similar construct also containing the enhancer gave similar frequencies. Some ectopic expression was seen in newly emerging neural crest cells, probably as a result of perdurance of P-galactosidase RNA or protein in the dorsally derived crest. Thus, the Wnt-l expression construct allows the efficient ectopic expression of cDNA sequences in the midbrain and in the dorsal aspect of much of the CNS.
An Shh ectopic expression construct (pWEXP-CShh) containing two tandem head to tail copies of a chick Shh cDNA was generated (Figure By utilizing this approach, ectopic expression of the chick Shh is distinguishable from that of the endogenous mouse Shh gene.
Chick Shh shows a high degree of sequence identity and similar expression to the mouse gene. Thus, it is highly likely that Shh function is widely conserved amongst vertebrates, a conclusion further supported by studies of the same gene in zebrafish.
Table 2 shows the results of several transgenic experiments in which the G o population was collected at 9.5 to 11.5dpc. Approximately half of the transgenic embryos identified at each stage of development had a clear, consistent CNS phenotype. As we expect, on the basis of control studies using the 5.5kb Wnt-l enhancer, that only half the transgenics will express the transgene, it is clear that in most embryos ectopically expressing chick Shh, an abnormal phenotype results.
TABLE 2 Summary of WEXP2-Chick Shh transgenic studies A
N
Age (dpc) Number of Number of Number of Embryos with Embryos Transgenics CNS phenotypea 9.5 37 11 6 (54.5%) 10.5 59 16 8(50%) 11.5 33 7 3 (42.9%) 25 Figures in parentheses, refer to the percentage of transgenic embryos with a CNS phenotype a In addition one 9.5pc and two 10.5pc transgenic embryos showed non-specific growth retardation, as occurs at low frequency in transgenic studies. These embryos were excluded from further analysis.
At 9.5dpc, embryos with a weaker phenotype show an open neural plate from the mid diencephalon to the myelencephalon. In embryos with a stronger phenotype at the same stage, the entire diencephalon is open and telencephalic and optic development is morphologically abnormal. As the most anterior diencephalic expression of Wnt-l is lower than that in more caudal regions, the differences in severity may relate to differences in the level of chick Shh expression in different Go embryos. At the lateral margins of the open neural folds, where Wnt-l is normally expressed, there is a thickening of the neural tissue extending from the diencephalon to myelencephalon. The cranial phenotype is similar at 10.5 and 11.5 dpc. However, there appears to be a retardation in cranial expansion of the CNS at later stages.
In addition to the dorsal cranial phenotype, there is a progressive dorsal phenotype in the spinal cord. At 9.5 dpc, the spinal cord appears morphologically normal, except at extreme rostral levels. However by 10.5dpc, there is a dorsal dysmorphology extending to the fore or hindlimbs. By 11.5 dpc, all transgenic embryos showed a dorsal phenotype along almost the entire spinal cord. Superficially, the spinal cord had a rippled, undulating appearance suggestive of a change in cell properties dorsally. This dorsal phenotype, and the cranial phenotype were examined by histological analysis of transgenic embryos.
Sections through a 9.5dpc embryo with an extreme CNS phenotype show a widespread dorsal perturbation in cranial CNS development. The neural/ectodermal junction in the diencephalon is abnormal. Neural tissue, which has a columnar epithelial morphology quite distinct from the squamous epithelium of the surface ectoderm, appears to spread dorsolaterally. The myelencephalon, like the diencephalon and midbrain, is open rostrally.
Interestingly, there are discontinuous dorso-lateral regions in the myelencephalon with a morphology distinct from the normal roof plate regions close to the normal site of Wnt-l expression. These cells form a tight, polarized epithelium with basely located nuclei, a 20 morphology similar to the floor plate and distinct from other CNS regions. Differentiation of dorsally derived neural crest occurs in transgenic embryos as can be seen from the presence of cranial ganglia. In the rostral spinal cord, the neural tube appeared distended dorsolaterally which may account for the superficial dysmorphology.
By 11.5dpc, CNS development is highly abnormal along the entire dorsal spinal cord to the hindlimb level. The dorsal half of the spinal cord is enlarged and distended. Dorsal sensory innervation occurs, however, the neuronal trajectories are highly disorganized. Most obviously, the morphology of dorsal cells in the spinal cord, which normally are elongated cells with distinct lightly staining nuclei and cytoplasm, is dramatically altered. Most of the dorsal half of the spinal cord consists of small tightly packed cells with darkly staining nuclei 30 and little cytoplasm. Moreover, there appears to be many more of these densely packed cells, leading to abnormal outgrowth of the dorsal CNS. In contrast, ventral development is normal, as are dorsal root ganglia, whose origins lie in neural cells derived from the dorsal spinal cord.
(vi) Ectopic Shh Expression Activates Floor Plate Gene Expression To determine whether ectopic expression of chick Shh results in inappropriate activation of a ventral midline development in the dorsal CNS, expression of two floor plate expressed genes, HNF-3P and mouse Shh, were examined. Whole mounts of transgenic embryos show ectopic expression of HNF-3p throughout the cranial Wnt-l expression domain. In addition to normal expression at the ventral midline, transcripts are expressed at high levels, in a circle just rostral to the mid/hindbrain junction, along the dorsal (actually lateral in unfused brain folds) aspects of the midbrain and, more weakly, in the roof plate of the myelencephalon. No expression is observed in the metencephalon which does not express Wnt-l. Thus, ectopic expression of Shh leads to the activation of HNF-3p throughout the cranial Wnt-l expression domain.
The relationship between chick Shh expression and the expression of HNF-31 in serial sections was also examined. Activation of HNF-3p in the brain at 9.5 and 10.5dpc is localized to the dorsal aspect in good agreement with the observed ectopic expression of chick Shh. Interestingly mouse Shh is also activated dorsally. Thus, two early floor plate markers are induced in response to chick Shh.
From 9.5dpc to 11.5dpc, the spinal cord phenotype becomes more severe. The possibility that activation of a floor plate pathway may play a role in the observed phenotype was investigated. In contrast to the brain, where ectopic HNF-30 and Shh transcripts are still present, little or no induction of these floor plate markers is observed. Thus, although the dorsal spinal cord shows a widespread transformation in cellular phenotype, this does not appear to result from the induction of floor plate development.
Example 3 Chick Sonic Hedgehog Mediates ZPA Activity Experimental Procedures Retinoic Acid Bead Implants 25 Fertilized white Leghorn chicken eggs were incubated to stage 20 and then implanted with AG1-X2 ion exchange beads (Biorad) soaked in 1 mg/ml retinoic acid (RA, Sigma) as described by Tickle, C. et al., (1985) Dev. Biol 109: 82-95. Briefly, the beads were soaked for 15 min in 1mg/ml RA in DMSO, washed twice and implanted under the AER on the anterior margin of the limb bud. After 24 or 36 hours, some of the implanted embryos were harvested and fixed overnight in 4% paraformaldehyde in PBS and then processed for whole mount in situ analysis as previously described. The remainder of the animals were allowed to develop to embryonic day 10 to confirm that the dose of RA used was capable of inducing mirror image duplications. Control animals were implanted with DMSO soaked beads and showed no abnormal phenotype or gene expression.
Plasmids Unless otherwise noted, all standard cloning techniques were performed according to Ausubel, F.M. et al., (1989) Current Protocols in Molecular Biology Greene Publishing Assoc. and Wiley Inerscience), and all enzymes were obtained from Boehringer Mannheim Biochemicals. pHH-2 is a cDNA contain the entire coding region of chicken Sonic hedgehog (SEQ ID No:l). RCASBP(A) and RCASBP(E) are replication-competent retroviral vectors which encode viruses with differing host ranges. RCANBP(A) is a variant of RCASBP(A) from which the second splice acceptor has been removed. This results in a virus which can not express the inserted gene and acts as a control for the effects of viral infection (Hughes, S.H. et al., (1987) J Virol. 61: 3004-3012; Fekete, D. et al., (1993) Mol.
Cell. Biol. 13: 2604-2613). RCASBP/AP(E) is version of RCASBP(E) containing a human placental alkaline phosphatase cDNA (Fekete, D. et al., (1993b) Proc. Natl. Acad Sci. USA 2350-2354). SLAX13 is a pBluescript SK+ derived plasmid with a second Cla I restriction site and the 5' untranslated region of v-src (from the adaptor plasmid CLA 12-Nco, Hughes, S.H. et al., (1987) J. Virol. 61: 3004-3012) cloned 5' of the EcoRI (and Clal) site in the pBluescript polylinker. RCASBP plasmids encoding Sonic from either the first (M or second (M2) methionine (at position 4) were constructed by first shuttling the 1.7kb Sonic fragment of pHH-2 into SLAX-13 using oligonucleotides to modify the 5' end of the cDNA such that either the first or second methionine is in frame with the NcoI site of SLAX-13.
The amino acid sequence of Sonic is not mutated in these constructs. The Ml and M2 Sonic Cla l fragments (v-src 5'UTR:Sonic) were each then subcloned into RCASBP(A), i RCANBP(A) and RCASBP(E), generating Sonic/RCAS-Al, Sonic/RCAS-A2, Sonic/RCAN-Al, Sonic/RCAN-A2, Sonic/RCAS-El and Sonic/RCAS-E2.
Chick Embryos, Cell Lines And Virus Production All experimental manipulations were performed on standard specific-pathogen free White Leghorn chick embryos (S-SPF) from closed flocks provided fertilized by SPAFAS (Norwich, Conn). Eggs were incubated at 37.5 0 C and staged according to Hamburger, V. et al., (1951) J. Exp. Morph. 88: 49-92. All chick embryo fibroblasts (CEF) were provided by i C. Cepko. S-SPF embryos and CEFs have previously been shown to be susceptible to 30 RCASBP(A) infection but resistant to RCASBP(E) infection (Fekete, D. et al., (1993b) Proc.
Natl. Acad Sci. USA 90: 2350-2354). Line 15b CEFs are susceptible to infection by both S.yi RCASBP(A) and These viral host ranges were confirmed in control experiments. CEF cultures were grown and transfected with retroviral vector DNA as described (Morgan, B.A.
et al., (1993) Nature 358: 236-239; Fekete, D. et al., (1993b) Proc. Natl. Acad. Sci. USA 2350-2354). All viruses were harvested and concentrated as previously described (Morgan, B.A. et al., (1993) Nature 358: 236-239; Fekete, D. et al., (1993b) Proc. Natl. Acad. Sci. USA 2350-2354) and had titers of approximately 108 cfu/ml.
Cell Implants A single 60mm dish containing line 15b CEFs which had been infected with either RCASBP/AP(E), Sonic/RCAS-El or Sonic/RCAS-E2 were grown to 50-90% confluence, lightly trypsinized and then spun at 1000 rpm for 5 min in a clinical centrifuge. The pellet was resuspended in 1 ml media, transferred to a microcentrifuge tube and then microcentrifuged for 2 min at 2000 rpm. Following a 30 min incubation at 370 C, the pellet was respun for 2 min at 2000 rpm and then lightly stained in media containing 0.01% nile blue sulfate. Pellet fragments of approximately 300um x 100pm x 50.tm were implanted as a wedge to the anterior region of hh stage 19-23 wing buds (as described by Riley, B.B. et al., (1993) Development 118: 95-104). At embryonic day 10, the embryos were harvested, fixed in 4% paraformaldehyde in PBS, stained with alcian green, and cleared in methyl salicylate (Tickle, C. et al., (1985) Dev. Biol 109: 82-95).
Viral Infections Concentrated Sonic/RCAS-A2 or Sonic/RCAN-A2 was injected under the AER on the anterior margin of stage 20-22 wing buds. At 24 or 36 hours post-infection, the embryos were harvested, fixed in 4% paraformaldehyde in PBS and processed for whole mount in situ analysis as previously described.
(ii) Co-Localization Of Sonic Expression And Zpa Activity ZPA activity has been carefully mapped both spatiaiiy and temporally within the limb 20 bud (Honig, L.S. et al., (1985) J. Embryol. exp. Morph. 87: 163-174). In these experiments small blocks of limb bud tissue from various locations and stages of chick embryogenesis (Hamburger, V et al., (1951) J. Exp. Morph. 88: 49-92) were grafted to the anterior of host limb buds and the strength of ZPA activity was quantified according to degrees of digit duplication. Activity is first weakly detected along the flank prior to limb bud outgrowth.
25 The activity first reaches a maximal strength at stage 19 in the proximal posterior margin of the limb bud. By stage 23 the activity extends the full length of the posterior border of the limb bud. The activity then shifts distally along the posterior margin so that by stage 25 it is no longer detectable at the base of the flank. The activity then fades distally until it is last detected at stage 29.
This detailed map of endogenous polarizing activity provided the opportunity to determine the extent of the correlation between the spatial pattern of ZPA activity and Sonic expression over a range of developmental stages. Whole mount in situ hybridization was used to assay the spatial and temporal pattern of Sonic expression in the limb bud. Sonic expression is not detected until stage 17, at the initiation of limb bud formation, at which time it is weakly observed in a punctate pattern reflecting a patchy expression in a few cells.
From that point onwards the Sonic expression pattern exactly matches the location of the ZPA, as determined by Honig, L.S. et al., (1985) J. Embryol. exp. Morph. 87: 163-174, both in position and in intensity of expression.
(iii) Induction Of Sonic Expression By Retinoic Acid A source of retinoic acid placed at the anterior margin of the limb bud will induce ectopic tissue capable causing mirror-image duplications (Summerbell, D. et al., (1983) In Limb Development and Regeneration Ala R. Liss) pp. 109-118; Wanek, N. et al., (1991) Nature 350: 81-83). The induction of this activity is not an immediate response to retinoic acid but rather takes approximately 18 hours to develop (Wanek, N. et al., (1991) Nature 350: 81-83). When it does develop, the polarizing activity is not found surrounding the implanted retinoic acid source, but rather is found distal to it in the mesenchyme along the margin of the limb bud (Wanek, N. et al., (1991) Nature 350: 81-83).
If Sonic expression is truly indicative of ZPA tissue, then it should be induced in the ZPA tissue which is ectopically induced by retinoic acid. To test this, retinoic acid-soaked beads were implanted in the anterior of limb buds and the expression of Sonic after various lengths of time using whole-mount in situ hybridization was assayed. As the limb bud grows, the bead remains imbedded proximally in tissue which begins to differentiate.
Ectopic Sonic expression is first detected in the mesenchyme 24 hours after bead implantation. This expression is found a short distance from the distal edge of the bead. By 20 36 hours Sonic is strongly expressed distal to the bead in a stripe just under the anterior ectoderm in a mirror-image pattern relative to the endogenous Sonic expression in the posterior of the limb bud.
Effects Of Ectopic Expression Of Sonic On Limb Patterning The normal expression pattern of Sonic, as well as that induced by retinoic acid, is consistent with Sonic being a signal produced by the ZPA. To determine whether Sonic expression is sufficient for ZPA activity, the gene was ectopically expressed within the limb bud. In most of the experiments we have utilized a variant of a replication-competent retroviral vector called RCAS (Hughes, S.H. et al., (1987) J Virol. 61: 3004-3012)) both as a vehicle to introduce the Sonic sequences into chick cells and to drive their expression. The 30 fact that there exists subtypes of avian retroviruses which have host ranges restricted to particular strains of chickens was taken advantage of to control the region infected with the Sonic/RCAS virus (Weiss, R. (et al.) (1984) RNA Tumor Viruses, Vol. 1 Weiss et al. eds., Cold Spring Harbor Laboratories) pp. 209-260); Fekete, D. et al., (1993a) Mol. Cell.
Biol. 13: 2604-2613). Thus a vector with a type E envelope protein (RCAS-E, Fekete, D. et al., (1993b) Proc. Natl. Acad Sci. USA 90: 2350-2354) is unable to infect the cells of the SPAFAS outbred chick embryos routinely used in our lab. However, RCAS-E is able to 9b infect cells from chick embryos of line 15b. In the majority of experiments, primary chick embryo fibroblasts (CEFs) prepared from line 15b embryos in vitro were infected. The infected cells were pelleted and implanted into a slit made in the anterior of S-SPF host limb buds. Due to the restricted host range of the vector, the infection was thus restricted to the graft and did not spread through the host limb bud.
To determine the fate of cells implanted and to control for any effect of the implant procedure, a control RCAS-E vector expressing human placental alkaline phosphatase was used. Alkaline phosphatase expression can be easily monitored histochemically and the location of infected cells can thus be conveniently followed at any stage. Within 24 hours following implantation the cells are dispersed proximally and distally within the anterior margin of the limb bud. Subsequently, cells are seen to disperse throughout the anterior portion of the limb and into the flank of the embryo.
Limb buds grafted with alkaline phosphatase expressing cells or uninfected cells give rise to limbs with structures indistinguishable from unoperated wild type limbs. Such limbs have the characteristic anterior-to-posterior digit pattern 2-3-4. ZPA grafts give rise to a variety of patterns of digits depending on the placement of the graft within the bud (Tickle, C. et al., (1975) Nature 254: 199-202) and the amount of tissue engrafted (Tickle, C. (1981) Nature 289: 295-298). In some instances the result can be as weak as the duplication of a single digit 2. However, in optima! cases the ZPA graft evokes the production of a full mirror image duplication of digits 4-3-2-2-3-4 or 4-3-2-3-4 (see Figure A scoring system has been devised which rates the effectiveness of polarizing activity on the basis of the most "i posterior digit duplicated: any graft which leads to the development of a duplication of digit 4 has been defined as reflecting 100% polarizing activity (Honig, L.S. et al., (1985) J. Embryol.
Exp. Morph. 87: 163-174).
Grafts of 15b fibroblasts expressing Sonic resulted in a range ofZPA-like phenotypes.
In some instances the resultant limbs deviate from the wild type solely by the presence of a mirror-image duplication of digit The most common digit phenotype resulting from grafting Sonic-infected CEF cells is a mirror-image duplication of digits 4 and 3 with digit 2 missing: 4-3-3-4. In many such cases the two central digits appear fused in a 4-3/3-4 pattern.
30 In a number of the cases the grafts induced full mirror-image duplications of the digits S"equivalent to optimal ZPA grafts 4-3-2-2-3-4. Besides the digit duplications, the ectopic expression of Sonic also gave rise to occasional duplications of proximal elements including the radius or ulna, the humerus and the coracoid. While these proximal phenotypes are not features of ZPA grafts, they are consistent with an anterior-to-posterior respecification of cell fate. In some instances, most commonly when the radius or ulna was duplicated, more complex digit patterns were observed. Typically, an additional digit 3 was formed distal to a duplicated radius.
The mirror-image duplications caused by ZPA grafts are not limited to skeletal elements. For example, feather buds are normally present only along the posterior edge of the limb. Limbs exhibiting mirror-image duplications as a result of ectopic Sonic expression have feather buds on both their anterior and posterior edges, similar to those observed in ZPA grafts.
While ZPA grafts have a powerful ability to alter limb pattern when placed at the anterior margin of a limb bud, they have no effect when placed at the posterior margin (Saunders, J.W. et al., (1968) Epithelial-Mesenchymal Interaction, Fleischmayer and Billingham, eds. (Baltimore: Williams and Wilkins) pp. 78-97). Presumably, the lack of posterior effect is a result of polarizing activity already being present in that region of the bud. Consistent with this, grafts of Sonic expressing cells placed in the posterior of limb buds never result in changes in the number of digits. Some such grafts did produce distortions in the shape of limb elements, the most common being a slight posterior curvature in the distal tips of digits 3 and 4 when compared to wild type wings.
Effect Of Ectopic Sonic Expression On Hoxd Gene Activity The correct expression of Hoxd genes is part of the process by which specific skeletal elements are determined (Morgan, B.A. et al., (1993) Nature 358: 236-239). A transplant of a ZPA into the anterior of a chick limb bud ectopically activates sequential transcription of Hoxd genes in a pattern which mirrors the normal sequence of Hoxd gene expression (Nohno, S 20 T. et-al., (1991) Cell 64: 1197-1205; Izpisua-Belmonte, J.C. et al., (1991) Nature 350: 585- 589). Since ectopic Sonic expression leads to the same pattern duplications as a ZPA graft, we reasoned that Sonic would also lead to sequential activation of Hoxd genes.
To test this hypothesis, anterior buds were injected with Sonic/RCAS-A2, a virus 0. which is capable of directly infecting the host strains of chicken embryos. This approach does not strictly limit the region expressing Sonic (being only moderately controlled by the timing, location and titer of viral injection), and thus might be expected to give a more variable result. However, experiments testing the kinetics of viral spread in infected limb buds indicate that infected cells remain localized near the anterior margin of the bud for at least 48 hours. Hoxd gene expression was monitored at various times post infection by 30 whole mount in situ hybridization. As expected, these genes are activated in a mirror-image pattern relative their expression in the posterior of control limbs. For example, after 36 hours Hoxd-13 is expressed in a mirror-image symmetrical pattern in the broadened distal region of Sinfected limb buds. Similar results were obtained with other Hoxd genes (manuscript in preparation).
92 Examnle 4 A Functionally Conserved Homolog ofDrosophila Hedgehog is Expressed in Tissues With Polarizing Activity in Zebrafish Embryos Experimental Procedures Cloning and Sequencing Approximately 1.5 x 106 plaques of a 33h zebrafish embryonic Xgt 1 cDNA library were screened by plaque hybridization at low stringency (McGinnis, W. et al., (1984) Nature 308: 428-433) using a mix of two hh sequences as a probe: a Drosophila hh 400bp EcoRI fragment and a murine Ihh 264bp BamHI-EcoRI exon 2 fragment. Four clones were isolated and subcloned into the EcoRI sites of pUC18 T3T7 (Pharmacia). Both strands of clone 8.3 were sequenced using nested deletions (Pharmacia) and internal oligonucleotide primers.
DNA sequences and derived amino acid sequences were analyzed using "Geneworks" (Intelligenetics) and the GCG software packages.
PCR amplification Degenerate oligonucleotides hh5.1 (SEQ ID No:30) and hh3.3 (SEQ ID No:31) were used to amplify genomic zebrafish DNA hh 5.1: AG(CA)GITG(CT)AA(AG)GA(AG)(CA)(AG)I(GCT)IAA hh 3.3: CTCIACIGCIA(GA)ICK(GT)IGCIA PCR was performed with an initial denaturation at 94C followed by 35 cycles of 47C for 1 20 min, 72C for 2min and 94 0 C for 1 min with a final extension at 72 0 C. Products were subcloned in pUC18 (Pharmacia).
*.oo In Situ Hybridization In situ hybridizations of zebrafish embryos were performed as described in Oxtoby, E. et al., (1993) Nuc. Acids REs. 21: 1087-1095 with the following modifications: Embryos 25 were rebydrated in ethanol rather than methanol series; the proteinase K digestion was reduced to 5 min and subsequent washes were done in PBTw without glycine; the antibody was preadsorbed in PBTw, 2mg/ml BSA without sheep serum; and antibody incubation was performed in PBTw, 2mg/ml BSA. Drosophila embryos were processed and hybridized as previously described.
30 Histology Stained embryos were dehydrated through ethanol:butanol series, as previously described (Godsave, S.F. et al., (1988) Development 102: 555-566), and embedded in Fibrowax. 8pm sections were cut on an Anglian rotary microtome RNA Probe Synthesis For analysis of Shh expression, two different templates were used with consistent results; phh[c] 8.3 linearized with Bgl II to transcribe an antisense RNA probe that excludes the conserved region, and (ii) phh[c] 8.3 linearized with Hind III to transcribe an antisense RNA that covers the complete cDNA. All in situ hybridizations were performed with the latter probe which gives better signal. Other probes were as follows: Axial Drallinearized p6TIN (StrAhle, U. et al., (1993) Genes Dev. 7: 1436-1446) using T3 RNA polymerase. gsc linearized with EcoRl and transcribed with T7: pax 2 Bar HI-linearized pcFl6 (Krauss, S. et al., (1991) Development 113: 1193-1206) using T7 RNA polymerase.
In situ hybridizations were performed using labelled RNA at a concentration of 1 ng/ml final concentration. Antisense RNA probes were transcribed according to the manufacturer's protocol (DIG RNA Labelling Kit, BCL).
Zebrafish Strains Wild type fish were bred from a founder population obtained from the Goldfish Bowl, Oxford. The mutant cyclops strain bl6 and the mutant notail strains bl60 and b195 were obtained from Eugene, Oregon. Fish were reared at 28 0 C on a 14h light/I Oh dark cycle.
RNA Injections The open reading frame of Shh was amplified by PCR, using oligonucleotides 5'-CTGCAGGGATCCACCATGCGGCTTTTGACGAG-3' (SEQ ID No:32), which contains S 20 a consensus Kozak sequence for translation initiation, and CTTATTCCACACGAGGGATT-3' (SEQ ID No:33), and subcloned into the BglII site of SpSP64T (Kreig, P.A. et al., (1984) Nuc.Acids Res. 12: 7057-7070). This vector includes and 3' untranslated Xenopus f3-Globin sequences for RNA stabilization and is commonly used for RNA injections experiments in Xenopus. In vitro transcribed Shh RNA at a concentration of approximately 100 pg/ml was injected into a single cell of naturally ospawned zebrafish embryos at one-cell to 4-cell stages using a pressure-pulsed Narishige microinjector. The injected volume was within the picolitre range. Embryos were fixed to 27 hrs after injection in BT-Fix (Westerfield, M. (1989) The Zebrafish Book, (Eugene: The University of Oregon Press)) and processed as described above for whole-mount in situ S 30 hybridizations with the axial probe.
Transgenic Drosophila An EcoR1 fragment, containing the entire Shh ORF, was purified from the plasmid phh[c]8.3 and ligated with phosphatased EcoR1 digested transformation vector pCaSpeRhs (Thummel, C.S. et al., (1988) Gene 74: 445-456). The recombinant plasmid, pHS Shh containing the Shh ORF in the correct orientation relative to the heat shock promoter, was selected following restriction enzyme analysis of miniprep DNA from transformed colonies and used to transform Drosophila embryos using standard microinjection procedures (Roberts, D.B. (1986), Drosophila, A Practical Approach, Roberts, ed., (Oxford: IRL Press) pp. 1-38).
Ectopic Expression In Drosophila Embryos Embryos carrying the appropriate transgenes were collected over 2 hr intervals, transferred to thin layers of 1% agarose on glass microscope slides and incubated in a plastic Petri dish floating in a water bath at 37 0 C for 30 min intervals. Following heat treatment, embryos were returned to 25C prior to being fixed for in situ hybridization with DIG labelled single stranded Shh, wg or ptc RNA probes as previously described (Ingham et al., (1991) Curr. Opin. Genet. Dev. 1: 261-267).
(ii) Molecular Cloning OfZebrafish Hedgehog Homologues In an initial attempt to isolate sequences homologous to Drosophila hh, a zebrafish genomic DNA library was screened at reduced stringency with a partial cDNA, hhPCR4.1, corresponding to the first and second exons of the Drosophila gene (Mohler, J. et al., (1992) Development 115: 957-971). This screen proved unsuccessful; however, a similar screen of a mouse genomic library yielded a single clone with significant homology to hh., subsequently designated Ihh. A 264bp BamHI-EcoRl fragment from this lambda clone containing sequences homologous to the second exon of the Drosophila gene was subcloned and, 20 together with the Drosophila partial cDNA fragment, used to screen a Xgtl 1 zebrafish cDNA library that was prepared from RNA extracted from 33h old embryos. This screen yielded four clones with overlapping inserts the longest of which is 1.6kb in length, herein referred to as Shh (SEQ ID (iii) A Family Of Zebrafish Genes Homologous To The Drosophila Segment Polarity Gene, Hedgehog Alignment of the predicted amino acid sequences of Shh (SEQ ID No:12) and hh (SEQ ID No:34) revealed an identity of 47%, confirming that Shh is a homolog of the Drosophila gene. A striking conservation occurs within exon 2: an 80 amino acid long domain shows 72% identity between Shh and Dros-HH. (Figure 9A). This domain is also highly conserved in all hh-related genes cloned so far and is therefore likely to be essential to the function of hh proteins. A second domain of approximately 30 amino acids close to the carboxy-terminal end, though it shows only 61% amino-acid identity, possesses 83% similarity between Shh and hh when allowing for conservative substitutions and could also, therefore, be of functional importance (Figure 9B). Although putative sites of posttranslational modification can be noted, their position is not conserved between Shh and hh.
Lee, J.J. et al., (1992) Cell 71: 33-50, identified a hydrophobic stretch of 21 amino acids flanked downstream by a putative site of signal sequence cleavage (predicted by the algorithm of von Heijne, G. (1986) Nuc. Acids Res. 11) close to the amino-terminal end of hh. Both the hydrophobic stretch and the putative signal sequence cleavage sites of hh, which suggest it to be a signaling molecule, are conserved in Shh. In contrast to hh, Shh does not extend N-terminally to the hydrophobic stretch.
Using degenerate oligonucleotides corresponding to amino-acids flanking the domain of high homology between Dros-HH and mouse Ihh exons 2 described above, fragments of the expected size were amplified from zebrafish genomic DNA by PCR. After subcloning and sequencing, it appeared that three different sequences were amplified, all of which show high homology to one another and to Dros-HH (Figure 10). One of these corresponds to Shh therein referred to as 2-hh(a) (SEQ ID No:16) and 2hh(b) (SEQ ID No:17), while the other two represent additional zebrafish hh homologs (SEQ ID No:5). cDNAs corresponding to one of these additional homologs have recently been isolated, confirming that it is transcribed. Therefore, Shh represents a member of a new vertebrate gene family.
(iv) Shh Expression In The Developing Zebrafish Embryo Gastrula stages Shh expression is first detected at around the 6 0% epiboly stage of embryogenesis in the dorsal mesoderm. Transcript is present in a triangular shaped area, corresponding to the 20 embryonic shield, the equivalent of the amphibian organizer, and is restricted to the inner cell layer, the hypoblast. During gastrulation, presumptive mesodermal cells involute to form the hypoblast, and converge towards the future axis of the embryo, reaching the animal pole at approximately 70% epiboly. At this stage, Shh -expressing cells extend over the posterior third of the axis, and the signal intensity is not entirely homogeneous, appearing stronger at the base than at the apex of the elongating triangle of cells.
S"This early spatial distribution of Shh transcript is reminiscent of that previously described for axial, a forkhead-related gene; however, at 80% epiboly, axial expression extends further towards the animal pole of the embryo and we do not see Shh expression in S•the head area at these early developmental stages.
By 100% epiboly, at 9.5 hours of development, the posterior tip of the Shh expression domain now constitutes a continuous band of cells that extends into the head. To determine the precise anterior boundary of Shh expression, embryos were simultaneously hybridized with probes of Shh and pax-2 (previously pax[b]), the early expression domain of which marks the posterior midbrain (Krauss, S. et al. (1991) Development 113: 1193-1206). By this stage, the anterior boundary of the Shh expression domain is positioned in the centre of the 96 animal pole and coincides approximately with that of axial. At the same stage, prechordal plate cells expressing the homeobox gene goosecoid (gsc) overlap and underlay the presumptive forebrain (Statchel, S.E. et al., (1993) Development 117: 1261-1274). Whereas axial is also thought to be expressed in head mesodermal tissue at this stage, we cannot be certain whether Shh is expressed in the same cells. Sections of stained embryos suggest that in the head Shh may by this stage be expressed exclusively in neuroectodermal tissue.
Somitogenesis By the onset of somitogenesis (approximately 10.5h of development), Shh expression in the head is clearly restricted to the ventral floor of the brain, extending from the tip of the diencephalon caudally through the hindbrain. At this stage, expression of axial has also disappeared from the head mesoderm and is similarly restricted to the floor of the brain; in contrast to Shh, however, it extends only as far as the anterior boundary of the midbrain. At this point, gsc expression has become very weak and is restricted to a ring of cells that appear to be migrating away from the dorsal midline.
As somitogenesis continues, Shh expression extends in a rostral-caudal progression throughout the ventral region of the central nervous system (CNS). Along the spinal cord, the expression domain is restricted to a single row of cells, the floor plate, but gradually broadens in the hindbrain and midbrain to become 5-7 cells in diameter, with a triangular shaped lateral extension in the ventral diencephalon and two strongly staining bulges at the tip of the forebrain, presumably in a region fated to become hypothalamus.
As induction of Shh in the floor plate occurs, expression in the underlying mesoderm begins to fade away, in a similar manner to axial (Strahle, U. et al., (1993) Genes Dev. 7: 1436-1446). This downregulation also proceeds in a rostral to caudal sequence, coinciding with the changes in cell shape that accompany notochord differentiation. By the 22 somite stage, mesodermal Shh expression is restricted to the caudal region of the notochord and in the expanding tail bud where a bulge of undifferentiated cells continue to express Shh at relatively high levels. Expression in the midbrain broadens to a rhombic shaped area; cellular rearrangements that lead to the 90* kink of forebrain structures, position hypothalamic tissue underneath the ventral midbrain. These posterior hypothalamic tissues 30 do not express Shh. In addition to Shh expression in the ventral midbrain, a narrow stripe of expressing cells extends dorsally on either side of the third ventricle from the rostral end of the Shh domain in the ventral midbrain to the anterior end of, but not including, the S epiphysis. The most rostral Shh expressing cells are confined to the hypothalamus. In the telencephalon, additional Shh expression is initiated in two 1-2 cell wide stripes.
By 36 hours of development, Shh expression in the ventral CNS has undergone further changes. While expression persists in the floor plate of the tailbud, more rostrally located floor plate cells in the spinal cord cease to express the gene. In contrast, in the hindbrain and forebrain Shh expression persists and is further modified.
At 26-28h, Shh expression appears in the pectoral fin primordia, that are visible as placode like broadenings of cells underneath the epithelial cell layer that covers the yolk. By 33 hrs of development high levels of transcript are present in the posterior margin of the pectoral buds; at the same time, expression is initiated in a narrow stripe at the posterior of the first gill. Expression continues in the pectoral fin buds in lateral cells in the early larva.
At this stage, Shh transcripts are also detectable in cells adjacent to the lumen of the foregut.
(vi) Expression Of Shh In Cyclops And Notail Mutants Two mutations affecting the differentiation of the Axial tissues that express Shh have been described in zebrafish embryos homozygous for the cyclops (cyc) mutation lack a differentiated floorplate (Hatta, K. et al., (1991) Nature 350: 339-341). By contrast, homozygous notail (ntl) embryos are characterized by a failure in notochord maturation and a disruption of normal development of tail structures (Halpern, M.E. et al., (1993) Cell 75: 99- 111).
A change in Shh expression is apparent in cyc embryos as early as the end of gastrulation; at this stage, the anterior limit of expression coincides precisely with Lie two pax-2 stripes in the posterior midbrain. Thus, in contrast to wild-type embryos, no Shh expression is detected in midline structures of the midbrain and forebrain. By the 5 somite 20 stage, Shh transcripts are present in the notochord which at this stage extends until rhombomere 4; however, no expression is detected in more anterior structures. Furthermore, no Shh expression is detected in the ventral neural keel, in particular in the ventral portions of the midbrain and forebrain.
At 24 hours of development, the morphologically visible cyc phenotype consists of a fusion of the eyes at the midline due to the complete absence of the ventral diencephalon. As at earlier developmental stages, Shh expression is absent from neural tissue. Shh expression in the extending tail bud of wild-type embryos is seen as a single row of floor plate cells throughout the spinal cord. In a cyc mutant, no such Shh induction occurs in cells of the 3 ventral spinal cord with the exception of some scattered cells that show transient expression near the tail. Similarly, no Shh expression is seen rostrally in the ventral neural tube.
However, a small group- of Shh expressing cells is detected underneath the epiphysis which presumably correspond to the dorsal-most group of Shh expressing cells in the diencephalon of wild-type embryos.
In homozygous notail (ntl) embryos, no Shh staining is seen in mesodermal tissue at 24 hours of development, consistent with the lack of a notochord in these embryos; by 99 contrast, expression throughout the ventral CNS is unaffected. At the tail bud stage, however, just prior to the onset of somitogenesis, Shh expression is clearly detectable in notochord precursor cells.
(vii) Injection Of Synthetic Shh Transcripts Into Zebrafish Embryos Induces Expression Of A Floor Plate Marker To investigate the activity of Shh in the developing embryo, an over-expression strategy, similar to that employed in the analysis of gene function in Xenopus, was adopted.
Newly fertilized zebrafish eggs were injected with synthetic Shh RNA and were fixed 14 or 24 hours later. As an assay for possible changes in cell fate consequent upon the ectopic activity of Shh, we decided to analyze Axial expression, since this gene serves as a marker for cells in which Shh is normally expressed. A dramatic, though highly localized ectopic expression of Axial in a significant proportion (21/80) of the injected embryos fixed after 24 hours of development is observed. Affected embryos show a broadening of the Axial expression domain in the diencephalon and ectopic Axial expression in the midbrain; however, in no case has ectopic expression in the telencephalon or spinal cord been observed.
Many of the injected embryos also showed disturbed forebrain structures, in particular smaller ventricles and poorly developed eyes. Amongst embryos fixed after 14h, a similar proportion (8/42) exhibit the same broadening and dorsal extension of the Axial stripe in the diencephalon as well as a dorsal extension of Axial staining in the midbrain; again, no changes in Axial expression were observed caudal to the hindbrain with the exception of an increased number of expressing cells at the tip of the tail.
(viii) Overexpression OfShh In Drosophila Embryos Activates The hh-Dependent Pathway *In order to discover whether the high degree of structural homology between the Drosophila and zebrafish hh genes also extends to the functional level, an overexpression system was used to test the activity of Shh in flies. Expression of Dros-HH driven by the HSP70 promoter results in the ectopic activation of both the normal targets of hh activity; the wg transcriptional domain expands to fill between one third to one half of each parasegment whereas ptc is ectopically activated in all cells except those expressing en (Ingham, P.W.
(1993) Nature 366:560-562). To compare the activities of the fly and fish genes, flies 30 transgenic for a HS Shh construct were generated described above and subjected to the same heat shock regime as H Shh transgenic flies. HS Shh embryos fixed immediately after the second of two 30 min heat shocks exhibit ubiquitous transcription of the Shh cDNA.
Similarly treated embryos were fixed 30 or 90 min after the second heat shock and assayed for wg or ptc transcription. Both genes were found to be ectopically activated in a similar manner to that seen in heat shocked H Shh embryos; thus, the zebrafish Shh gene can activate the same pathway as the endogenous hh gene.
Examle Cloning, Expression and Localization of Human Hedgehogs Experimental Procedures Isolation of human hedgehog cDNA clones.
Degenerate nucleotides used to clone chick Shh (Riddle et al., (1993) Cell 75:1401- 1416) were used to amplify by nested PCR human genomic DNA. The nucleotide sequence of these oligos is as follows: vHH50:5'-GGAATTCCCAG(CA)GITG(CT)AA(AG)GA(AG)(CA)(AG)I(GCT)TIAA-3' (SEQ ID NO:18); vHH30:5'-TCATCGATGGACCCA(GA)TC(GA)AAICCIGC(TC)TC-3' (SEQ ID NO: 19); vHH3I:5'-GCTCTAGAGCTCIACIGCIA(GA)IC(GT)IGGIA-3' (SEQ ID The expected 220 bp PCR product was subcloned into pGEM7zf (Promega) and sequenced using Sequenase v2.0 Biochemicals). One clone showed high nucleotide similarity to mouse Ihh and mouse Shh sequence (Echelard et al., (1993) Cell 75:1417-1430) and it was used for screening a human fetal lung 5'-stretch plus cDNA library (Clontech) in 2.
gtl 0 phage. The library was screened following the protocol suggested by the company and two positive plaques were identified, purified, subcloned into pBluescript SK+ (Stratagene) and sequenced, identifying them as the human homologues of Shh (SEQ ID NO:6) and Ihh (SEQ ID NO:7).
SOne clone contained the full coding sequence of a human homolog of Shh as well as 150 bp of 5' and 36 bp of 3' untranslated sequence. The other clone, which is the human 25 homolog of Ihh, extends from 330 bp 3' of the coding sequence to a point close to the predicted boundary between the first and second exon. The identity of these clones was e.o determined by comparison to the murine and chick genes. The protein encoded by human Shh has 92.4% overall identity to the mouse Shh, including 99% identity in the aminoterminal half. The carboxyl-terminal half is also highly conserved, although it contains short S 30 stretches of 16 and 11 amino acids not present in the mouse Shh. The human Ihh protein is 96.8% identical to the mouse Ihh. The two predicted human proteins are also highly related, particularly in their amino-terminal halves where they are 91.4% identical. They diverge significantly in their carboxyl halves, where they show only 45.1% identity. The high level of similarity in the amino portion of all of these proteins implies that this region encodes domains essential to the activity of this class of signaling molecules.
Io Northern blotting Multiple Tissue Northern Blot (Clontech) prepared from poly A+RNA isolated from human adult tissues was hybridized with either full length 3 2 p-labeled human Shh clone or 32 p-labeled human Ihh clone following the protocol suggested by the company.
Digoxigenin in situ hybridization.
Sections: tissues from normal human second trimester gestation abortus specimens were washed in PBS and fixed overnight at 4 0 C paraformaldehyde in PBS, equilibrated 24 hours at 4°C in 50% sucrose in PBS and then placed in 50% sucrose in oct for one hour before embedding in oct. Cryostat sections (10-25 mm) were collected on superfrost plus slides (Fisher) and frozen at -80 0 C until used. Following a postfixation in 4% paraformaldehyde the slides were processed as in Riddle et al., (1993) Cell 75:1401-1416 with the following alterations: proteinase K digestion was performed at room temperature from 1-15 minutes (depending on section thickness), prehybridization, hybridization and washes time was decreased to 1/10 of time.
Whole-mounts: tissues from normal second trimester human abortus specimens were washed in PBS, fixed overnight at 4 0 C in 4% paraformaldehyde in PBS and then processed as in Riddle et al., (1993) Cell 75:1401-1416.
Isolation of an Shh PI clone.
The human Shh gene was isolated on a PI clone from a P1 library (Pierce and Sternberg, 1992) by PCR (polymerase chain reaction) screening. Two oligonucleotide primers were derived from the human Shh sequence. The two olignucleotide primers used for 25 PCR were: SHHF5'-ACCGAGGGCTGGGACGAAGATGGC-3' (SEQ ID NO:43) SHR5'-CGCTCGGTCGTACGGCATGAACGAC-3' (SEQ ID NO:44) The PCR reaction was carried using standard conditions as described previously (Thierfelder et al., 1994) except that the annealing temperature was 65 0 C. These primers amplified a 119 bp fragment from human and P1 clone DNA. The P1 clone was designated SHHP1. After the P1 clone was isolated these oligonucleotides were used as sequencing primers. A 2.5KbEcoRI fragment that encoded a CA repeat was subcloned from this P1 clone using methods described previously (Thierfelder et al. 1994). Oligonucleotide primers that amplified this CA repeat sequence were fashioned from the flanking sequences: 35 SHHCAF5'-ATGGGGATGTGTGTGGTCAAGTGTA-3' (SEQ ID SHHCARS'-TTCACAGACTCTCAAAGTGTATTTT-3' (SEQ ID NO:46) /0/ Mapping the human Ihh and Shh genes.
The human Ihh gene was mapped to chromosome 2 using somatic cell hybrids from NIGMS mapping pannel 2 (GM10826B).
The Shh gene was mapped to chromosome 7 using somatic cell hybrids from NIGMS mapping panel 2 (GM10791 and GM 10868).
Linkage between the limb deformity locus on chromosome 7 and the Shh gene was demonstrated using standard procedures. Family LD has been described previously (Tkukurov et (1994) Nature Genet. 6:282-286). A CA repeat bearing sequence near the Shh gene was amplified from the DNA of all members of Family LD by PCR using the SHHCAF and SHHCAR primers. Linkage between the CA repeat and the LD disease gene segregating in Family LD was estimated by the MLINK program (Oct, 1967). Penetrance was set at 100% and the allele frequencies were determined using unrelated spouses in the LD family.
Interspecific Backcross Mapping.
SInterspecific backcross progeny were generated by mating (C57BL/6J x M. spretus) Fl females and C57BL/6J males as described (Copeland and Jenkins, (1991) Trends Genet.
7:113-118). A total of 205 N2 mice were used to map the Ihh and Dhh loci. DNA isolation, restriction enzyme digestions, agarose gel electrophoresis, Southern blot transfer and hybridization were performed essentially as described (Jenkins et al., (1982) J. Virol. 43:26- 36). All blots were prepared with Hybond-N+ nylon membrane (Amersham). The probe, an 1.8kb EcoRI fragment of mouse cDNA, detected a major fragment of 8.5 kb in C57BL/6j DNA and a major fragment 6.0 kb in M. spretus DNA following digestion with Bgll.
25 The Shh probe, an 900 bp Smai fragment of mouse cDNA, detected HinclI fragments of and 2.1 kb as well as 4.6 and 2.1 The Dhh probe, and 800 bp BamHilEcoRi fragment of mouse genomic DNA, detected major fragments of 4.7 and 1.3 kb and 8.2 and 1.3 kb following digestion with SphI. The presence or absence of M spretus specific fragments was followed in backcross mice.
A description of the probes and RFLPs for loci used to position the Ihh, Shh and Dhh loci in the interspecific backcross has been reported. These include: Fnl, Vil and Acrg, chromosome 1 (Wilkie et al., (1993) Genomics 18:175-184), Gnail, En2, 116, chromosomes (Miao et al., (1994) PNAS USA 91:11050-11054) and Pdgfb, Gdcl and Rarg, chromosome 15 (Brannan et al., (1992) Genomics 13:1075-1081). Recombination distances were calculated as described (Green, (1981) Linkage, recombination and mapping. In "Genetics and Probability in Animal Breeding Experiments", pp. 77-113, Oxford University Press, NY) using the computer program SPRETUS MADNESS. Gene order was determined by /02 minimizing the number of recombination events required to explain the allele distribution patterns.
(ii) Expression of Human Shh and Ihh To investigate the tissue distribution of Shh and Ihh expression, poly(A)+RNA samples from various adult human tissues were probed with the two cDNA clones. Of the tissues tested, an Ihh-specific message of -2.7 kb is only detected in liver and kidney. Shh transcripts was not detected in the RNA from any of the adult tissues tested. All the samples contained approximately equal amounts of intact RNA, as determined by hybridization with a control probe.
The hedgehog family of genes were identified as mediators of embryonic patterning in flies and vertebrates. No adult expression of these genes had previously been reported.
These results indicate that Ihh additionally plays a role in adult liver and kidney. Since the hedgehog genes encode intercellular signals, Ihh may function in coordinating the properties of different cell types in these organs. Shh may also be used as a signaling molecule in the adult, either in tissues not looked at here, or at levels too low to be detected under these conditions.
In situ hybridization was used to investigate the expression of Shh in various midgestational human fetal organs. Shh expression is present predominantly in endoderm derived tissues: the respiratory epithelium, collecting ducts of the kidney, transitional epithelium of the ureter, hepatocytes, and small intestine epithelium. Shh was not detectable S in fetal heart or placental tissues. The intensity of expression is increased in primitive differentiating tissues (renal blastema, base villi, branching lung buds) and decreased or absent in differentiated tissues glomeruli). Shh expression is present in the mesenchyme immediately abutting the budding respiratory tubes. The non-uniform pattern of Shh expression in hepatocytes is consistent with expression of other genes in adult liver (Dingemanse et al., (1994) Differentiation 56:153-162). The base of villi, the renal blastema, and the lung buds are all regions expressing Shh and they are areas of active growth and differentiation, suggesting Shh is important in these processes.
(iii) The Chromosomal Map Location of Human Shh and Ihh.
Since Shh is known to mediate patterning during the development of the mouse and chick and the expression of Shh and Ihh are suggestive of a similar role in humans, mutations 35 in these genes would be expected to lead to embryonic lethality or congenital defects. One way of investigating this possibility is to see whether they are genetically linked to any known inherited disorders.
/03 Shh- and Ihh-specific primers were designed from their respective sequences and were used in PCR reactions on a panel of rodent-human somatic cell hybrids. Control rodent DNA did not amplify specific bands using these primers. In contrast, DNA from several rodenthuman hybrids resulted in PCR products of the appropriate size allowing us to assign Shh to chromosome 7q and Ihh to chromosome 2.
One of the central roles of chick Shh is in regulating the anterior-posterior axis of the limb. A human congenital polysyndactyly has recently been mapped to chromosome 7q36 (Tsukurov et al., (1994) Nature Genet. 6:282-286; Heutink et al., (1994) Nature Genet.
6:287-291). The phenotype of this disease is consistent with defects that might be expected from aberrant expression of Shh in the limb. Therefore, the chromosomal location of Shh was mapped more precisely, in particular in relation to the polysyndactyly locus.
A P1 phage library was screened using the Shh specific primers for PCR amplification and clone SHHP1 was isolated. Clone SHHPI contained Shh sequence. A Southem blot of an EcoRl digest of this phage using probe demonstrated that a 2.5 Kb EcoRI fragment contained a CA repeat. Nucleotide sequence analysis of this subcloned EcoRI fragment~ demonstrated that the CA repeat lay near the EcoRI sites. Primers flanking the CA repeat we:re designed and used to map the location of Shh relative to other markers on 7q in individuals of a large kindred with complex polysyndactyly (Tsukurov et al., (1994) Nature Genet. 6:282-286). Shh maps close to D75550 on 7q36, with no recombination events seen in this study. It is also extremely close to, but distinct from, the polysyndactyly locus with one recombination event observed between them (maximum lod score 4.82, 0.05).
One unaffected individual (pedigree ID V-10 in Tsukurov et al., (1994) Nature Genet. 6:282- 286) has the Shh linked CA repeat allele found in all affected family members. No recombination was observed between the locus En2 and the Shh gene (maximum lod score 25 1.82, e 0.0).
(iv) Chromosomal mapping of the Murine Ihh, Shh and Dhh genes.
The murine chromosomal location of Ihh, Shh and Dhh was determined using an t* interspecific backcross mapping panel derived from crosses of [(C57BL/6J x M. spetrus)F1 X C57BL/J)] mice. cDNA fragments from each locus were used as probes in Southern blot hybridization analysis of C57BL/6J and M. spretus genomic DNA that was separately .digested with several different restriction enzymes to identify informative restriction fragment length polymorphisms (RFLPs) useful for gene mapping. The strain distribution pattern of each RFLP in the interspecific backcross was then determined by following the S. 35 presence or absence of RFLPs specific for M. spretus in backcross mice.
Ihh mapped to the central region of mouse chromosome 1, 2.7 cM distal of Fnl and did not recombine with Vil in 190 animals typed in common, suggesting that the two loci are within 1.6 cM (upper 95% confidence level) (Fig. 16). Shh mapped to the proximal region of /O 4j mouse chromosome 5, 0.6 cM distal of En2 and 1.9 cM proximal of 116 in accordance to Chang et al., (1994) Development 120:3339-3353. Dhh mapped to the very distal region of mouse chromosome 15, 0.6 cM distal of Gdcl and did not recombine with Rarg in 160 animals typed in common, suggesting that the two loci are within 1.9 cM of each other (upper 95% confidence level) (Fig. 16).
Interspecific maps of chromosome 1, 5 and 15 were compared with composite mouse linkage maps that report the map location of many uncloned mouse mutations (compiled by M.T. Davisson, T.H. Roderick, A.L. Hillyard and D.P. Doolittle and provided from GBASE, a computerized database maintained at The Jackson Laboratory, Bar Harbor, ME). The hemimelic extra-toe (Hx) mouse mutant maps 1.1 cM distal to En2 on chromosome 5 (Martin et al., (1990) Genomics 6:302-308), a location in close proximity to where Shh has been positioned. Hx is a dominant mutation which results in preaxial polydactyly and hemimelia affecting'all four limbs (Dickie, (1968) Mouse News Lett 38:24; Knudsen and Kochhar, (1981) L. Embryol. Exp. Morph. 65: Suppl. 289-307). Shh has previously been shown to be expressed in the limb (Echelard et al., (1993) Cell 75:1417-1430). To determine whether Shh and Hx are tightly linked we followed their distribution ,n a backcross panel in which Hx was segregating. Two recombinant,: between Shh and Hx were identified, thus excluding the possibili y that the two loci are .llelic and these observations are again consistent with those of Chang et al., (1994) Development 120:3339-3353. W'hile there are several other mutations in the vicinity of Ihh and Dhh, none is an obvious candidate for an alteration in the corresponding gene.
The central region of mouse chromosome 1 shares homology with human chromosome 2q (summarized in Fig. 16). Placement of Ihh in this interval suggests the human homolog of Ihh will resice on 2q, as well. Similarly, it is likely that human homolog 25 of Dhh will reside on human chromosome 12q.
Example 6 "Proteolytic Processing Yields Two Secreted Forms of Sonic Hedgehog Experimental Procedures 30 In vitro Translation and Processing Mouse and chick sonic hedgehog coding sequences were inserted into the vector pSP64T (kindly provided by D. Melton) which contains an SP6 phage promoter and both and 3' untranslated sequences derived from the Xenopus laevis P-Globin gene. After restriction endonuclease digestion with Sal I to generate linear templates, RNA was transcribed in vitro using SP6 RNA polymerase (Promega, Inc.) in the presence of 1 mM cap structure analog (m 7 G(5')ppp(5')Gm; Boehringer-Mannheim, Inc.) Following digestion with RQI DNase I (Promega, Inc.) to remove the DNA template, transcripts were purified by phenol:choloroform extraction and ethanol precipitation.
Rabbit reticulocyte lysate (Promega, Inc.) was used according to the manufacturer's instructions. For each reaction, 12.5 ul of lysate was programmed with 0.5-2.0 pg of in vitro transcribed RNA. The reactions contained 20 pCi of Express labeling mix (NEN/DuPont, Inc.) were included. To address processing and secretion in vitro, 1.0-2.0 pl1 of canine pancreatic microsomal membranes (Promega, Inc.) were included in the reactions. The final reaction volume of 25 pl was incubated for one hour at 300C. Aliquots of each reaction (between 0.25 and 3.0 pi) were boiled for 3 minutes in Laemmli sample buffer (LSB: 125 mM Tris-Hcl [pH 2% SDS; 1% 2-mercaptoethanol; 0.25 mg/ml bromophenol blue) before separating on a 15% polyacrylamide gel. Fixed gels were processed for fluorography using EnHance (NEN/DuPont, Inc.) as described by the manufacturer.
Glycosylation was addressed by incubation with Endoglycosidase H (Endo H; New England Biolabs, Inc.) according to the:manufacturer's directions. Reactions were carried out for 1-2 hr at 37 0 C before analyzing reaction products by polyacrylamide gel electrophoresis
(PAGE).
Xenopzs Oocyte Injection and Labe!ing Oocytes were enzymatically defolliculated and rinsed with OR2 (50 mM HEPES [pH 82 mM NaCI, 2.5 mM KC1, 1.5 mM Na2HPO4). Healthy stage six oocytes were 20 injected with 30 ng of in vitro transcribed, capped mouse Shh RNA (prepared as described above). Following a 2 hr recovery period, healthy injected oocytes and uninjected controls were cultured at room temperature in groups of ten ia 96-well dishes containing 0.2 ml of OR2 (supplemented with 0.1 mg/ml Gentamicin and 0.4 mg/ml BSA) per well. The incubation medium was supplemented with 50 pCi of Express labeling mix. Three days after injection, the culture media were collected and expression of Shh protein analyzed by immunoprecipitation. Oocytes were rinsed several tines in OR2 before lysing in TENT mM Tris-HCl [pH 150 mM NaCI, 2mM EDTA; 1% Triton-X-100; 10 pl/oocyte) supplemented with 1 ,ug/mi aprotinin, 2 gg/ml leupeptin and ImM phenylmethylsufonylfluoride (PMSF). After centrifu ation at 13000 x g for 10 minutes at 30 40C, soluble protein supematants were recovered and analyzed by immunoprecipitation (see below).
Cos Cell Transfection and Labeling Cos cells were cultured in Dulbecco's Modified Eagle Medium (DMEM; Sigma, Inc.) supplemented with 10% fetal bovine serum (Gibco/BRL), 2 mM L-Glutamine (Gibco/BRL) and 50 mU/ml penicillin and 50 .ig/ml streptomycin (Gibco/BRL). Subconfluent cos cells in mm or 60 mm dishes (Falcon, Inc.) were transiently transfected with 2 mg or 6 mg supercoiled plasmid DNA, respectively. Between 42 and 44 hr post-transfection, cells were labeled for 4-6 hr in 0.5 ml (35 mm dishes) or 1.5 ml (60 mm dishes) serum-free DMEM lacking Cysteine and Methionine (Gibco/BRL) and supplemented with 125 iCi/ml each of Express labeling mix and L-35S-Cysteine (NEN/DuPont). After labeling, media were collected and used for immunoprecipitation. Cells were rinsed with cold PBS and lysed in the tissue culture dishes by the addition of 0.5 ml (35 mm dishes) or 1.5 ml (60 mm dishes) TENT (with protease inhibitors as described above) and gentle rocking for 30 minutes at Lysates were cleared by centrifugation (13000 x g for 5 min. at 40C) and the supematants were analyzed by immunoprecipitation (see below).
Baculovirus Production and Infection A recombinant baculovirus expressing mouse sonic hedgehog with a myc epitope tag inserted at the carboxy terminus was generated using the Baculogold kit (Pharmingen, Inc.).
The initial virus production used Sf 9 cells, followed by two rounds of amplification in High Five cells (Invitrogen, Inc.) in serum-free medium (ExCell 401; Invitrogen, Inc.). A baculovirus lacking Shh coding sequences was also constructed as a control. For protein induction, High Five cells were infected at a multiplicity of approximately 15. Three days later, medium and cells were collected by gentle pipetting. Cells were collected by centrifugation (1000 x g) and the medium was recovered for Western blot analysis. Cell 20 pellets were washed twice in cold PBS and lysed in TENT plus protease inhibitors (see above) by rotating for 30 minutes at 4 0 C in a microcentrifuge tube. The lysate was cleared as described above prior to Western blotting.
Western Blotting For Western blotting, 0.25 ml samples of media of the total) were precipitated with TCA and redissolved in 15 tl of LSB. Cell lysate samples of total) were brought to a final volume of 15 p. with water and concentrated (5X) LSB Samples were boiled S minutes prior to separation on a 15% acrylamide gel. Proteins were transferred to PVDF membrane (Immobilon-P; Millipore, Inc.) and blocked in BLOTTO w/v non-fat dried milk in PBS) containing 0.2% Tween-20. Hybridoma supernatant recognizing the human c- 30 myc epitope (9E10; Evan, G.I. et al., (1985) Mol. Cell. Biol. 5:3610-3616) was added at a dilution of 1:200 for one hour followed by a 1:5000 dilution of Goat anti-Mouse-Alkaline phosphatase conjugate (Promega, Inc.) for 30 minutes. Bands were visualized using the Lumi-Phos 530 reagent (Boehringer-Mannheim) according to the manufacturer's directions.
107 Immunoprecipitation Cell lysates (Xenopus oocytes or cos cells) were brought to 0.5 ml with TENT (plus protease inhibitors as above). Media samples (OR2 or DMEM) were cleared by centrifugation at 13000 x g for 5 min. (4 0 C) and 10X TENT was added to a final concentration of IX (final volume: 0.5-1.5 ml). The c-myc monoclonal antibody hybridoma supernatant was added to 1/20 of the final volume. Samples were rotated for 1 hr at then 0.1 ml of 10% protein A-Sepharose CL-4B (Pharmacia, Inc.) was added. Samples were rotated an additional 14-16 h. Immune complexes were washed 4 times with 1.0 ml TENT. Immunoprecipitated material was eluted and denatured by boiling for 10 minutes in 25 pl IX LSB. Following centrifugation, samples were separated on 15% acrylamide gels and processed for fluorography as described previously. Samples for Endo H digestion were eluted and denatured by boiling for 10 minutes in the provided denaturation buffer followed by digestion with Endo H for 1-2 hr at 370C. Concentrated (SX) LSB was added and the samples were processed for electrophoresis as described.
For immunoprecipitation with the anti-mouse Shh serum, samples (Cos cell lysates and DMEM) were precleared by incubating 1 hr on ice with 3 pl pre-immune serum, followed by the addition of 0.1 ml 10% Protein A-Sepharose. After rotating for 1 hr at 4 C, supematants were recovered and incubatec .or 1 hr on ice with 3 pl depleted anti-mouse Shh serum (see below).. incubation with Proteirn A-Sepharose, w shing, elution and 20 electrophoresis were then performed as described above.
Immunofluorescent Staining of Cos Cells Twenty-four hours after transfection, cells were transferred to 8-chamber slides (Lab- Tek, Inc.) and allowed to attach an additional twenty-four hours. Cells were fixed in 2% paraformaldehyde/0.1% glutaraldehyde, washed in PBS and permeabilized in 1% Triton-X- 100 (Munro, S. and Pelham, (1987) Cell 48:899-907). After washing in PBS, cells were treated for 10 minutes in 1 mg/ml sodium borohydride. Cells were incubated with the c-myc monoclonal antibody hybridoma supernatant (diluted 1:10) and the affinity purified mouse Sonic hedgehog antiserum (diluted 1:4) for 45 minutes followed by incubation in 1:100 Goat-anti Mouse IgG-RITC plus 1:100 Goat anti Rabbit IgG FITC (Southern 30 Biotechnology Associates, Inc.) for 45 minutes. DAPI (Sigma, Inc.) was included at 0.3 i.
g/ml The slides were mounted in Slo-Fade (Molecular Probes, Inc.) and photographed on a Leitz DMR compound microscope.
Antibody Production and Purification A PCR fragment encoding amino acids 44-143 of mouse Sonic hedgehog was cloned in frame into the Eco RI site of pGEX-2T (Pharmacia, Inc.). Transformed bacteria were 1o g induced with IPTG and the fusion protein purified on a Glutathione-Agarose affinity column (Pharmacia, Inc.) according to the manufacturer's instructions. Inoculation of New Zealand White rabbits, as well as test and production bleeding were carried out at Hazelton Research Products, Inc.
To deplete the serum of antibodies against Glutathione-S-transferase (GST) and bacterial proteins, a lysate of E. coli transformed with pGEX-2T and induced with IPTG was coupled to Affi-Gel 10 (Bio-Rad, Inc.) The serum was incubated in batch for two hours with the depletion matrix before centrifugation (1000 x g for 5 min.) and collection of the supernatant. To make an affinity matrix, purified bacterially expressed protein corresponding to the amino terminal two-thirds of mouse Sonic hedgehog was coupled to Affi-Gel 10 (Bio- Rad, Inc.). The depleted antiserum was first adsorbed to this matrix in batch, then transferred to a column. The matrix was washed with TBST (25 mM Tris-HCI [pH 140 mM NaCI, mM KCI, 0.1% Triton-X-100), and the purified antibodies were eluted with ten bed volumes of 0.15 M Glycine,.[pH The solution was neutralized with one volume of 1 M Tris-HCI [pH and dialyzed against 160 volumes of PBS.
Other antibodies have been generated against hedgehog proteins and three polyclonal rabbit antisera obtained to hh proteins can be characterized as follows:Ab77 -reacts only with the carboxyl processed chick Shh peptide (27 kd); Ab79 -reacts with amino processed chick, mouse and human Shh peptide (19 kd). Weakly reacts with 27 kd peptide from chick and 20 mouse. Also reacts with mouse Ihh; and Ab80 -reacts with only amino peptide (19kd) of chick, mouse and human.
(ii) In Vitro Translated Sonic Hedgehog is Proteolytically Processed and Glycosylated The open reading frames of chick and mouse Shh encode primary translation products of 425 and 437 amino acids, respectively, with predicted molecular masses of 46.4 kilodaltons (kDa) and 47.8 kDa (Echelard, Y. et al., (1993) Cell 75:1417-1430; Riddle, R.D.
et al., (1993) Cell 75:1401-1416). Further examination of the protein sequences revealed a short stretch of amino terminal residues (26, for chick, 24 for mouse) that are highly hydrophobic and are predicted to encode signal peptides. Removal of these sequences would 30 generate proteins of 43.7 kDa (chick Shh) and 45.3 kDa (mouse Shh). Also, each protein contains a single consensus site for N-linked glycosylation (Tarentino, A.L. et al., (1989) Methods Cell Biol. 32:111-139) at residue 282 (chick) and 279 (mouse). These features of the Shh proteins are summarized in Figure 11.
A rabbit reticulocyte lysate programmed with in vitro translated messenger RNA encoding either chick or mouse Shh synthesizes proteins with molecular masses of 46 kDa and 47 kDa, respectively. These values are in good agreement with those predicted by 9 examination of the amino acid sequences. To examine posttranslational modifications of Shh proteins, a preparation of canine pancreatic microsomal membranes was included in the translation reactions. This preparation allows such processes as signal peptide cleavage and core glycosylation. When the Shh proteins are synthesized in the presence of these membranes, two products with apparent molecular masses of approximately 19 and 28 kDa (chick), or 19 and 30 kDa (mouse) are seen in addition to the 46 kDa and 47 kDa forms.
When the material synthesized in the presence of the membranes is digested with Endoglycosidase H (Endo the mobilities of the two larger proteins are increased. The apparent molecular masses of the Endo H digested forms are 44 kDa and 26 kDa for chick Shh, and 45kDa and 27 kDa for mouse Shh. The decrease in the molecular masses of the largest proteins synthesized in the presence of the microsomal membranes after Endo H digestion is consistent with removal of the predicted signal peptides. The mobility shift following Endo H treatment indicates that N-linked glycosylation occurs, and that the 26 kDa (chick) and 27 kDa (mouse) proteins contain the glycosylation sites.
The appearance of the two lower molecular weight bands (hereafter referred to as the "processed forms") upon translation in the presence of microsomal membranes suggests that a proteolytic event in addition to signal peptide cleavage takes place. The combined molecular masses of the processed forms (19 kDa and 26 kDa for chick; 19 kDa and 27 kDa for mouse) add up to approximately the predicted masses of the signal peptide cleaved S 20 proteins (44 kDa for chick and 45 kDa for mouse) suggesting that only a single additional cleavage occurs.
The mouse Shh protein sequence is 12 amino acid residues longer than the chick sequence (437 versus 425 residues). Alignment of the chick and mouse Shh protein sequences reveals that these additional amino acids are near the carboxy terminus of the 25 protein (Echelard, Y. et al., (1993) Cell 75:1417-1430). Since the larger of the processed forms differ in molecular mass by approximately 1 kDa between the two species, it appears that these peptides contain the carboxy terminal portions of the Shh proteins. The smaller processed forms, whose molecular masses are identical, presumably consist of the amino terminal portions.
30 (iii) Secretion ofShh Peptides To investigate the synthesis of Shh proteins in vivo, the mouse protein was expressed in several different eukaryotic cell types. In order to detect synthesized protein, and to facilitate future purification, the carboxy terminus was engineered to contain a twenty-five amino acid sequence containing a recognition site for the thrombin restriction protease followed by a ten amino acid sequence derived from the human c-myc protein and six consecutive histidine residues. The c-myc sequence serves as an epitope tag allowing /1 0 detection by a monoclonal antibody (9E10; Evan, G.I. et al., (1985) Mol. Cell Biol. 5:3610- 3616). The combined molecular mass of the carboxy terminal additions is approximately 3 kDa.
Xenopus laevis oocytes Immunoprecipitation with the c-myc antibody detects several proteins in lysates of metabolically labeled Xenopus laevis oocytes injected with Shh mRNA. Cell lysates and medium from 35 S labeled oocytes injected with RNA encoding mouse Shh with the c-myc epitope tag at the at the carboxy terminus, or from control oocytes were analyzed by immunoprecipitation with c-myc monoclonal antibody. A band of approximately 47 kDa is seen, as is a doublet migrating near 30 kDa. Treatment with Endo H increases the mobility of the largest protein, and resolves the doublet into a single species of approximately 30 kDa.
These observations parallel the behaviors seen in vitro. Allowing for the added mass of the carboxy terminal additions, the largest protein would correspond to the signal peptide cleaved form, while the doublet would represent the glycosylated and unglycosylated larger processed form. Since the epitope tag was placed at the carboxy terminus of the protein, the identity of the 30 kDa peptide as the carboxy terminal portion of Shh is confirmed. Failure to detect the 19 kDa species supports its identity as an amino terminal region of the protein.
To test whether Shh is secreted by Xenopus oocytes, the medium in which the injected oocytes were incubated was probed by immunoprecipitation with the c-myc 20 antibody. A single band migrating slightly more slowly than the glycosylated larger Sprocessed form was observed. This protein is insensitive to Endo H. This result is expected S since most secreted glycoproteins lose sensitivity to Endo H as they travel through the Golgi apparatus and are modified by a series of glycosidases (Kornfeld, R. and Komfeld, (1985) Annu. Rev. Biochem. 54:631-664). The enzymatic maturation of the Asn-linked carbohydrate moiety could also explain the slight decrease in mobility of the secreted larger protein versus the intracellular material. Following Endo H digestion, a band with a slightly lower mobility than the signal peptide cleaved protein, is also apparent, suggesting that some Shh protein is secreted without undergoing proteolytic processing. Failure to detect this protein in the medium without Endo H digestion suggests heterogeneity in the extent of carbohydrate "i 30 modification in the Golgi preventing the material from migrating as a distinct band.
Resolution of this material into a single band following Endo H digestion suggests that the carbohydrate structure does not mature completely in the Golgi apparatus. Structural differences between the unprocessed protein and the larger processed form could account for this observation (Komfeld, R. and Kornfeld, (1985) Annu. Rev. Biochem. 54:631-664).
Cos cells The behavior of mouse Shh in a mammalian cell type was investigated using transfected cos cells. Synthesis and secretion of the protein was monitored by immunoprecipitation using the c-myc antibody. Transfected cos cells express the same Sonic hedgehog species that were detected in the injected Xenopus oocytes, and their behavior following Endo H digestion is also identical. Furthermore, secretion of the 30 kDa glycosylated form is observed in cos cells, as well as the characteristic insensitivity to Endo H after secretion. Most of the secreted protein co-migrates with the intracellular, glycosylated larger processed form, but a small amount of protein with a slightly lower mobility is also detected in the medium. As in the Xenopus oocyte cultures, some Shh which has not undergone proteolytic processing is evident in the medium, but only after Endo H digestion.
Baculovirus infected cells To examine the behavior of the mouse Shh protein in an invertebrate cell type, and to potentially purify Shh peptides, a recombinant baculovirus was constructed which placed the Shh coding sequence, with the carboxy terminal tag, under the control of the baculoviral Polyhedrin gene promoter. When insect cells were infected with the recombinant baculovirus, Shh peptides could be detected in cell lysates and medium by Western blotting S* with the c-myc antibody.
20 The Shh products detected in this system were similar to those described above.
However, virtually no unprocessed protein was seen in cell lysates, nor was any detected in the medium after Endo H digestion. This suggests that the proteolytic processing event occurs more efficiently in these cells than in either of the other two cell types or the in vitro translation system. A doublet corresponding to the glycosylated and unglycosylated 30 kDa forms is detected, as well as the secreted, Endo I resistant peptide as seen in the other expression systems. Unlike the other systems, however, all of the secreted larger processed form appears to comigrate with the glycosylated intracellular material.
(iv) Secretion of a Highly Conserved Amino Terminal Peptide To determine the behavior of the amino terminal portion of the processed Sonic hedgehog protein, the c-myc epitope tag was positioned 32 amino acids after the putative signal peptide cleavage site (Figure 12). Cos cells were transfected with Shh expression constructs containing the c-myc tag at the carboxy terminus or near the amino terminus.
When this construct was expressed in cos cells, both the full length protein and the smaller processed form (approximately 20 kDa due to addition of the c-myc tag) were detected by immunoprecipitation of extracts from labeled cells. However, the 20 kDa product is barely I 2.
detected in the medium. In cells transfected in parallel with the carboxy terminal c-myc tagged construct, the full length and 30 kDa products were both precipitated from cell lysates and medium as described earlier.
As the amino terminal c-myc tag may affect the secretion efficiency of the smaller processed form, the expression of this protein was examined in cos cells using an antiserum directed against amino acids 44 through 143 of mouse Shh (Figure 12). After transfection with the carboxy-terminal c-myc tagged construct, immunoprecipitation with the anti-Shh serum detected a very low level of the smaller processed form in the medium despite a strong signal in the cell lysate. This recapitulates the results with the myc antibody.
To examine the subcellular localization of Shh proteins, cos cells were transfected with the carboxy terminal tagged Shh construct and plated on multi-chamber slides, fixed and permeabilized. The cells were incubated simultaneously with the anti-Shh serum and the cmyc antibody followed by FITC conjugated Goat anti-Rabbit-IgG and RITC conjugated Goat anti-Mouse-IgG. DAPI was included to stain nuclei. Strong perinuclear staining characteristic of the Golgi apparatus was observed with the anti-Shh serum. The same subcellular region was also stained using the c-myc antibody. The coincidence of staining patterns seen with the two antibody preparations suggest that the low level of the smaller processed form detected in the medium is not due to its retention in the endoplasmic reticulum.
20 Hedgehog Processing In summary, the results discussed above demonstrate that the mouse and chick Shh genes encode secreted glycoproteins which undergo additional proteolytic processing. Data indicate that this processing occurs in an apparently similar fashion in a variety of cell types suggesting that it is a general feature of the Shh protein, and not unique to any particular 25 expression system. For mouse Shh, data indicate that both products of this proteolytic processing are secreted. These observations are summarized in Figure 13.
It was observed that the 19 kDa amino peptide accumulates to a lower level in the medium than the 27 kDa carboxyl peptide. This may reflect inefficient secretion or rapid turnover of this species once secreted. Alternatively, the smaller form may associate with the 30 cell surface or extracellular matrix components making it difficult to detect in the medium.
The insensitivity of the secreted, larger form to Endo H is a common feature of secreted glycoproteins. During transit through the Golgi apparatus, the Asn-linked carbohydrate moiety is modified by a series of specific glycosidases (reviewed in Kornfeld, R. and Komfeld, (1985) Annu. Rev. Biochem 54:631-664; Tarentino, A.L. et al., (1989) Methods Cell Biol. 32:111-139). These modifications convert the structure from the immature "high mannose" to the mature "complex" type. At one step in this process, a Golgi enzyme, a- //3 mannosidase II, removes two mannose residues from the complex rendering it insensitive to Endo H (Kornfeld, R. and Kornfeld, (1985)Annu. Rev. Biochem 54:631-664).
Based on the observed molecular masses of the processed forms of mouse and chick Shh, the predicted secondary proteolytic cleavage site would be located near the border of the sequences encoded by the second and third exons. Interestingly, this region marks the end of the most highly related part of the hedgehog proteins. The amino terminal (smaller) form would contain the most highly conserved portion of the protein. In fact, the amino acids encoded by exons one and two (exclusive of sequences upstream of the putative signal peptide cleavage sites) share 69% identity between Dros-HH and mouse Shh, and 99% identity between chick and mouse Shh. Amino acid identity in the region encoded by the third exon is much lower 30% mouse to Drosophila and 71% mouse to chick (Echelard, Y. et al., (1993) Cell 75:1417-1430). Therefore, the two processed forms of Shh may have conserved as well as divergent signaling activities separated into distinct coding exons in the Shh gene. Furthermore, the observation that some unprocessed protein is secreted by Xenopus oocytes and cos cells raises the possibility that it may have a separate function.
The biochemical behavior of mouse Shh appears to be quite similar to that described for the Drosophila Hedgehog (Dros-HH) protein (Lee, J.L. et al., (1992) Cell 71:33-50; Tabata, T. et al., (1992) Genes Dev. 6:2635-2645). In vitro translatin" of Dros-HH mRNA, in the presence of mnicro.nomes, rvealed products with molecular masses 20 corresponding to full length protein, as well as to the product expected after cleavage of the predicted internal (Type II) signal peptide (Lee, J.L. et al., (1992) Cell 71:33-50).
Interestingly, no additional, processed forms were observed. However, such forms could have been obscured by breakdown products migrating between 20 and 30 kDa. When an RNA encoding a form of the protein lacking the carboxy-terminal 61 amino acids was 25 translated, no breakdown products were seen, but there is still no evidence of the proteolytic processing observed with mouse Shh. A similar phenomenon has been observed in these experiments. A reduction in the extent of proteolytic processing is seen when a mouse Shh protein lacking 10 carboxy-terminal amino acids is translated in vitro or expressed in cos cells (data not shown). This suggests that sequences at the carboxy termini of Dros-HH 30 proteins act at a distance to influence the efficiency of processing.
In vivo, processing of Dros-HH has been demonstrated (Tabata, T. et al., (1992) Genes Dev. 6:2635-2645). Immunoblots of lysates from Schneider cells transfected with a hh expression vector reveal two smaller molecular weight forms similar to those described for mouse Shh. These products were also detected in extracts of larvae and imaginal discs derived from flies expressing a heat shock inducible hh construct. Thus, it is clear that there are also several distinct forms of Dros-HH proteins.
(vi) Hedgehog Signaling In order to satisfy the criteria for intercellular signaling, hedgehog proteins must be detected outside of their domains of expression. This has been clearly demonstrated for Dros-HH. Using an antiserum raised against nearly full length Dros-HH protein, Tabata and Kornberg (Tabata, T. and Komberg, (1992) Cell 76:89-102) detect the protein in stripes that are slightly wider than the RNA expression domains in embryonic segments, and just anterior to the border of the RNA expression domain in wing imaginal discs. Similarly, Taylor, et. al., (1993) Mech. Dev. 42:89-96, detected Dros-HH protein in discrete patches within cells adjacent to those expressing hh RNA in embryonic segments using an antiserum directed against an amino-terminal portion of Dros-HH which, based on the proteolytic processing data (Tabata, T. et al., (1992) Genes Dev. 6:2635-2645), is not likely to recognize the carboxyl cleavage product.
The detection of Dros-HH beyond cells expressing the hh gene is consistent with the phenotype of hh mutants. In these animals, cellular patterning in each embryonic parasegment in disrupted resulting in an abnormal cuticular pattern reminiscent of that seen in wg mutants. Further analysis has revealed that the loss of hh gene function leads to loss of wg expression in a thin stripe of cells just anterior to the hh expression domain (Ingham, P.W.
and Hidalgo, (1993) Development 117:283-291). This stggests that Dros-HH acts to maintain wg expression in neighboring cells. The observation that ubiquitously expressed 20 Dros-HH leads to ectopic activation of wg supports this model (Tabata, T. and Komberg, (1992) Cell 76:89-102). In addition to these genetic studies, there is also indirect evidence that Dros-HH acts at a distance from its site of expression to influence patterning of the epidermis (Heemskerk, J. and DiNardo, (1994) Cell 76:449-460).
The apparent effect of Dros-HH on neighboring cells, as well as on those located at 25 a distance from the site of hh expression is reminiscent of the influence of the notochord and floor plate on the developing vertebrate CNS, and of the ZPA in the limb. The notochord (a site of high level Shh expression) induces the formation of the floor plate in a contact dependent manner, while the notochord and floor plate (another area of strong Shh expression) are both capable of inducing motomeurons at a distance (Placzek, M. et al., 30 (1993) Development 117:205-218; Yamada, T. et al., (1993) Cell 73:673-686).
Moreover ZPA activity is required not only for patterning cells in the extreme posterior of the limb bud where Shh is transcribed, but also a few hundred microns anterior of this zone. Several lines of evidence indicate that Shh is able to induce floor plate (Echelard, Y. et al., (1993) Cell 75:1417-1430; Roelink, H. et al., (1994) Cell 76:761-775) and mediate the signaling activity of the ZPA (Riddle, R.D. et al., (1993) Cell 75:1401-1416). Since it has been shown that Shh is cleaved, it can be speculated that the processed peptides may have distinct activities. The smaller amino terminal form, which appears to be more poorly secreted, less stable or retained at the cell surface or in the extracellular matrix, may act locally. In contrast, the larger carboxy terminal peptide could possibly function at a distance.
In this way, Shh peptides may mediate distinct signaling functions in the vertebrate embryo.
Example 7 Sonic hedgehog and Fgf-4 act through a signaling cascade and feedback loop to integrate growth and patterning of the developing limb bud Experimental Procedures Cloning of Chicken Fgf-4 and Bmp-2 A 246 bp fragment of the chicken Fgf-4 gene was cloned by PCR from a stage 22 chicken limb bud library. Degenerate primers were designed against previously cloned Fgf-4 and Fgf-6 genes: fgf5' (sense) AAA AGC TTT AYT GYT AYG TIG GIA THG G (SEQ ID No:38) and fgf3' (antisense) AAG AAT TCT AIG CRT TRT ART TRT TIG G (SEQ ID No:39). Denaturation was at 94°C for 2 min, followed by 30 cycles of 94°C for 30 sec, for 60 sec, and 72*C for 30 see, with a final extension at 72 0 C for 5 min. The PCR product was subcloned into the Bluescript SK+ vector. A clone was sequenced and confirmed as Fgf- 4 by comparison with previously published Fgf-4 genes and a chicken Fgf-4 gene sequence kindly provided by Lee Niswander.
BMP-related sequences were amplified from a stage 22 posterior limb bud cDNA library prepared in Bluescript using primers and conditions as described by Basler, et al.
(1993). Amplified DNAs were cloned and used to screen a stage 22 limb bud library prepared in X-Zap (Stratagene). Among the cDNAs isolated was chicken Bmp-2. Its identity 25 was confirmed by sequence comparison to the published clones (Francis, et al., (1994) Development 120:209-218) and by its expression patterns in chick embryos.
Chick Surgeries and Recombinant Retroviruses 30 All experimental manipulations were performed on White Leghorn chick embryos (S- SPF) provided by SPAFAS (Norwich, Conn). Eggs were staged according to Hamburger and Hamilton (1951) J. Exp. Morph. 88:49-92.
Viral supematants of Sonic/RCAS-A2 or a variant containing an influenza hemaglutinin epitope tag at the carboxyl terminus of the hedgehog protein (Sonic7. 1/RCAS- A2, functionally indistinguishable from Sonic/RCAS-A2), were prepared as described (Hughes, et al., (1987) J. Virol. 61:3004-13; Fekete and Cepko, (1993) Mol. Cell. Biol.
13:2604-13; Riddle, et al., (1993) Cell 75:1401-16). For focal injections the right wings of stage 18-21 embryos were transiently stained with nile blue sulfate (0.01 mg/ml in Ringer's solution) to reveal the AER. A trace amount of concentrated viral supernatant was injected beneath the AER.
The AER was removed using electrolytically sharpened tungsten wire needles. Some embryos had a heparin-acrylic bead soaked in FGF-4 solution (0.8 mg/ml; a gift from Genetics Institute) or PBS stapled to the limb bud with a piece of 0.025mm platinum wire (Goodfellow, Cambridge UK) essentially as described by Niswander et al, (1993) Cell 75:579-87.
Limbs which were infected with Sonic/RCAS virus after AER removal were infected over a large portion of the denuded mesoderm to ensure substantial infection. Those embryos which received both an Fgf-4 soaked bead and virus were infected only underneath the bead.
In Situ Hybridizations and Photography Single color whole mount in situ hybridizations were performed as described (Riddle, et al., (1993) Cell 75:1401-16). Two color whole mount in situ hybridizations were performed essentially as described by Jowett and Lettice (1994) Trends Genet. 10:73-74.
The second color detection was developed using 0.125mg/ml magenta-phos (Biosynth) as the 0 substrate. Radioactive in situ hybridizations on 5pm sections was performed essentially as 20 described by Tessarollo, et al. (1992) Development 115:11-20.
The following probes were used for whole mount and section in situ hybridizations: Sonic: 1.7kb fragment of pHH2 (Riddle, et al., (1993) Cell 75:1401-16). Bmp-2: 1.5 kb fragment encoding the entire open reading frame. Fgf-4: 250 bp fragment described above.
Hox d-11: a 600 bp fragment, Hoxd-13: 400 bp fragment both including 5' untranslated sequences and coding sequences upstream of the homeobox. RCAS: 900 bp SalI-Clal S fragment of RCAS (Hughes et al., (1987) J Virol. 61:3004-12).
(ii) Relationship of Sonic to Endogenous Bmp-2 and Hoxd Gene Expression 30 The best candidates for genes regulated by Sonic in vivo are the distal members of the Hoxd gene cluster, Hoxd-9 through -13, and Bmp-2. Therefore, the relationships of the S* expression domains of these genes in a staged series of normal chick embryos were analyzed.
Hoxd-9 and Hoxd-10 are expressed throughout the presumptive wing field at stage 16 (Hamburger and Hamilton, (1951) J. Exp. Morph. 88:49-92), prior to the first detectable expression of Sonic at early stage 18. Hoxd-11 expression is first detectable at early stage 18, the same time as Sonic, in a domain coextensive with Sonic. Expression of Hoxd-12 and Hoxd-13 commence shortly thereafter. These results suggest that Sonic might normally induce, directly or indirectly, the expression of only the latter three members of the cluster, even though all five are nested within the early limb bud.
As limb outgrowth proceeds Sonic expression remains at the posterior margin of the bud. In contrast the Hoxd gene expression domains, which are initially nested posteriorly around the Sonic domain, are very dynamic and lose their concentric character. By stage 23 the Hoxd-11 domain extends anteriorly and distally far beyond that of Sonic, while Hoxd-13 expression becomes biased distally and displaced from Sonic.
While it is not clear whether Bmp-2 is expressed before Sonic (see Francis et. al., (1994) Development 120:209-218) Bmp-2 is expressed in a mesodermal domain which apparently overlaps and surrounds that of Sonic at the earliest stages of Sonic expression. As the limb bud develops, the mesodermal expression of Bmp-2 remains near the posterior limb margin, centered around that of Sonic, but in a larger domain than Sonic. This correspondence between Sonic and Bmp-2 expression lasts until around stage 25, much longer than the correspondence between Sonic and Hoxd gene expression. After stage Bmp-2 expression shifts distally and is no longer centered on Sonic.
(iii) Relationship ofSonic to Induced Bmp-2 and Hoxd Gene Expression The fact that the expression domains of the Hoxd genes diverge over time from that of 20 Sonic hedgehog implies that Sonic does not directly regulate their later patterns of expression.
This does not preclude the possibility that the later expression domains are genetically downstream of Sonic. If this were the case, exogenously expressed Sonic would be expected to initiate a program of Hoxd gene expression which recapitulates that seen endogenously.
Therefore, the spatial distribution of Hoxd gene expression at various times following Sonic misexpression was compared. The anterior marginal mesoderm of early bud (Stage 18-20) wings was injected at a single point under the AER with a replication competent virus that expresses a chicken Sonic cDNA. Ectopic Sonic expressed by this protocol leads to both anterior mesodermal outgrowth and anterior extension of the AFR.
The Sonic and Hoxd gene expression domains in the infected limbs were analyzed in 30 sectioned and intact embryos. Viral Sonic message is first detected approximately 18 hours after infection at the anterior margin, at the same time as, and approximately coextensively with, induced Hoxd-11. This suggests that Sonic can rapidly induce Hoxd-11 expression and that the lag after injection represents the time required to achieve Sonic expression. By hours post infection distal outgrowth of infected cells combined with lateral viral spread 35 within the proliferating cells leads to viral expression in a wedge which is broadest at the distal margin and tapers proximally. By this time, Hoxd-11 expression has expanded both antero-proximally and distally with respect to the wedge of Sonic-expressing cells, into a domain which appears to mirror the more distal aspects of the endogenous Hoxd-11 domain.
n, g Weak Hoxd-13 expression is also detected at 35 hours in a subset of the Sonic expressing domain at its distal margin. 51 hours after infection the relationship of Sonic and Hoxd-11 expression is similar to that seen at 35 hours, while the induced Hoxd-13 expression has reached wild type levels restricted to the distal portions of the ectopic growth. Thus the ectopic Hoxd expression domains better reflect the endogenous patterns of expression than they do the region expressing Sonic. This suggests that there are multiple factors regulating Hoxd expression but their actions lie downstream of Sonic.
Since the endogenous Bmp-2 expression domain correlates well with that of Sonic, and Bmp-2 is induced by ZPA grafts, it was looked to see if Bmp-2 is also induced by Sonic.
Bmp-2 is normally expressed in two places in the early limb bud, in the posterior mesoderm and throughout the AER (Francis, et al., (1994) Development 120:209-218). In injected limb buds additional Bmp-2 expression is seen in both the anterior mesoderm and in the anteriorly extended AER. The domain of Bmp-2 expression is slightly more restricted than that of viral expression, suggesting a delay in Bmp-2 induction. Bmp-2 expression in both the mesoderm and ectoderm is thus a downstream target of Sonic activity in the mesoderm. In contrast to the expression domains of the Hoxd genes, the endogenous and ectopic Bmp-2 expression domains correlate well with that of Sonic. This suggests that Bmp-2 expression is regulated more directly by Sonic than is expression of the Hoxd genes.
20 (iv) The AER and Competence to Respond to Sonic Ectopic activation of Hoxd gene expression is biased distally in virally infected regions, suggesting that ectodermal factors, possibly from the AER, are required for Hoxd gene induction by Sonic. To test this, Sonic virus was injected into the proximal, medial 25 mesoderm of stage 21 limb buds, presumably beyond the influence of the AER. Although the level of Sonic expression was comparable to that observed in distal injections, proximal misexpression of Sonic did not result in ectopic induction of the Hoxd genes or Bmp-2, nor did it result in any obvious morphological effect (data not shown). The lack of gene induction following proximal misexpression of Sonic suggests that exposure to Sonic alone is insufficient to induce expression of these genes.
This was tested more rigorously by injection of Sonic virus into the anterior marginal mesoderm of stage 20/21 limb buds after the anterior half of the AER had been surgically removed. Embryos were allowed to develop for a further 36 to 48 hours before harvesting.
During this time the AER remaining on the posterior half of the limb bud promotes almost wild type outgrowth and patterning of the bud. Gene expression was monitored both in sectioned and intact embryos. In the presence of the AFR, Sonic induces both anterior mesodermal proliferation and expression of Hoxd-ll, Hoxd-13 and Bmp-2. In the absence of the overlying AER, Sonic does not induce either mesodermal proliferation or expression of /i/ these genes above background. Signals from the AER are thus required to allow both the proliferative and patterning effects of Sonic on the mesoderm.
Since application of FGF protein can rescue other functions of the AER such as promoting PD outgrowth and patterning, it was sought to determine whether FGFs might also promote mesodermal competence to respond to Sonic. FGF-4-soaked beads were stapled to AER-denuded anterior mesoderm which was infected with Sonic virus. Gene expression and mesodermal outgrowth were monitored as described previously. In the presence of both Sonic virus and FGF-4 protein, Hoxd-11, Hoxd-13 and Bmp-2 expression are all induced.
The expression levels of the induced genes are similar to or greater than the endogenous expression levels, and are equivalent in magnitude to their induction in the presence of the AER. Thus Fgf-4 can induce the competence of the mesoderm to respond to Sonic.
Sonic alone is insufficient to induce either gene expression or mesodermal proliferation in the absence of the AER, while the combination of Sonic and FGF-4 induces both proliferation and gene expression. It was than asked whether FGF-4 alone has any effect on gene induction or mesodermal proliferation. Application of FGF-4 in the absence of Sonic virus does not induce Hoxd or Bmp-2 gene expression above control levels, however FGF-4 alone induces mesodermal outgrowth. These results suggest that mesodermal gene activation requires direct action of Sonic on the mesoderm and that proliferative response to Sonic is indirec:t, due to the induction of FGFs.
Sonic Induces Polarized Fgf-4 Expression in the AER gf-4 is expressed in a graded fashion in the AER of the mouse limb bud, with maximal expression at the posterior region of the AER tapering to undetectable levels in the 25 anterior ridge (Niswander and Martin, (1992) Development 114:755-68). Therefore, it was appropriate to investigate whether Fgf-4 is asymmetrically expressed in the chick AER, and whether its expression is induced by Sonic. A fragment of the chicken Fgf-4 gene was cloned from a stage 22 chicken limb library by PCR using degenerate primers designed from mouse Fgf-4 and Xenopus e-Fgf sequence; based on information provided by L. Niswander and G.
Martin. Assignment of gene identity was based on primary sequence as well as comparison of expression patterns with that of murine Fgf-4 (Niswander and Martin, (1992) Development S 114:755-68). Whole mount in situ hybridization analysis showed strong limb expression of chick Fgf-4 in the AER. Fgf-4, like Bmp-2, is expressed all the way to the posterior border of the AER, but its anterior domain ends before the morphological end of the AER creating a 35 posterior bias that has also been observed by Niswander et al., (1994) Nature (in press).
Expression is first detected in the distal AER at about stage 18. As outgrowth proceeds the posterior bias develops. Expression peaks around stage 24/25 and then fades by stage 28/29.
/la The expression domain of Fgf-4 becomes posteriorly biased as Sonic is expressed in the posterior mesoderm. This observation is consistent with Sonic influencing the expression of Fgf-4 in the posterior AER. To test the effect of Sonic on Fgf-4 expression in the AER, stage 18-20 embryos were infected with Sonic virus in a single point at their anterior margin beyond the anterior limit of the AER. The embryos were harvested one to two days later, when an extension of the anterior AER became apparent. The expression of Fgf-4 was analyzed by in situ hybridization Fgf-4 expression is induced in the anteriormost segment of the AER, in a region which is discontinuous with the endogenous expression domain, and overlies the domain of viral Sonic infection. This result contrasts with the Bmp-2 expression induced in the extended AER, which is always continuous with the endogenous expression domain. The asymmetry of the induced Fgf-4 expression indicates that Sonic polarizes the extended AER, much as a ZPA graft does (Maccabe and Parker, (1979) J. Embryol. Exp.
Morph. 53:67-73). Since FGFs by themselves are mitogenic for limb mesoderm, these results are most consistent with Sonic inducing distal proliferation indirectly, through the induction ofmitogens in the overlying AER.
(vi) Reciprocal Regulation of Sonic by Fgf-4 Sonic thus appears to be upstream of Fgf-4 expression in the AER. However, since 20 the AER is required to maintain polarizing activity in the posterior mesoderm (Vogel and Tickle, (1993) Development 19:199-206; Niswander et al., (1993) Cell 75:579-87), Sonic may also be downstream of the AER. If Sonic is regulated by the AER and the AER by Sonic, this would imply that they are reinforcing one another through a positive feedback loop.
25 To test whether the AER dependence of ZPA activity is controlled at the level of transcription of the Sonic gene, Sonic expression following removal of the AER from the posterior half of the limb bud was assayed. Sonic expression is reduced in an operated limb compared to the contralateral control limb within ten hours of AER removal, indicating that Sonic expression is indeed AER dependent. The dependence of Sonic expression on signals from the AER suggests that one of the functions of the AER is to constrain Sonic expression to the more distal regions of the posterior mesoderm.
In addition to their mitogenic and competence-inducing properties, FGFs can also substitute for the AER to maintain the ZPA. In order to test whether FGFs can support the expression of Sonic, beads soaked in FGF-4 protein were stapled to the posterior-distal tips of limb buds after posterior AER removal. Embryos were assayed for Sonic expression approximately 24 hours later, when Sonic expression is greatly reduced in operated limb buds which had not received an FGF-4 bead. Strong Sonic expression is detectable in the posterior mesoderm, slightly proximal to the bead implant, and reflecting the normal domain of Sonic expression seen in the contralateral limb. With the finding that FGF-4 can maintain Sonic expression, the elements required for a positive feedback loop between Sonic expression in the posterior mesoderm and Fgf-4 expression in the posterior AER are established (see also Niswander et al. (1994) Nature (in press)).
The induction of Bmp-2 expression by Sonic requires signals from the AER, and its domain correlates over time with that of Sonic. Therefore, it was interesting to learn if the continued expression of Bmp-2 also requires signals from the AER, and if so, whether they could be replaced by FGF-4. To test this, Bmp-2 expression following posterior AER removal, and following its substitution with an FGF-4 bead was assayed. Bmp-2 expression fades within hours of AER removal, and can be rescued by FGF-4. These data indicate that the maintenance of Bmp-2 expression in the posterior mesoderm, like that of Sonic, is dependent on signals from the AER, which are likely to be FGFs.
(vii) The Mesodermal Response to Sonic It has been found that only mesoderm underlying the AER is responsive to Sonic, apparently because the AER is required to provide competence signals to the limb mesoderm.
Fgf-4, which is expressed in the AER, car substitute for th- AER in this regard, and thus might act in combination with Sonic to promote Hoxd and Bmp-2 gene expression in the mesoderm. FGFs may be permissive factors in a number of instructive pathways, as they are 20 also required for activins to pattern Xenopus axial mesoderm (Cornell and Kimelman, (1994) Development 120:2187-2198; LaBonne and Whitmnan, (1994) Development 120:463-472).
The induction of Hoxd and Bmp-2 expression in response to Sonic and FGF-4 in the absence of an AER suggests that the mesoderm is a direct target tissue of Sonic protein.
Since Sonic can induce Fgf-4 expression in the AER, it follows that Sonic also acts indirectly 25 on the mesoderm through the induction of competence factors in the AER.
(viii) Downstream Targets and a Cascade of Signals Induced by Sonic The five AbdB-like Hoxd genes, Hoxd-9 through -13, are initially expressed in a i nested pattem centered on the posterior of the limb bud, a pattern which suggests they might be controlled by a common mechanism (Dolle, et al., (1989) Cell 75:431-441; Izpisua- Belmonte, et al., (1991) Nature 350:585-9). The analysis of the endogenous and induced domains of Hoxd gene expression suggests that Sonic normally induces expression of Hoxd- 11, -12 and -13. In contrast it was found that Hoxd-9 and -10 expression initiate before Sonic mRNA is detectable. This implies that at least two distinct mechanisms control the initiation of Hoxd gene expression in the wing bud, only one of which is dependent on Sonic.
Several observations suggest that the elaboration of the Hoxd expression domains is not controlled directly by Sonic, but rather by signals which are downstream of Sonic. The Hoxd expression domains rapidly diverge from Sonic, and evolve into several distinct /22 subdomains. Moreover these subdomains appear to be separately regulated, as analysis of the murine Hoxd-11 gene promoter suggests that it contains independent posterior and distal elements (Gerard, et al., (1993) Embo. J. 12:3539-50). In addition, although initiation of Hoxd-11 through -13 gene expression is dependent on the AER, their expression is maintained following AER removal (Izpisua-Belmonte, et al., (1992) Embo. J. 11:1451-7).
As Sonic expression fades rapidly under similar conditions, this implies that maintenance of Hoxd gene expression is independent of Sonic. Since ectopic Sonic can induce a recapitulation of the Hoxd expression domains in the limb, it can be concluded that although indirect effectors appear to regulate the proper patterning of the Hoxd expression domains, they are downstream of Sonic. Potential mediators of these indirect effects include Bmp-2 in the mesoderm and Fgf-4 from the AER.
In contrast to the Hoxd genes, Bmp-2 gene expression in the posterior limb mesoderm appears to be continually regulated by Sonic. It was found that both endogenous and ectooic Bmp-2 expression correspond to that of Sonic. Furthermore, continued Bmp-2 expression is dependent on the AER and can be rescued by FGF-4. It is likely that this is an indirect Sconsequence of the fact that Sonic expres:ion is also maintained by the AER and can be rescued by FGF-4., In fact, Bmp-2 expression might be a direct response of cells to secreted Sonic protein. The differences between Bmp-2 and Hoxd gene expression suggest that multiple pathways downstream of Sonic regulate gene expression in the mesoderm.
20 Bmp-2 itself is a candidate for a secondary signaling molecule in the cascade of patterning events induced by Sonic. Bmp-2 is a secreted molecule of the TGF-P family and it expression can be induced by Sonic. This appears to be an evolutionarily conserved pathway, as Dros-HH, the Drosophila homolog of Sonic, activates the expression of dpp, the homolog of Bmp-2, in the eye and wing imaginal discs (Heberlein, et al., (1993) Cell 75:913- 25 26; Ma, et al., (1993) Cell 75:927-38; Tabata and Korberg, (1994) Cell 76:89-102).
Expression of Dros-HH is normally confined to the posterior of the wing disc. Ectopic expression of Dros-HH in the anterior of the disc results in ectopic expression of dpp and ultimately in the duplication of wing structure with mirror image symmetry (Bassler and Struhl, (1994) Nature 368:208-214). This effect is strikingly parallel to the phenotypic results of ectopic expression of Sonic in the chick limb.
(ix) Regulation of Sonic Expression Sonic expression is activated in the posterior of the limb bud very early during mesodermal outgrowth (Riddle et al., (1993) Cell 75:1401-16). The factors which initiate this localized expression are not yet identified but ectopic expression of Hoxb-8 at the anterior margin of the mouse limb bud results in the activation of a second domain of Sonic expression under the anterior AER (Charit6 el al., (1994) Cell 78:589-601). Since retinoic acid is known to be able to induce the expression of Hoxb-8 and other Hox genes in vitro i23 (Mavilio et al., (1988) Differentiation 37:73-79) it is possible that endogenous retinoic acid acts to make cells competent to express Sonic by inducing expression of upstream Hox genes, either in the very early limb bud or in the flank prior to the limb bud formation.
Several lines of evidence suggest that once induced Sonic expression is dependent on signals from the posterior AER. Following its initiation in the posterior limb mesoderm, the Sonic expression domain moves distally as the limb bud grows out, always remaining subjacent to the AER. Similarly, Sonic expression can also be induced on the anterior margin of the limb bud by implantation of a retinoic acid bead, but the induced ectopic expression is limited to the mesoderm directly underlying the AER (Riddle, et al., (1993) Cell 75:1401-16).
In addition, ZPA activity fades rapidly following removal of the AER (Niswander, et al., (1993) Cell 75:579-87; Vogel and Tickle, (1993) Development 119:199-206), and ZPA grafts only function when placed in close proximity to the AER (Tabin, (1991) Cell 66:199-217; Tickle, (1991) Development Supp. 1:113-21). The observation that continued Sonic expression depends on signals from th- posterior AER reveals the mechanism underlying these observations.
The reliance of Sonic expression on AER-derived signals suggests an explanation for the distal shift in Sonic expression duing limb development (Riddle et al., (1993) Cell 75:1401-16). Signals from the AER also promote distal outgrowth of the mesodermal cells of the progress :one, which in turn results in the distal displacement of the AER. Hence, as 0 maintenance of Sonic expression requires signals from the AER, its expression domain will be similarly displaced.
It was found that replacement of the AER with FGF-4 soaked beads results in the maintenance of Sonic expression. This result is consistent with the previous findings that ZPA activity can be maintained in vivo and in vitro by members of the FGF family 5 (Anderson, et al., (1993) Development 117:1421-33; Niswander et al., (1993) Cell 75:1401- 16 Vogel and Tickle, (1993) Development 119:199-206). Since Fgf-4 is normally expressed in the posterior AER, these results suggest that Fgf-4 is the signal from the ectoderm involved in maintaining Sonic expression.
O 30 Sonic and Regulation and Maintenance of the AER Sonic can induce anterior extensions of the AER which have an inverted polarity relative to the endogenous AER. This polarity is demonstrated by examining the expression of two markers in the AER. In normal limbs Bmp-2 is expressed throughout the AER, while Fgf-4 is expressed in the posterior two thirds of the AER. In the extended AER resulting 35 from ectopic Sonic expression, Bmp-2 is again found throughout the AER, while Fgf-4 expression is biphasic, found at either end of the AER, overlying the anterior and posterior mesodernnal domains expressing Sonic. These results are consistent with previous observations that antero-posterior polarity of the AER appears to be regulated by the la q underlying mesoderm, and that ZPA grafts lead to the induction of ectopic, polarized AER tissue (Maccabe and Parker, (1979) J. Embryol. Exp. Morph. 53:67-73). Our results also suggest that the normal AP polarity of the AER is a reflection of endogenous Sonic expression. The induced AER is sufficient to promote complete PD outgrowth of the induced structures (Riddle et al., (1993) Cell 75:1401-16). Hence whatever factors are necessary to maintain the AER are also downstream of Sonic.
(xi) A Positive Feedback Loop Between Sonic and Fgf-4 The induction of Fgf-4 expression by Sonic in the ectopic AER, and the maintenance of Sonic. expression by FGF-4 suggest that Sonic and Fgf-4 expression are normally sustained by a positive feedback loop. Sucli a feedback loop would allow the coordination of mesodermal outgrowth and patterning. This coordination is possible because Sonic patterns mesodermal tissue and regulates Fgf-' expression, while FGF-4 protein induces mesodermal proliferation and maintains Sonic expression. Moreover mesodermal tissue can only be patterned by Sonic in the context rf a competence activity provided by F8f4. Thus patterning is always coincident with proliferation.
It remaias possible that exogenously applied Fgf-4 might be mimicking the activity of a different memnber of the FGF farmily. For example, Fgf-2 is expressed in'the limb 20 mesoderm and the AER (Savage et al,, (1993) Development Dynamics 198:159-70) and has similar effects on limb tissue as Fgf.-4 (Niswander and Martin, (1993) Nature 361:68-71; Niswander, et al., (1993) Cell 75:579-87; Riley, et al., (1993) Development 118:95-104; Fallon, et al., (1994) Science 264:104-7).
25 (xii) Coordinated Regulation of Limb Outgrowth and Patterning Patterning and outgrowth of tle developing limb are known to be regulated by two S major signaling centers, the ZPA and AER. The identification of Sonic and FGFs as molecular mediators of the activities of the ZPA and AER has allowed for dissociation of the 30 activities of these signaling centers from their regulation, and investigation of the signaling pathways through which they function.
The results presented above suggest that the ability of cells to respond to Sonic protein is dependent on FGFs produced by the AER. It was also found that Sonic induces a cascade of secondary signals involved in regulating mesodermal gene expression patterns. In addition evidence was found for a positive feedback loop initiated by Sonic, which maintains expression of Sonic in the posterior mesoderm and Fgf-4 in the AER. The feedback loop described suggests a mechanism whereby outgrowth and patterning along the AP and PD axes of the limb can be coordinately regulated.
/23 The results described above further suggest that Sonic acts as a short range signal which triggers a cascade of secondary signals whose interplay determines the resultant pattern of structures. The data suggest a number of inductive pathways that can be combined to generate a model (Figure 14) which describes how Sonic, in coordination with the AER, acts to pattern mesodermal tissues along the anterior-posterior limb axis, while simultaneously regulating proximal-distal outgrowth.
Following its induction, Sonic signals to both the limb ectoderm and mesoderm.
Sonic imposes a distinct polarity on the forming AER, including the posteriorly biased expression of Fgf-4, and the AER becomes dependent on continued Sonic expression. The mesoderm, as long as it is receiving permissive signals from the overlying ectoderm, responds to the Sonic signal by expreF sing secondary signaling molecules such as BIrp-2 and by activating Hoxd genes. Bmp-2 expression is directly dependent on continued Sonic S expression, while the continued expresskin of the Hoxd genes, rapidly becomes Sonic.
independent. In a reciprocal fashion, maintenance of Sonic hedgehog expression in the posterior mesoderm becomes dependent :n signals from the AER. Since the factors expressed by the AER are not only:. equired for the maintenance of Sonic expression and activity,.-but are also mitogenic, jrowt: and patterning become inextricably linked.
Coordination of limb development through interdependent signaling centers forces the AP and PD structures to be induced and. .atterned in tandem. The pathways elucidated herein thus provide a molecular framework for the controls governing limb patterning Example 8 Sonic, BMP-4, and Hox Gene Expre'sion Suggest a Conserved Pathway in Patterning the Vertebrate and Drosophila Gut Experimental Procedure In Situ Hybridization and Photography 30 BMP probes were isolated usinig primers designed to amplify members of thi; TGF- "i and BMP families (Basler, K. et al., (1993) Cell 73:687-702, eight independent 120.bp BMP fragments were amplified from a stage 22 chicken posterior limb bud plasmid cDNA library.
These fragments were pooled and used to screen an unamplified stage 22 limb bud lambda zap cDNA library constructed as in Riddle et al., (1993) Cell 75:1401-16. Among the BMP related clones isolated were an approximately 1.9 kb cDNA clone corresponding to chicken BMP-2 and an approximately 1.5 kb cDNA clone corresponding to chicken BMP-4. Both clones contain the entire coding regions. The Sonic clone was obtained as described in Riddle et al, (1993) Cell 75:1401-16. Digoxigenin-UTP labeled RNA probes were transcribed as per Riddle et al., (1993) Cell 75:1401-16. Briefly, harvested chick embryos were fixed overnight in 4% paraformaldehyde, washed in PBS then processed for whole mount in situ hybridization methods are per Riddle et al., (1993)Cell 75:1401-16. Embryos were photographed from either ventral or dorsal surfaces under transmitted light using a Nikon zoom stereo microscope with Kodak Ektar 100 ASA film. Whole mount in situ hybridization embryos and viscera were processed for sectioning as described in Riddle et al., (1993)Cell 75:1401-16. 15-25 .m transverse sections were air dried and photographed with brightfield or numarski optics using a Zeiss Axiophot microscope and Kodak Ektar 25 ASA film.
Chick Embryos and Recombinant Retroviruses A retroviral vector engineered to express a full length cDNA of chicken Sonic, as in Riddle et al. (1993) Cell 75:1401-16, was injected unilaterally into stage 8-13 chicken embryos targeting the definitive endoderm at the mid-embryo level. At this stage the CIP has not formed and neither Sonic nor BMP-4 are expressed in the region injected. Injections were performed on the ventral surface on embryos cultured with their ventral surface facing up (New, D.A.T. (1955) Embryol. Exp. Morph. 3:320-31. Embryos were harvested 18-28 hours after injection and prepared for who'b mount in situ hybridization (see above description of in 20 situ experiment), hybridized with Sonic or BMP-4 digoxigenin labeled probes.
In Situ Hybridization with Hox Genes Cloned cDNA of the chicken homologues of Hoxa-9,-10,-11,-13; b-9, c-9,-10,-11; d- 25 9,-10,-11,-12,and -13 were used to transcribe digoxigenen-UTP labeled riboprobes for whole mount in situ hybridization. Domestic chick embryos were harvested into PBS and eviscerated. The visceral organ block was fixed in 4% paraformaldehyde overnight and processed for whole mount in situ hybridization. Methods and photographic technique as described above.
(ii) Expression of Sonic and BMP-4 in Stage 13 Chick Embryos Determined by Whole Mount In Situ Hybridization Chick gut morphogenesis begins at stage 8 (Hamberger and Hamilton, (1987) Nutr.
6:14-23 with a ventral in-folding of the anterior definitive endoderm to form the anterior intestinal portal (AIP) (Romanoff, (1960) The Avian Embryo, The Macmillan Co., NY.
This lengthens posteriorly forming the foregut. A second wave of endodermal invagination is initiated posteriorly at stage 13, creating the caudal intestinal portal (CIP). The CIP extends /27 anteriorly forming the hindgut. Sonic expression, previously noted in the endoderm of the vertebrate gut (Riddle et al., (1993) Cell 75:1401-16; Echelard et al., (1993) Cell 75:1417- 1430), is expressed early in a restricted pattern in the endodermal lips of the AIP and CIP.
Sonic expression is detected in the endoderm of the AIP and CIP in pre gut closure stages. At later stages, stage 28 embryos, Sonic is expressed in the gut in all levels (fore-, mid-, and hind-gut) restricted to the endoderm. Sonic is known to be an important inductive signal in other regions of the embryo including the limb bud (Riddle et al., (1993) Cell 75:1401-16) and neural tube (Echelard et al., (1993) Cell 75:1417-1430; Kraus et al., (1994) Cell 75:1437- 1444; Roelink et al., (1994) Cell 76:761-775). Since primitive gut endoderm is known to cause gut-specific mesodermal differentiation when combined with non-gut mesenchyme (Haffen et al., (1987) Nutr. 6:14-23), we speculated that Sonic might function as an inductive signal to the visceral mesoderm. A potential target gene for the action of Sonic was suggested by analogy to the Drosophila imaginal discs where, Dros-HH, the homologue of vertebrate Sonic, activates the expression of the TGF-P related gene dpp in adjacent cells (Tabata abd Komberg, (1994) Cell 76:89-102; Heberlein et al., (1993) Cell 75:913-926; Ma et al., (1993) Cell 75:913-926; Basler et al., (1993) Cell 73:687-702). There are two vertebrate homologues of dpp, BMP-2 and BMP-4. The earliest detectable expression of BMP-4 occurs simultaneously with the first observable expression of Sonic in the developing gut. BMP-4 is expressed in a domain abutting Sonic at the AIP and the CIP, but is restricted 20 to the adjacent ventral mesoderm. BMP-4 gut expression persists into later stage embryos, stage 33 embryos, in the visceral mesoderm only. The tissue restricted expression of both genes is maintained in all stages studied. BMP-2 is not expressed in the gut at the AIP or CIP, but is expressed in clusters of cells in the gut mesoderm in later stages, a pattern distinct from that of BMP-4.
9 (iii) Ectopic Expression of Sonic Induces Ectopic Expression of BMP-4 in Mesodermal Tissues of the Developing Chick 9 To test whether Sonic is capable of inducing BMP-4 in the mesoderm we an ectopic expression system previously used to study the role of Sonic in limb development was utilized (Riddle et al., (1993) Cell 75:1401-16). A replication competent retrovirus engineered to express Sonic was injected unilaterally into the presumptive endoderm and visceral mesoderm at mid-embryo positions in stage 8-13 chick embryos in vitro (New, D.A.T. (1955) Embryol. Exp. Morph. 3:320-321). When embryos were examined by in situ hybridization 18-26 hours later, the normal wild type expression of Sonic is detected at the AIP, CIP, and in the midline (neural tube and notochord). Ectopic Sonic expression is present unilaterally on the left ventral surface. Also, wild type Sonic expression is seen in the floor plate of the neural tube and notochord. Ectopic expression is seen unilaterally in the /11 visceral endoderm, its underlying splanchnic mesoderm, and somatic mesoderm. BMP-4 expression can be seen induced in the mesoderm at the site of injection, in addition to its normal expression in the mesoderm of the CIP. Wild type BMP-4 expression is seen in the most dorsal aspects of the neural tube and symmetrical lateral regions adjacent to the neural tube. Induced BMP-4 expression is present unilaterally in the splanchnic mesoderm at the site of Sonic viral injection, and not in the visceral endodermn.
Since BMP-4 is, itself, a secreted protein, it could function as a secondary signal in an inductive cascade, similar to the signal cascades from Dros-HH to dpp in Drosophila imaginal discs (Tabata abd Kornberg, (1994) Cell 76:89-102; Heberlein et al., (1993) Cell 75:913-926; Ma et al., (1993) Cell 75:913-926; Basler et al., (1993) Cell 73:687-702) and from Sonic to BMP-2 in the limb bud. In the gut, BMP-4 could act as a secondary signal either as part of a feedback loop to the endoderm or within the visceral mesoderm. This latter possibility is consistent with the finding that in mice homozygous for a deletion in the BMP-4 gene, the ventral mesoderm fails to close.
(iv) Expression ofHox Genes in the Developing Chick Gut There is a striking parallel between the apparent role of Sonic as an endoderm-tomesoderm signal in early vertebrate gut morphogenesis and that of its Drosophila 20 homologue, Dros-HH. Dros-HH like Sonic) is expressed in the Drosophila gut endoderm from the earliest stages of morphogenesis (Taylor et al., (1993) Mech. Dev. 42:89-96). Its putative receptor, patched, is found in the visceral mesoderm implicating Dros-HH (like Sonic) in endodermal-mesodermal inductive interactions. This led to consideration whether other genes known to be involved in regulating Drosophila gut development might also play a role in regulating chick gut morphogenesis. Regionally specific pattern in Drosophila gut endoderm is regulated by a pathway involving restricted expression of homeotic genes in the mesoderm (McGinnis and Krumlauf, (1992) Cell 68:283-302). Although the basis for S patterning the vertebrate gut is poorly understood, in several other regions of the embryo Hox genes have been implicated as key regulators of patterns. Vertebrate Hox genes are expressed in overlapping anteroposterior domains which correlate with structural boundaries in the developing hindbrain, vertebrae, and limbs (McGinnis and Krumlauf, (1992) Cell 68:283- 302). Whole mount in situ hybridization was used to test whether these genes are also expressed in the developing vertebrate hindgut and whether their domains of expression correlate with morphologic borders of the chick gut.
Lumenal gut differentiation creates three morphologically and physiologically distinct regions: fore-, mid-, and hind- gut. The fore-gut and hind-gut are the derivatives of the primitive gut tubes initiated at the AIP and CIP respectively. Ultimately these tubes meet and fuse at the yolk stalk around stage 24-28. The midgut is formed from both foregut and hindgut primordia, just anterior and posterior to the yolk stalk.
The most posterior derivative of the hindgut is the cloaca, the common gut-urogenital opening. The rest of the hindgut develops into the large intestine. The midgut/hindgut border is demarcated by a paired tubal structure, the ceca (analogous to the mammalian appendix), which forms as budding expansions at the midgut/hindgut border at stage 19-20.
Anterior to the ceca, the midgut forms the small intestine.
The expression pattern of the 5' members of the Hox gene clusters in the chick hindgut by whole mount in situ hybridization was studied. Hox gene expression patterns in the gut are dynamic. They are initially expressed (by stage 10) in broad mesodermal domains extending anteriorly and laterally. Later they become restricted. By stage 25, the Abd-B like genes of the Hoxa and Hoxd cluster are regionally restricted in their expression in hindgut mesoderm. The most anteriorly expressed gene, Hoxa-9, has an anterior border of expression within the mesoderm of the distal midgut (to a point approximating the distal third of the midgut length). Each successive gene within the A and D Hox clusters has a more posterior domain of expression. Hoxa-10, Hoxd-9 and Hoxd-10 are restricted in their expression to the ceca. Hoxa-ll and Hoxd-11 have an anterior limit of expression in the mid-ceca at the approximate midgut/hindgut boundary (Romanoff, A.L. (1960) The Avian Embryo, The Macmillan Co. NY). Hoxd-12 has an anterior limit at the posterior border of the ceca and 20 extends posteriorly throughout the hindgut to the cloaca. Hoxa-13 and Hoxd-13 are expressed in the most posteriorly restricted domain, in the ventral mesoderm surrounding the cloaca. Hoxa-13 and Hoxd-13 are the only Abd-B like genes which are also expressed within the gut endoderm, from the ceca to the cloaca.
The only member of the B or C Hox clusters which we found to be expressed in the 25 hindgut is Hoxc-9. The expression of Hoxc-9 overlaps with its paralogues Hoxa-9 and Hoxd- 9 in the midgut mesoderm, but has a sharp posterior boundary, complementary to Hoxa-11 and Hoxd- 11 in the mid-ceca.
The restricted expression of the Abd-B like Hox genes appear to demarcate the successive regions of the gut which will form the cloaca, the large intestine, the ceca, the mid-ceca at the midgut/hindgut border, and the lower portion of the midgut (perhaps identifying that portion of the midgut derived from the posterior gut tube3). Moreover, these molecular events presage regional distinctions. Expression of all Hox genes could be detected by stage 14, well before the hindgut lumen is closed (by stage 28) and is maintained in subsequent stages studied. Cytodifferentiation of the hindgut mesoderm and epithelium begins later, at stages 29-31 (Romanoff, A.L. (1960) The Avian Embryo, The Macmillan Co.
NY).
These results suggest that specific Hox genes might be responsible for regulating morphogenesis of the gut. Consistent with this, there is an apparent homeotic alteration in 134 the gut of a transgenic mouse in which the anterior limit of expression of Hoxc-8 is shifted rostrally: a portion of foregut epithelium mis-differentiates as midgut (Pollock and Bieberich, (1992) Cell 71:911-923).
Conservation in the Expression of Regulatory Genes Involved in the Formation of Vertebrate and Drosophila Gut There is an intriguing parallel between the expression patterns of Sonic, BMP-4, and the Hox genes in the vertebrate gut and those of their homologues during Drosophila gut morphogenesis (Figure 15). This conservation is of particular interest because in Drosophila the role played by these genes has been clarified genetically. Dros-HH (like its vertebrate homologue, Sonic) is expressed at the earliest stages in the gut endoderm and may be a signal to visceral mesoderm (Taylor et al., (1993) Mech. Dev. 42:89-96). Nothing is known directly of the relationship between Dros-HH expression and activation of expression of other genes in the Drosophila gut. However, in Drosophila imaginal discs, Dros-HH is known to activate the expression of dpp in a signaling cascade (Kraus et al., (1994) Cell 75:1437-1444; Heberlein et al., (1993) Cell 75:913-926; Ma et al., (1993) Cell 75:913-926; Basler et al., (1993) Cell 73:687-702). Later in gut development, the production of dpp in the mesoderm contributes to the regulation of the expression of homeotic genes in both the mesoderm and 20 the endoderm (Bienz, M. (1994) TIG 10:22-26). Drosophila homeotic genes are expressed in the gut visceral mesoderm and their expression is known to determine the morphologic borders of the midgut. This involves proper induction of gene expression in the adjacent endoderm, one of the mediators of the interaction is dpp (Bienz, M. (1994) TIG 10:22-26). If Dros-HH is required for the ultimate activation of the homeotic genes in the Drosophila 25 midgut, this would parallel the situation in the vertebrate limb bud where Sonic functions as an upstream activator of the Hox genes (Riddle et al., (1993) Cell 75:1401-1416), perhaps in a signaling cascade involving BMP-2.
The extraordinary conservation in the expression of regulatory genes in the vertebrate and Drosophila gut strongly suggests a conservation of patterning mechanisms. Pathways established by genetic studies in Drosophila provide direct insights into the molecular basis for the regionalization and morphogenesis of the vertebrate gut.
Example 9 Bacterially Expressed Hedgehog Proteins Retain Motorneuron-inducing Activity Various fragments of the mouse Shh gene were cloned into the pET1 D vector as fusion proteins with a poly(His) leader sequence to facilitate purification. Briefly, fusion genes encoding the mature M-Shh protein (corresponding to Cys-25 through Ser-437 of SEQ ID No. 11) or N-terminal containing fragments, and an N-terminal exogenous leader having 131 the sequence M-G-S-S-H-H-H-H-H-H -L-V-P-R-G-S-H-M (SEQ ID No: 47) were cloned in pETI ID and introduced into E. coli. The poly(His)-Shh fusion proteins were purified using nickel chclate chromatography according to me vendor's instructions (Qiagen catalog 30210), and the poly(His) leader cleaved from the purified proteins by treatment with thrombin.
Preparations of the purified Shh proteins were added to tissue explants (neural tube) obtained from chicken embryos and cultured in a defined media no serum). M-Shh protein was added to final concentrations of between 0.5pM to 5nM, and differentiation of the embryonic explant tissue to moiomeuron phenotype was detected by expression of Islet-1 antigen. The bacterially produced protein was demonstrated to be active in the explant cultures at concentrations as low as 5 to 50pM. An Shh polypeptide containing all 19kd of the amino terminal fragment and approximately 9kd of the carboxyl terminal fragment (see Example 6) displayed both motor neuron inducing activity and weak floor plate inducing activity, indicating that these activities likely reside with the N-temminal fragment.
All of the above-cited references and publications arc hereby incorporated by reference.
Equivalents Those skilled in the art will recognize, or be able to ascertain using no more than S2 routine experimentation, numerous equivalents to the specific polypeptides, nucleic acids, S 20 methods, assays and reagents described herein. Such equivalents are considered to be within the scope of this invention and arc covered by the following claims.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
S"i The reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that that prior art forms part of the common general knowledge in Australia.
/32 SEQUENCE LISTING GENERAL INFORMATION:
APPLICANT:
NAME: President and Fellows of Harvard College STREET: 124 Mt. Auburn Street CITY: Cambridge STATE: MA COUNTRY: USA POSTAL CODE (ZIP): 02138 NAME: Imperial Cancer Research Technology Ltd.
STREET: Sardinia Street CITY: London STATE: N/A COUNTRY: United Kingdom POSTAL CODE (ZIP): WC2A3NL (ii) TITLE OF INVENTION: Vertebrate Embryonic Pattern-Inducing Proteins and Uses Related Thereto (iii) NUMBER OF SEQUENCES: 47 (iv) COMPUTER READABLE FORM: MEDIUM TYPE: Floppy disk COMPUTER: IBM PC compatible OPERATING SYSTEM: PC-DOS/MS-DOS 30
SOFTWARE:
*o 0 CURRENT APPLICATION DATA: APPLICATION NUMBER: (vi) PRIOR APPLICATION DATA: APPLICATION NUMBER: US 08/176,427 FILING DATE: 30-DEC-1993 (vi) PRIOR APPLICATION DATA: S(A) APPLICATION NUMBER: US 08/356,060 FILING DATE: 14-DEC-1994 (viii) ATTORNEY/AGENT INFORMATION: too**: NAME: Vincent, Matthew P.
REGISTRATION NUMBER: 36,709 S REFERENCE/DOCKET NUMBER: HMI-006PC (ix) TELECOMMUNICATION INFORMATION: TELEPHONE: (617) 227-7400 TELEFAX: (617) 227-5941 INFORMATION FOR SEQ ID NO:1: SEQUENCE CHARACTERISTICS: LENGTH: 1278 base pairs TYPE: nucleic acid STRANDEDNESS: both TOPOLOGY: linear 133 (ii) MOLECULE TYPE: CDNA (ix) FEATURE: NAME/KEY: CDS LOCATION: 1. .1277 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:i: ATG GTC GAA ATG CTG CTG TTG ACA AGA ATT CTC TTG GTG GGC TTC ATC Met Val Giu Met Leu Leu Leu Thr Arg Ile Leu Leu Val Gly Phe Ile TGC OCT CTT TTA GTC TCC Cys Ala Leu Leu Val Ser TCT GGG CTG ACT TGT GGA CCA GGC AGG GGC Ser Giy Leu 25 Thr CYS Gly Pro Gly Arg Gly ATT GOA AAA AGG Ile Gly Lys Arg AGG CAC CCC Arg His Pro AAA AAG CTG ACC Lys Lys Leu Thr 40 GAG AAG ACC CTA Oiu Lys Thr Leu
CCG
Pro
GGG
Gly TTA GCC TAT AAG Leu Ala Tyr Lys GCC AG? GGA AGA Ala Ser Gly Arg CAG TTT Gin Phe s0 ATT CCC AAT GTG Ile Pro Asn Val 192
TAT
Tyr GAA GGG AAG ATC Glu Gly Lys Ile
ACA
Thr 70 AGA AAC TCC GAG Arg Asn Ser Giu
AGA
Arg 75 TTT AAA t73,A CTA Phe Lys Glu Leu
ACC
Thr CCA AAT TAC AAC Pro Ann Tyr Asn
CCT
Pro as GAC ATT ATT TTT Asp Ile Ile Phe GAT GAA GAG AAC Asp Glu Giu Asn ACG GGA Thr Gly 240 288 336 OCT GAC AGA A-1a Asp Arg GCG ATC TCG Ala Ile Ser 215
CTG
Leu 100 ATG ACT CAG CGC Met Thr Gin Arg AAG GAC AAG CTG AAT GCC CTG Lys Asp Lys Leu Asn Ala Leu GTO ATG AAC CGO Val Met Asn Gin
TG
Trp 120 CCC 000 GTG AAG Pro Oly Val Lys
CTO
Leu 125 COG GTG ACC Arg Vai Thr GAG GGC GJlu Gly 130 TOG GAC GAG GAT Trp Asp Oiu Asp
GC
Giy 135 CAT CAC TCC GAG His His Ser Glu
GAA
Oiu 140 TCG CTG CAC TAC Ser Leu His Tyr
GAG
Glu 145 GOT COC 0CC GTO GAC ATC ACC ACO TCG Gly Arg Ala Val Asp Ile Thr Thr Ser
OAT
Asp 155 COG GAC CGC AGC Arg Asp Arg Ser
AAG
Lys 160 TAC GGA ATG CTG Tyr Gly Met Leu 0CC Ala 165 CGC CTC 0CC GTC Arg Leu Ala Val 0CC GOC TTC GAC Ala Gly Phe Asp TGG OTC Trp Val 175 TAC TAC GAG Tyr Tyr Glu
TCC
Ser 180 AAG 0CC CAC ATC Lys Ala His Ile TGC TCC GTC AAA Cys Ser Val Lys GCA OAA AAC Ala Giu Asn 190 TCA GTG GCA GCG Ser Val Ala Ala 195 AAA TCA GGA Lys Ser Gly
GGC
Gly 200 13 Z/ TGC TTC CCT GGC TCA Cys Phe Pro Gly Ser 205 GCC ACA G'rG Ala Thr Val CAC CTG GAG His Leu Giu 210 CAT GGA GGC ACC AAG CTG GTG AAG His Gly Gly Thr Lys Leu Val Lys
GAC
Asp 220 CTG AGC CCT G Leu Ser Pro Gly
GAC
Asp 225 CCC GTG CTG GCT Arg Val Leu Al1a
GCT
Ala 230 GAC GCG GAC GGC Asp A1a Asp Giy
CG
Arg 235 CTG CTC TAC ACT Leu Leu Tyr Ser
GAC
Asp 240 TTC CTC ACC TTC Phe Leu Thr* Phe
CTC
Leu 245 GAC CGG ATG GAC Asp Arg Met Asp
AGC
Ser 250 TCC CGA AAG CTC Ser Arg Lys Leu TTC TAC Phe Tyr 255 GTC ATC GAG Val Ile Glu CAC CTG CTC His Leu Leu 275 COG CAG CCC CGG Arg Gin Pro Arg
GCC
Ala 265 CGG CTG CTA CTG Arg Leu Leu Leu ACG GCG GCC Thr Ala Ala 270 CCC ACA CG Ala Thr Gly TTT GTG CCC CCC Phe Val Ala Pro
CAC
Gin 280 CAC AAC CAG TCG His Asn Gin Ser TCQ ACC Ser Thr 290 ACT GGC CAG GCG Ser Gly Gin Ala
CTC
Leu 295 TTC CCC AGC AAC Phe Ala Ser Asn
GTG
Val 300 AAG CCT 0CC CAA Lys Pro Gly Gin
S
CGT
30 Arg 305 CTC TAT GTG CTG Val Tyr Val.Leu
GC
Giy 310 GAG GGC GGG CAG Giu Gly Gly Gin
CAG
Gin 315 CTG CTC CCC C Leu Leu Pro Ala
TCT
Ser 320 GTC CAC AGC CTC Val His Ser Val
TCA
Ser 325 TTG CCC GAG GAG Leu Arg Giu Ciu
GCG
Ala 330 TCC GGA GCC TAC 0CC CCA Ser Gly Ala Tyr Ala Pro 960 1008 1056 1104 CTC ACC 0CC Leu Thr Ala TAC CCC GTC Tyr Ala Val 355
CAG
GIn 340 GGC ACC ATC CTC Gly Thr Ile Leu
ATC
Ile 345 AAC CGG GTG TTG Asn Arg Val Leu CCC TCC TGC Ala Ser Cys 350 TTC GCA CCA Phe Ala Pro ATC GAG GAG CAC Ile Glu~ Giu His TGG GCC CAT TGG Trp Ala His Trp
GCC
Ala 365 TTC CC Phe Arg 370 TTG OCT CAG CCC Leu Ala Gin Giy CTG 0CC 0CC CTC Leu Ala Ala Leu
TGC
Cys 380 CCA GAT COG CC Pro Asp Gly Ala
ATC
Ile 385 CCT ACT 0CC 0CC Pro Thr Ala Ala
ACC
Thr 390 ACC ACC ACT GGC Thr Thr Thr Gly
ATC
Ile 395 CAT TGG TAC His Trp Tyr TCA CG Ser Arg .400 CTG CAT Leu His 415 1152 1200 1248 CTC CTC TAC CC Leu Leu Tyr Arg
ATC
Ile 405 GGC AGC TGG CTG Cly Ser Trp Val
CTG
Leu 410 GAT GOT GAC C Asp Gly Asp Ala CCG CTG GGC ATG GTG GCA CCG GCC AGC TG Pro Leu Gly Met Val Ala Pro Ala Ser 420 425 INFORMATION FOR SEQ ID NO:2: SEQUENCE CHARACTERISTICS: LENGTH: 1190 base pairs TYPE: nucleic acid STR.ANDEDNESS: both TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURE: NAME/KEY: CDS LOCATION: 1..1191 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:2: 1277 ATG GCT CTG CCG GCC AGT CTG TTG CCC CTG TGC TGC TTG ,Met Ala Leu Pro Ala Ser Leu Leu Pro Leu Cys Cys Leu GCA CTC TTG Ala Leu Leu
C.
C
C
p GCA CTA TCT GCC CAG AGC TGC GGG CCG Ala Leu Ser Ala 20 Gin Ser Cys Gly GGC CGA OGA CCG GTT GGC CGG Gly Arg Gly Pro Val Gly Arg CGG CGT TAT GTG CGC AAG CAA CTT Arg Arg Tyr Val Arg Lys Gln GTG CCT CTG CTA Val Pro Leu Leu Leu 40
TAC
Tyr AAG CAG TTT Lys Gln Phe GTG CCC AGT ATG CCC Val Pro 50 Ser Met Pro GAG CG Giu Arg ACC CTG GOC GCG Thr Leu Gly Ala 000 CCA GCG GAG Gly Pro Ala Glu 144 192 240
GGG
Gly 65 AGO GTA ACA AGO Arg Val Thr Arg TCG GAG CGC TTC Ser Giu Arg Phe CGG GAC Arg Asp 75 CTC GTA CCC Leu Val Pro
AAC
Asn TAC AAC CCC GAC 45 Tyr Asn Pro Asp
ATA
Ile as ATC TTC AAG OAT Ile Phe Lys Asp
GAG
Glu 90 GAG AAC AOC GOC Glu Asn Ser Gly GCA GAC Ala Asp CGC CTG ATO Arg Leu Met
ACA
Thr 100 GAG COT TGC AAA Oiu Arg Cys Lys
GAG
Glu 105 CGG OTO AAC OCT CTA 0CC ATC Arg Val Asn Ala Leu Ala Ile 110 GCG GTG ATG AAC ATG TOG CCC Ala Val Met Asn Met Trp Pro 12.5
GGA
Gly 120 GTA CGC CTA CGT GTG ACT GAA GGC Val Arg Leu Arg Val Thr 0Th Oly 125 TGG GAC GAG Trp Asp Glu 1.1 n GAC GOC CAC Asp Gly His GCA CAG GAT TCA Ala Gin Asp Ser
CTC
Leu 'I A CAC TAC GAA GOC His Tyr Olu Oly
CGT
Arg 145 /3' GCC TTG GAC ATC ACC ACG TCT GAC CGT Ala Leu Asp Ile Thr Thr Ser Asp Arg GAC CGT AAT AAG TAT GGT Arg Asn Lys Tyr Gly 160 TTG TTG GCG CGC CTA GCT GTG GAA GCC Leu Leu Ala Arg Leu Ala Val Glu Ala 165
GGA
Gly 170 TTC GAC TGG GTC Phe Asp Trp Val TAC TAC Tyr Tyr 175 GAG TCC CGC Glu Ser Arg
AAC
Asn 180 CAC ATC CAC GTA TCG GTC AAA His Ile His Val Ser Val Lys 185 GCT GAT AAC TCA CTG Ala Asp Asn Ser Leu 190 GCG GTC CGA Ala Val Arg 195 GCC GGA GGC TGC Ala Gly Gly Cys
TTT
Phe 200 CCG GGA AAT GCC Pro Gly Asn Ala GTG CGC TTG Val Axg Leu CGG AGC Arg Ser 210 GGC GAA CGG AAG Gly Glu Arg Lys
GGG
Gly 215 CTG AGG GAP. CTA Leu Arg Glu Leu
CAT
His 220 CGT GGT GAC TGG Arg Gly Asp Trp,
GTA
Val 225 CTG GCC GCT GAT Leu Ala Ala Asp
GCA
Al a 230 GCG GGC CGA GTG Ala Gly Arg Val
GTA
Val 235 CCC ACG CCA GTG Pro Thr Pro Val
CTG
Leu 240 CTC TTC CTG GAC Leu Phe Leu Asp
CGG
Arg 245 GAT CTG CAG CC Asp Leu Gln Arg
CGC
Arg 250 GCC TCG TTC GTG Ala Ser Phe Val GCT GTG Ala Val 255
C
GAG ACC GAG CGG Giu Thr Glu Arg 260 CCT CCG CGC AAA Pro Pro Arg Lys CTG TTG CTC ACA Leu Leu Leu Thr 265 CCT GCT CCA GGT Pro Ala Pro Gly
CCC
Pro
GAC
Asp 285 TGG CAT CTG Trp His Leu 270 TTT GCA CCG Phe Ala Pro GTG TTC GCT Val Phe Ala 275 GCT CGC GGG CCA Ala Arg Gly Pro GTG TTC Val Phe 290 GCG CGC CGC TTA Ala Arg Arg Leu GCT GGC GAC TCG Ala Gly Asp Ser CTG GCT CCC GGC Leu Ala Pro Gly
GGG
Gly 305 GAC GCG CTC CAG Asp Ala Leu Gin GCG CGC GTA GCC Ala Arg Val Ala GTG C CGC GAG Val Ala Arg Glu
GAA
Glu 320 960 1008 4 45 0CC GTG GGC GTG Ala Val Gly Val
TTC
Phe 325 GCA CCG CTC ACT Ala Pro Leu Thr
C
Ala 330 CAC COG ACG CTG His Gly Thr Leu CTG GTC Leu Val 335 AAC GAC GTC Asn Asp Val GCC CAC CGC Ala His Arg 355 GCC TCC TGC TAC Ala Ser Cys Tyr
C
Ala.
345 CTT CTA GAG ACT Val Leu Glu Ser CAC CAC TG His Gln Trp, 350 CTC GGC GCT Leu Gly Ala 1056 1104 GCC TTC CCC CCT Ala Phe Ala Pro TTG CGG Leu Arg 360 CTC CTG CAC Leu Leu His j37 CTG CTC CCT GGG GGT GCA GTC CAG CCG ACT GGC ATG Leu Leu Pro Gly Gly Ala Val Gin Pro Thr Gly Met 370 375 380 CAT TGG TAC TCT His Trp Tyr Ser 1152 z) CGC CTC CTT TAC CGC TTG GCC GAG GAG TTA ATG GGC TG Arg Leu Leu Tyr Arg Leu Ala Glu Glu Leu Met Gly 385 390 395 INFORM4ATION FOR SEQ ID NO:3: SEQUENCE CHARACTERISTICS: LENGTH: 1056 base pairs TYPE: nucleic acid STRANDEDNESS: both TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 1190 (ix) FEATURE: NAME/KEY: CDS LOCATION: 1056 (xi) SEQUENCE DESCRIPTION: SEQ, ID NO:3: GAG CGC TTC AAA GAG CTC ACC CCC AAC TAC AAT CCC GAC ATC Glu Axg Phe Lys Glu Leu Thr Pro Asn Tyr Asn Pro Asp Ile 30 1 ;in ATC TTC Ile Phe AAG GAC GAG GAG AAC Lys Asp Glu AAG GAC CGT Lys Asp Arg Glu Asn ACG GGT CC Thr Gly Ala CGC CTC ATG ACC Arg Leu Met Thr CAG CCC TGC Gin Arg Cys CAG TGG CCT Gin Trp Pro CTG AAC TCA CTG Leu Asn Ser Leu
GCC
Ala ATC TOT CTC ATG Ile Ser Val Met GGT GTG Cly Val.
s0 AAA CTG CGG GTG Lys Lieu Arg Val GAA GGC CGG GAT Glu Gly Arg Asp
GAA
Glu GAT GGC CAT CAC Asp Gly His His 192
TCA
45 Ser 65 GAG GAG TCT TTA Glu Glu Ser Leu TAT GAG GGC CC Tyr Glu Gly Arg GTG CAT ATC ACC Val Asp Ile Thr TCA GAC CT GAC OGA AAT AAG TAT GGA Ser Asp Arg Asp Arg Asn Lys Tyr Gly as GAG GCC GGC TTC GAO TGG GTG TAT TAC Glu Ala Gly Phe Asp Trp Val Tyr Tyr
CTG
Leu 90 CTG CC CGC TTA Leu A-la Arg Leu GCA GTG Ala Val GAG TCC AAG CC GlU Ser Lys Ala CAC GTG CAT His Val His 110 GCT GCC TGC Gly Gly Cys TGC TCT GTC AAG TOT GAG CAT Cys Ser Val Lays Ser Glu His GOC GCT GOC AAG Ala Al1a Ala Lys TTT CCT GCC GGA GCC CAG GTG CGC CTA GAG AAC GGG GAG CGT GTG GCC Phe Pro 130 Ala Gly Ala Gin Arg Leu Glu Asn Gly Glu Arg Val Ala 140 CTG TCA GC'r GTA AAG Leu Ser 145 Ala Val Lys
CCA
Pro 150 GGA GAC CGG GTG CTG GCC ATG GGG GAG GAT Gly Asp Arg Val Leu Ala Met Gly Glu Asp GGG ACC CCC ACC Gly Thr Pro Thr
TTC
Phe 165 AGT GAT GTG CTT Ser Asp Val Leu
ATT
Ile 170 TTC CTG GAC CGC Phe Leu Asp Arg GAG CCA Giu Pro 175 AAC CGG CTG Asn Arg Leu CGG CTG GCG Arg Leu Ala 195 GCT TTC CAG GTC Ala Phe Gln Val GAG ACT CAG GAT Glu Thr Gin Asp CCT CCG CGT Pro Pro Arg 190 GAC AAT CAT Asp Asn His CTC ACG CCT GCC Leu Thr Pro Ala
CAC
His 200 CTG CTC TTC ATT Leu Leu Phe Ile
GCG
Ala 205 ACA GAA Thr Glu 210 CCA GCA GCC CAC Pro Ala Ala His CGG GCC ACA TTT Arg Ala Thr Phe
CC
Ala 220 AGC CAT GTG CAA Ser His Val Gin
CCA
Pro 225 GGC CAA TAT GTG Gly Gin Tyr Val
CTG
Leu 230 GTA TCA GGG GTA Val Ser Gly Val
CCA
Pro 235 GGC CTC CAG CCT Gly Leu Gin Pro
GCT
Ala 240
C
CGG GTG GCA OCT Arg Val Ala Ala
GTC
Val 245 TCC ACC CAC GTG Ser Thr His Val CTT GGG TCC TAT Leu Gly Ser Tyr GCT CCT Ala Pro 255 =TC ACA AGG Leu Thr Arg TTT GCA GCT Phe Ala Ala 275
CAT
His 260 GGG ACA CTT GTG Gly Thr Leu Val
GTG
Val 265 GAG GAT GTG GTG Glu Asp Val Val GCC TCC TGC Ala Ser Cys 270 TTC TGG CCC Phe Trp Pro GTG GCT GAC CAC Val Ala Asp His CTG GCT CAG TTG Leu Ala Gin Leu
GCC
Ala 285 iCTG CGA "Teu Arg 290 CTG TTT CCC AGT Leu Phe Pro Ser GCA TGG G0G0C AGC Ala Trp Giy Ser
TG
Trp 300 ACC CCA AGT GAG Thr Pro Ser Giu
GGT
Gly 305 GTT CAC TCC TAC CCT CAG ATG CTC TAC Val His Ser Tyr Pro Gin Met Leu Tyr
CC
Arg 315 CTG GGG CGT CTC Leu Gly Arg Leu CTA GAA GAG AGC ACC TTC CAT CCA CTG Leu Glu Giu Ser Thr Phe His Pro Leu 325
GC
Gly 330 ATG TCT GGG GCA Met Ser Gly Ala GGA AGC Gly Ser 335 1008 1056 TGAAGGGACT CTAACCACTG CCCTCCTGGA ACTGCTGTGC GTGGATCC INFORMATION FOR SEQ ID NO:4:
ATG
met
I
SEQUENCE CHARACTERISTICS: LENGTH: 1313 base pairs TYPE: nucleic acid STRANDEDNESS: both TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (ix) FEATURZE: NAME/KEY: CDS, LOCATION: 1. .1314 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4: CTG CTG CTG CTG GCC AGA TGT TTT CTG GTG ATC CTT GCT TCC TCG Leu Leu Leu Leu Ala Arg Cys Phe Leu Val Ile Leu Ala Ser Ser
CTG
Leu CTG GTG TGC CCC Leu Val Cys Pro GGG CTG GCC TGT Gly Leu Ala Cys 25 GGG CCC GGC AGO GGG TTT GGA Gly Pro Gly Arg Gly Phe Gly AAG AGG CGG Lys Arg Arg CAC CCC AAA AAG CTG ACC CCT TTA His Pro Lys Lys Leu Thr Pro Leu GCC TAC AAG CAG TTT Ala Tyr Lys Gln Phe AGC GGC AGA TAT GAA Ser Gly Arg Tyr Glu
ATT
Ile
GGG
Gly CCC AAC GTA CC Pro Asn Val Ala so AAG ATC ACA AGA Lys Ile Thr Arg GAG AAG Glu Lys 55 ACC CTA GGG GCC Thr Leu Gly Ala 96 144 192 240 288 TCC GAA CGA TTT Ser Glu Arg Phe
AAG
Lys 75 GAA CTC ACC CCC Glu Leu Thr Pro
AAT
Asn.
so TAC AAC CCC GAC Tyr Asn Pro Asp
ATC
Ile ATA TTT AAG GAT Ile Phe Lys Asp
GAG
Giu 90 GAA AAC ACG G;GA Giu Asn Thr Gly GCA GAC Ala Asp CGG CTG ATG Arg Leu Met TCT GTG ATG Sex Val Met 115
ACT
Thr 100 CAG AGG TGC AAA Gin Arg Cys Lys AAG TTA AAT GCC Lys Leu Asn Ala TTG GCC ATC Leu Ala Ile 110 ACC GAG GGC Thr Giu Gly AAC CAG TGG CCT Asn Gin Trp Pro
GGA
Gly 120 GTG AGG CTG Val Arg Leu CGA GTG Arg Val 125 TGG GAT Trp Asp 130 GAG GAC GOC CAT Glu Asp Gly His
CAT
His 135 TCA GAG GAG TCT CTA CAC TAT GAG GOT Ser Glu Glu Ser Leu His Tyr Glu Gly 432 480
CGA
Arg 145 GCA GTG GAC ATC ACC ACG TCC GAC CGG Ala Val Asp Ile Thr Thr Ser Asp Arg
GAC
Asp 155 CGC AGC AAG TAC Arg Ser Lys Tyr ATG CTG GCT CGC Met Leu Ala Arg
CTG
Leu 165 GCT GTG GAA GCA Ala Val Giu Ala ATC CAC TGT TCT Ile His Cys Ser 185 GGT TTC GAC TGG GTC TAC TAT Phe Asp Trp Val Tyr Tyr 175 GAA TCC AAA Glu Ser Lys GCT CAC Ala His 180 GTG AAA GCA GAG AAC TCC GTG Val Lys Ala Glu Asn Ser Val 190 GCG GCC AAA Ala Ala Lys 195 TCC GGC GGC TGT Ser Gly Gly Cys
TTC
Phe 200 CCG GGA TCC GCC Pro Gly Ser Ala
ACC
Thr 205 GTG CAC CTG Val His Leu GAG CAG Glu Gin 210 GGC GGC ACC.AAG Gly Gly Thr Lys
CTG
Leu 215 GTG AAG GAC TTA Val Lys Asp Leu CGT CCC GGA GAC Arg Pro Gly Asp 220 TAC AGC GAC TTC Tyr Ser Asp Phe
CGC
Arg C'rC Leu 240
GTG
Val 225 CTG GCG GCT GAC Leu Ala Ala Asp_ CAG GGC CGG CTG Gin Gly Arg Leu
CTG
Leu 235 ACC TTC CTG GAC Thr Phe Leu Asp GAC GAA GGC GCC Asp Glu Gly Ala
AAG
Lys 250 AAG GTC TTC TAC Lys Val Phe Tyr GTG ATC Val Ile 255 GAG ACG CTG Glu Thr Leu
GAG
Glu 260 CCG CGC GAG CGC Pro Arg Glu Arg
CTG
Leu 265 CTG CTC ACC GCC Leu Leu Thr Ala GCG CAC CTG Ala His Leu 270 GGG CCA AGC Gly Pro Ser 4* CTC TTC Leu Phe GTG GCG CCG CAC AAC Val Ala Pro His Asn 275 TTT GCC AGC CGC GTG Phe Ala Ser Arg Val 295 TCG GGG CCC ACG Ser Gly Pro Thr
CCC
Pro 285 GCG CTC Ala Leu 290 CGC CCC GGG CAG Arg Pro Gly Gin
CGC
Arg 300 GTG TAC GTG GTG Val Tyr Val Val 816 864 912 960 1008 1056
GCT
Ala 305 GAA CGC GGC GGG Glu Arg Gly Gly
GAC
Asp 310 CGC CGG CTG CTG Arg Arg Leu Leu
CCC
Pro 315 GCC GCG GTG CAC Ala Ala Val His
AGC
Ser 320 GTG ACG CTG CGA GAG GAG GAG GCG GGC Val. Thr Leu Arg Glu Glu Glu Ala Gly
GCG
Ala 330 TAC GCG CCG CTC Tyr Ala Pro Leu ACG GCG Thr Ala 335 *4 CAC GGC ACC His Gly Thr ATT CTC Ile Leu 340 ATC AAC CGG GTG CTC GCC TCG TGC Ile Asn Arg Val Leu Ala Ser Cys 345 TAC GCT GTC Tyr Ala Val 350 TTC COC CTG Phe Arg Leu ATC GAG GAG Ile Glu Glu 355 CAC AGC TGG GCA His Ser Trp Ala
CAC
His 360 CGG GCC TTC GCG Arg Ala Phe Ala 1104 GCG CAC Ala His 370 GCG CTG CTG 0CC Ala Leti Leu Ala
GCG
Ala 375 CTG GCA CCC GCC Leu Ala Pro Ala
CGC
Arg 380 ACG GAC GOC GG Thr Asp Gly Gly 1152
GGC
Gly 385 41I GGG GGC AGC ATC CCT GCA GCG CAA TCT Gly Gly Ser Ile Pro Ala Ala Gin Ser 390 ACG GAA GCG AGG Thr Glu Ala Arg
GGC
Gly 400 1200 GCG GAG CCG ACT Ala Glu Pro Thr
GCG
Ala 405 GGC ATC CAC TGG Gly Ile His Trp
TAC
Tyr 410 TCG CAG CTG CTC Ser Gin Leu Leu TAC CAC Tyr His 415 1248 ATT GGC ACC TGG CTG TTG GAC AGC Ile Gay Thr Trp Leu Leu Asp Ser 420
GAG
Glu 425 ACC ATG CAT CCC Thr Met His Pro TTG GGA ATG Leu Gly Met 430 1296 GCG GTC AAG TCC AGC TG Ala Val Lys Ser Ser 435 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 1256 base pairs TYPE: nucleic acid STRANDEDNESS: both TOPOLOGY: linear (ii) MOLECUL TYPE: cDNA (ix) FEATURE: ZNAME/KEY: CDS LOCATION: 1. .1257 (xi) SEQUENTCE DESCRIPTION: SEQ ID 1313 ATG CGG CTT rrG ACG AGA GTG CTG CTG GTG TCT CTT CTC ACT Met Arg Leu Leu Thr Arg Val Leu Leu Val Ser Leu Leu Thr CTG TCC Leu Ser TTG GTG GTG Leu Val Val AGA AGA CAT 45 Arg Arg His GGA CTG GCC TGC Gly Leu Ala Cys CCT GOC AGA GGC Pro Gly Arg Gly TAC GGC AGA Tyr Gly Arg GAG TTC ATA Gin Phe Ile CCG AAG AAG CTG Pro Lys Lys Leu CCT CTC GCC TAC Pro Leu Ala Tyr
AAG
Lys CCT AAT Pro Asn so GTC GCG GAG AAG Val Ala Glu Lys TTA GGG GCC AGC Leu Gly Ala Ser AGA TAC GAG GGC Arg Tyr Glu Gly AAG ATA ACG CGC AAT Lys Ile Thr Arg Asn AAT CCC GAC ATI ATC Asn Pro Asp Ile Ile GAG AGA TTr AAA GAA CTT ACT CCA AAT Glu Arg Phe Lys Glu Leu Thr Pro Asn TI'T AAG GAT GAG Phe Lys Asp Glu GAG AAC Glii Asn 4!1 ACG GGA GCG Thr Gly Ala GAC AGG Asp Arg CTC ATG ACA CAG AGA Leu Met Thr Gin Arg 100 GTA ATG AAC CAC TGG Val Met Asn His Trp 115 1142 TGC AAA GAC AAG CTG AAC TCG CTG GCC ATC TCT Cys Lys Asp Lys Leu Asn Ser Leu Ala Ile Ser 105 110 CCA GGG GTT AAG CTG CGT GTG ACA GAG GGC TGG Pro Gly Val Lys Leu Arg Val. Thr Giu Gly Trp GAT GAG GAC GGT CAC Asp Giu 130 Asp Gly His
CAT
His
ACC
Thr IS50 TTT GAA GAA TCA CTC Phe GJlu Glu Ser Leu CAC TAC GAG GGA His Tyr Giu Gly 140 AGC AAA TAC GGG Ser Lys Tyr Gly
AGA
Arg
ACA
Thr 160
GCT
Al a 145 GTT GAT ATT ACC Val Asp Ile Thr TCT GAC CGA GAC Ser Asp Arg Asp
AAG
Lys 155 CTG TCT CGC CTA Leu Ser Arg Leu
GCT
Ala 165 GTG GAG GCT GGA Val Glu Ala Gly
TTT
Phe 170 GAC TGG GTC TAT Asp Trp Val Tyr TAC GAG Tyr Glu 175 TCC AAA GCC Ser Lys Ala GCG AAA TCT Ala Lys Ser 195
CAC
His 180 A.TT CAT TGC TCT Ile His Cys Ser
GTC
Val.
185 AAA OCA GAA AAT Lys Ala Glu Asn TCG GTT GCT Ser Val Ala 190 TCG CTC CAG Ser Leu Gln GGG GGC TGT TTC Gly Gly Cys Phe
CCA
Pro 200 GGT TCG OCT CTG Gly Ser Ala Leu
GTC
Val 205 576 624 672 GAC GGA ASp Gly 21.0 GGA CAG AAG GCC Gly Gin Lys Ala AAG GAC CTG AAC Lys Asp Leu Asn
CCC
Pro 220 GGA GAC AAG GTG Gly Asp Lys Val
CTG
Leu 225 OCO GCA GAC AGC Ala Ala Asp Ser
GCG
Ala 230 OGA AAC CTG GTG Gly Asn Leu Val
TTC
Phe 235 AGC GAC TTC ATC Ser Asp Phe Ile
ATG
Met 240 TTC ACA GAC Phe Thr Asp ACG CAA GAA Thr Gin Glu CGA GAC Arg Asp 245 TCC ACG ACG CGA Ser Thr Thr Arg
CGT
Arg 250 GTG TTT TAC GTC Val. Phe Tyr Val.
ATA GAA le Glu 255
CCC
Pro 260 GTT GAA AAG ATC Val Glu Lys Ile
ACC
Thr 26S CTC ACC GCC OCT Leu Thr Ala Ala CAC CTC CTT His Leu Leu 270 ACC GCC GCG Thr Ala Ala 45, TTT GTC CTC Phe Val. Leu 275 TAT GCC AGC Tyr Ala Ser 290 AGC GGT CAG Ser Gly Gin 305 GAC AAC TCA ACG Asp Asn Ser Thr
GAA
Glu 280 GAT CTC CAC ACC Asp Leu His Thr AGT GTC AGA Ser Vai Arg GGA CAA AAO GTG Gly Gin Lys Val.
ATG
Met 300 GTT GTT GAT GAT Va]. Val Asp ALsp CTT AAA TCT GTC ATC GTG CAG Leu Lys Ser Va]. Ile Vai Gin CGG ATA TAC ACG GAG GAG Arg Ile Tyr Thx Giu Giu 315 320 CAG CGG GGC TCG Gln Arg Gly Ser
TTC
Phe 325 GCA CCA GTG ACT Ala Pro Val. Thr ILf 3
GCA
Al a 330 CAT GGG ACC ATT GTG GTC 1008 His Gly Thr Ile Val Val.
335 3 GAC AGA ATA Asp .Arg Ile GCG CAT TTG Ala His Leu 355
CTG
Leu 340 GCG TCC TGT TAC Ala Ser Cys Tyr
GCC
Al a 345 GTA ATA GAG GAC Val Ile Glu Asp CAG GGG CTT Gin Gly Leu 350 GTG TCA TCA Va). Ser Ser 1056 1104 GCC TTC GCG CCC Ala Phe Ala Pro
GCC
Al a 360 AGG CTC TAT TAT Arg Leu Tyr Tyr TTC CTG Phe Leu 370 TCC CCC AAA ACT Ser Pro Lys Thr
CCA
Pro 375 GCA GTC GGT CCA Ala Val. Gly Pro
ATG
Met 380 CGA CTT TAC AAC Arg Leu Tyr Asn
AGG
Arg 385 AGG GGG TCC ACT Arg Gly Ser Thr
GGT
Gly 390 ACT CCA GGC TCC Thr Pro Gly Ser
TGT
Cys 395 CAT CAA ATG GGA His Gin Met Gly
ACG
Thr 400 1152 1200 1248 TGG CTT TTG GAC Trp Leu Leu Asp
AGC
Ser 405 AAC ATG CTT CAT Asn Met Leu His
CCT
Pro 410 TTG GGG ATG TCA Leu Gly Met Ser GTA AAC Val Asn 415 TCA ACC TG Ser Ser INFORMATION FOR SEQ ID NO:6: Wi SEQUJENCE CHARACTERISTICS: LENGTH: 1425 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA 1256 (ix) FEATURE: NAME/KEY: CDS LOCATION: 1. .1425 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6:
ATG
Met :1 CTG CTG CTG C AGA TGT CTG CTC CTA GTC CTC GTC TCC Leu Leu Leu Ala Arg Cys Leu Leu Leu Val Leu Val Ser TCG CTG Ser Leu CTC GTA TOC TCG GGA CTG C TGC GCA CCC CCC AGG, CCC TTC COG AAG Leu Val Cys 5cr Gly Leu Ala Cys Gly Pro Gly Arg Gly Phe Gly Lys AGO AGG CAC CCC AAA AAG CTG ACC CCT TTA CC Arg Arg His Pro Lys Lys Leu Thr Pro Leu Ala Is TAC AAG CAG TTT ATC Tyr Lys Gin Phe Ile CCC AA'T Pro Asn GTG GCC GAG AAG ACC CTA GGC Val Ala Giu Lys Thr Leu Gly 55 GCC AGC GGA Ala Ser Gly AGG TAT GAA GGG Arg TPyr Glu Gly ACC CCC AAT TAC Thr Pro Asn Tyr
AAG
Lys ATC TCC AGA AAC TCC GAG CGA TTT AAG Ile Ser Arg Asn Ser Glu Arg Phe Lys 70 GAA CTC Giu Leu 75 AAC CCC GAC.ATC Asn Pro Asp Ile TTT AAG GAT GAA Phe Lys Asp Giu AAC ACC GGA Asn Thr Gly GCG GAC AGG Ala Asp Arg CTG ATG ACT Leu Met Thr GTG ATG AAC Vai Met Asn 115
CAG
Gin 100 AGG TGT AAG GAC Arg Cys Lys Asp
AAG
Lys 105 TTG AAC GCT TTG GCC ATC TCG Leu Asn Ala Leu Ala Ile Ser 110 CAG TGG CCA GGA Gin Trp, Pro Gly AAA CTG CGG GTG Lys Leu Arg Val
ACC
Thr 125 GAG GGC TGG Giu Giy Trp GAC GAA Asp Glu 130 GAT GGC CAC CAC Asp Gly His His
TCA
Ser 135 GAG GAG TCT CTG Giu Giu Ser Leu
CAC
His 140 TAC GAG GGC CGC Tyr Giu Gly Arg
GCA
Al a 145 GTG GAC ATC AC-C Val Asp Ile Thr TCT GAC CGC GAC Ser Asp Arg Asp
CGC
Arg 155 AGC AAG TAC GGC Ser Lys Tyr Gly
ATG
Met 160 CTG GCC CGC CTG 30 Leu Ala Arg Leu
GCG
Ala 165 GTG GAG GCC GGC Val Giu Ala Gly
TTC
Phe 170 GAC TGG GTG TAC Asp Trp Vai Tyr TAC GAG Tyr Giu 175 TCC AAG GCA Ser Lys Ala GCC AAA TCG Ala Lys Ser 195
CAT
His ATC CAC TG(" TCG Ile His Cys Ser GTG AAA GCA GAG AA Val Lys Ala Giu As 185 GGC TCG GCC ACG GT Gly Ser Ala Thr Va C TCG GTG GCG n Ser Val Ala .190 G CAC CTG GAG 1 His Leu Glu GGA GGC TGC TTC Gly Gly Cys Phe
CCG
Pro 200 576 624 672 CAG GGC Gin Gly 210 GGC ACC AAG CTG, Gly Thr Lys Leu
GTG
Val 215 AAG GAC CTG AGC Lys Asp Leu Ser
CCC
Pro 220 GGG GAC CGC GTG Gly Asp Arg Val *45 CTG Leu 225 C GCG GAC GAC Ala Ala Asp Asp
CAG
Gin 230 GGC COG CTG CTC Gly Arg Leu Leu
TAC
.Tyr 235 AGC GAC TTC CTC Ser Asp Phe Leu
ACT
Thr 240 TTC CTG GAC CGC Phe Leu Asp Arg GAC GGC GCC AAG Asp Giy Ala Lys GTC TTC TAC GTG Val Phe Tyr Vai ATC GAG le Giu 255 ACG CGG GAG Thr Arg Glu
CCG
Pro 260 CGC GAG CGC CTG Arg Giu Arg Leu
CTG
Leu 265 CTC ACC GCC GCG Leu Thr Ala Ala CAC CTG CTC His Leu Leu 270 'rrT GTG GCG Phe Val Ala 275 CCG CAC AAC GAC Pro His Asn Asp GCC ACC GGG GAG CCC Ala Thr Gly Giu Pro 225 GAG GCG TCC Clu Ala Ser TCG GGC Ser Gly 290 TCG GGG CCG CCT Ser Gly Pro Pro
TCC
Ser 295 GGG GGC GCA CTG Cly Gly Ala Leu
GCC
Gly 300 CCT CGG GCG CTG Pro Arg Ala Leu
TTC
Phe 305 GCC AGC CGC GTG Ala Ser Arg Val
CC
Arg 310 CCC GGC CAC CC Pro Gly Gin Arg
GTG
Val 315 TAC GTG GTG GCC Tyr Val Val Ala
GAG
Glu 320 COT GAC GGC GAC Arg Asp Gly Asp CCC CTC CTG CCC Arg Lou Leu Pro
CC
Ala 330 OCT OTG CAC AGC Ala Val His Ser GTG ACC Val Thr 335 1008 CTA AGC GAG Leu Ser Glu
GAG
Glu 340 CCC GCG CCC CC Ala Ala Gly Ala
TAC
Tyr 345 C CCC CTC ACG Ala Pro Leu Thr CCC CAG GCC Ala Gin Cly 350 GTC ATC GAG Val Ile Glu ACC ATT CTC ATC AAC CCC GTG Thr Ile Leu Ile Asn Arg Val 355 CCC TCG TCC TAC Ala Ser Cys Tyr
C
Ala 365 1056 1104 1152* GAG CAC Glu His 370 ACC TGG C. CAC Ser Trp Ala His
CGG
Arg 375' GCC rTC GC CCC Ala Phe Ala Pro CCC CTC GC CAC Arg Leu Ala His I.
C
30 Ala 385 CTC CTG OCT GCA Leu Leu Ala Ala
CTG
Leu 390 CC CCC CC CC Ala Pro Ala Arg
ACG
Thr 395 CAC CCC CCC CCC Asp Arg Oly Cly
GAC
Asp 400 ACC CCC CCC CCC Ser Gly Cly Oly CCC CCC CCC GC Arg Cly Cly Cly
GCC
Cly 410 CCC AGA GTA CCC CTA ACC Gly Arg Val Ala Leu Thr 1200 1248 1296 1344 OCT CCA GGT Ala Pro Gly CAC TGG TAC His Trp Tyr 435
OCT
Al a 420 CC GAC CCT CCC Ala Asp Ala Pro
GGT
Gly 425 CC CCC CCC ACC Ala Oly Ala Thr CC CCC ATC Ala Gly Ile 430 CTC CTG CAC Leu Leu Asp TCG CAC CTC CTC Ser Gln Leu Leu CAA ATA CCC ACC Gn Ile Cly Thr
TGG
Trp 445 AGC GAG Ser Ciii 450 CC CTG CAC CCC Ala Leu His Pro
CTG
Leu 455 CCC ATG CCG GTC Gly Met Ala Val
AAC
Lys 460 TCC ACC NNN AC Ser Ser Xaa Ser 1392
CCC
Arg 465 CCC CCC GGG GCA Cly Ala Gly Gly
GGG
Gly 470 C CGG GAG GG Ala Arg Clii Gly
GCC
Ala 475 1425 INFORMATION~ FOR SEQ ID NO:7: SEQuENCE CHARACTER.ISTICS: LENGTH: 940 base pairs STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: c-DNA (ix) FEATURE: NAME/KEY:. CDS LOCATION: 1. .940 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7: CGG CGC CTC ATG ACC CAG CGC TGC AAG GAC CGC CTG AAC TCG CTG GOT Arg Arg Leu Met Thr Gin Arg Cys Lys Asp Arg Leu Asn Ser Leu Ala ATC TCG GTG le Ser Vai GGC TGG GAO Gly Trp Asp AAC CAG TGG CCC GGT GTG AAG CTG CGG Asn Gin Trp Pro Gly Val Lys Leu Arg GTG ACC GAG Val Thr Glu GAG GAO 000 CAC CAC TCA GAG GAG TCO CTG CAT TAT GAG Giu Asp Gly His His Ser Glu Giu Ser Leu His Tyr Giu 144 GGC CGC Gly Arg so GCG GTG GAO ATO Ala Val Asp Ile
ACC
Thr 55 ACA TCA GAO Thr Ser Asp OGO GAO Arg Asp 000 AAT AAG TAT Arg Asn Lys Tyr .30O GGA Gly 65 OTG OTO 000 000 Leu Leu Ala Axg OCA GTG GAG 000 Ala Val Glu Ala TTT GAO TGG GTG Phe Asp Trp Val
TAT
Tyr so TAO GAG TOA AAG Tyr Giu Ser Lys CAO GTG CAT TOO TOO OTO AAG TCO GAG His Val His Cys Ser Val Lys Ser Oiu CAC TCG His Ser G00 GOA GC Ala Ala Ala CTG GAG AGT Leu Glu Ser 125
AAG,
Lys 100 AOG G00 GOC TOO TTO OCT 000 GGA GCC Thr Gly Gly Cys Phe Pro Ala Oly Ala GGG-GC 00 GT OTO Gly Ala Arg Val
GOO
Al a 120 TI'G TCA 0CC GTO Leu Ser Ala Val
AGO
Arg 125 GAG OTA CGO Gin Val Arg 110 COG GOA GAO Pro Gly Asp AGO GAT GTG Ser Asp Val OGT GTG Arg Val 130 O*TG 000 ATG G Leu Ala Met Gly
GAG
Olu 135 OAT 000 AGO COO Asp Gly Ser Pro ACC TTC Thr Phe 140 c'ro Leu 145 ATT TTO OTO GAO OGO GAG 000 CAO AGO Ile Phe Leu Asp .Arg Glu Pro His Arg AGA G00 TTO CAG Arg Ala Phe Gin ATO GAG ACT Ile Glu Thr CAG GAO CCC COA OGO 000 OTO Gin Asp Pro Pro Arg Arg Leu 165 170 GCA OTO ACA CCC Ala Leu Thr Pro GOT GAO Ala His 175 CTG CTC TTT Leu Leu Phe CC ACA TTT Ala Thr Phe 195 GGG GTG CCA Gly Val Pro 210
ACG
Thr 180 GCT GAC AAT CAC Ala Asp Asn His
ACG
Thr 185 /1/7 GAG CCG GCA GCC Giu Pro Ala Ala CGC TTC CGG Arg Phe Arg 190 CTG GTG GCT Leu Val Ala GCC AGC CAC GTG Ala Ser His Val
CAG
Gin 200 CCT GGC CAG TAC Pro Gly Gin Tyr
GTG
Val 205 57 6 624 672 GGC CTG CAG Gly Leu Gin GCC CGC GTG GCA Ala Arg Val Ala
GCT
Ala 220 GTC TCT ACA CAC Val Ser Thr His
GTG
Val 225 GCC CTC GGG GCC Ala Leu Gly Ala CC CCG CTC ACA Ala Pro Leu Thr
AAG
Lys 235 CAT GGG ACA CTG His Gly Thr Leu
GTG
Val 240 GTG GAG GAT GTG Val Glu Asp Val
GTG
Val 245 .GCATCC TGC TTC Ala Ser Cys Phe
GCG
Ala 250 GCC GTG GCT GAC Ala Val Ala Asp CAC CAC His His 255 TTG, GCA Leu Ala 768 816 CTG GCT CAG Leu Ala Gln
TTG
Leu 260 GCC TTC TGG CCC Ala Phe Trp Pro
CTC,
Leu 265S AGA CTC TTT CAC Arg Leu Phe His GGC AGC TGG Tip Gly Ser Trp 275 ACC CCG GCGG Thr Pro Gly GGT GTG CAT TGG G1,y Val His Trp
TAC
Tyr 285 CCC CAG CTG Pro Gin Leu C.
C
C
C
C
CC C C C CTC TAC 30 Leu Tyr 290 CTG GGC Leu Gly 305 CGC CTG GGG CGT Arg Leu Gly Arg CTG CTA GAA GAG Leu Leu Glu Glu
GGC
Gly 300 AGC TTC CAC CCA Ser Phe His Pro ATC TCC GGG Met Ser Gly
GCA
Ala 310 GGG AGC TGA Gly Ser Xaa INFORMATION FOR SEQ ID NO:8: SEQUENCE CHARACTERISTICS: LENGTH: 425 amino acids TYPE: amino acid TOPOLOGY: linear 45 (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:8: Met Val Glu Met Leu Leu Leu Thr Arg Ile Leu Leu Val Gly 1 5 10 Phe Ile Cys Ala Leu Leu Val Ser Ser Gly Leu Thr Cys Gly Pro 25 Ile Gly Lys Arg Arg His Pro Lys Lys Leu Thr Pro Leu 40 Gly Arg Gly Ala Tyr Lys Gin Phe Ile Pro Asn Val Ala Giu Lys Thr Leu Gly Ala Ser Gly Arg 55 Tyr Giu Gly Lys Ile Thr Arg Asn Ser Giu Arg Phe Lys Giu Leu Thr 65 70 75 Pro Asn Tyr Asn Pro Asp Ile Ile Phe Lys Asp Giu Glu Asn Thr Gly 90 Ala Asp Arg Leu Met Thr Gin Arg Cys Lys Asp Lys Leu Asn Ala Leu 100 105 110 Ala Ile Ser Val Met Asn Gin Trp Pro Gly Val Lys Leu Arg Val. Thr.
115 120 125 Glu Gly Trp Asp Giu Asp Gly His His Ser Giu Giu Ser Leu His Tyr 130 135 140 Giu Gly Arg Ala Val Asp X1s Thr Thr Ser Asp Arg Asp Arg Ser Lys 143 150 155 160 Tyr Gly Met Leu Ala Arq Leu Ala Val Glu Ala Gly Phe Asp Trp Val.
165 170 175 Tyr Tyr Ser Lys Ala His Ile His Cys Ser Val. Ty Ala Gus 180 185 190 Ser Val Ala Ala Lys Ser Gly Gly Cys Phe Pro Gly Ser Ala Thr Val 195 200 205 *9 *His Leu Glu His Gly Gly Thr Lys Leu Val. Lys Asp Leu Ser Pro Gly S210 215 220 *Asp Arg Val Leu Ala Ala Asp Ala Asp Gly Arg Leu Leu Tyr Ser Asp 3S 225 230 235 240 Phe L eu Thr Phe Leu Asp Arg Met Asp Ser Ser Arg Lys Leu Phe Tyr 245 250 255 Val Ile Glu Thr Arg Gin Pro Arg Ala Arg Leu Leu Leu Thr Ala Ala 26D 265 270 His Leu Leu Phe Va]. Ala Pro Gin His Asn Gin Ser Giu Ala Thr Giy 275 280 285 4Li *Ser Thr Ser Gly Gin Ala Leu Phe Ala Ser Asn Val Lys Pro Gly Gin 290 295 300 Arg Val Tyr Val Leu Gly Giu Gly Gly Gin Gin Leu Leu Pro Ala Ser 305 310 315 320 Val. His Ser Val Ser Leu Arg Giu Giu Ala Ser Gly Ala Tyr Ala Pro 325 330 335 Leu Thr Ala Gin Gly Thr Ile Leu Ile Asn Arg Val Leu Ala Ser Cys 340 345 350 Tyr Ala Val Ile Glu 355 Phe Arg Leu Ala Gin 370 Ile Pro Thr Ala Ala 385 Leu Leu Tyr Arg Ile 405 Pro Leu Gly Met Val 420 INFORMATION FOR Glu Gl y Thr 390 Gly Al a His Leu 375 Thr Ser Pro Ser Trp 360 Leu Ala Thr Thr Trp Val Ala Ser 425 Ll 1 Ala His Trp Ala Phe Ala Pro 365 Ala Leu Cys Pro Asp Gly Ala 380 Gly Ile His Trp, Tyr Ser Arg 395 400 Leu Asp Giy Asp Ala Leu His 410 415 SEQ ID NO: 9: Wi SEQUENCE CHARACTERISTICS: LENGTH: 396 amino acids TYPE: amino acid TOPOLOGY: linear 000.
*00 a 00* 400 000.
Met Ala Arg Val 40 Gly Tyr Arg Ala Trp Arg 145 (ii) MOLECULE (xil SEQUENCE Ala Leu Pro Ala Leu Ser Ala Gin Arg Tyr Val Arg 35 Pro Ser Met Pro 50 Arg Val Thr Arg Asn Pro Asp Ile Leu Met Thr Glu 100 Val Met Asn Met 115 Asp Glu Asp Gly 130 Ala Leu Asp Ile TYPE: protein DESCRIPTION: SEQ ID NO:9: Ser Leu Leu Pro Leu Cys Cys Leu Ala Leu Leu' Ser Lys Glu Gly 70 Ile Arg Trp His Thr 150 Cys Gin Arg 55 Ser Phe Cys Pro His 135 Thr Gly Leu 40 Thr Giu Lys Lys Gly 120 Al a Ser Pro 25 Val Leu Arg Asp Glu 105 Val Gin Asp Gly Arg Pro Leu Gly Ala Phe Arg 75 Giu Glu 90 Arg Val Arg Leu Asp Ser Arg Asp 155 Gly Leu Ser Asp Asn Asn Arg Leu 140 Arg Pro Tyr Gly Lelu Ser Ala Val 125 His Asn Val Lys Pro Val Gly Leu 110 Thr Tyr Lys Gly Arg Gin Phe Ala Giu Pro Asn s0 Ala Asp Al a Ile Giu Gly Glu Gly Tyr Gly 160 Leu Leu Ala Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr 165 175 Glu Ser Ala Val Arg Se r 210 Val iLeu 225 Leu.Phe Glu..Thr Val0h 2.Val -Phe 290 Gly Asp 305 Ala Val Asn Asp Ala His Leu Leu 370 Arg Leu) 385 Arg Arg 195 Gly Ala Leu Glu Ala 275 Al a Ala cay Val Arg 355 Pro Lieu Asn 180 Ala Glu Ala Asp Arg 260 Ala Arg Leu Val Leu 340 Ala Gly Tyr His Gly Arg Asp Arg 245 Pro Arg Arg Gin Phe 325 Ala Phe Gly Arg Ile Gly Lys Ala 230 Asp Pro Gly Leu Pro 310 Al a Ser Ala Ala Leu 390 His Cys Gly 215 Al a Leu Arg Pro Arg 295 Ala Pro Cys Pro Val 375 Ala Val Phe 200 Leu 'sly L~ys 8 0 -Ja yr .so (An (i.W Ser 185 Pro Arg A rg I ,eu Pro
CAY
Val Th r Ala 345 Arg P,;.o G!.u Val Gly Glu Val Arg 250 Leu Ala Asp Ala Ala 330 Val Leu Thr [Leu *Lys Ala Asp *Asn Ala Thr 205 *Leu His Arg 220 Val. Pro Thr 235 Ala Ser Phe Leiu Thr Pro Pro Gly Asp 285 Ser, Val Leu 300 Axe Val Ala 315 His Gly Thr Leo Glu Ser Leu His Ala 365 Gly Met His 380 Met Gly 395 Asn 190 Val Gly Pro Val Trp 270 Phe Ala Arg [Leu ifis 350 eu rrp Ser Arg Asp Val Ala 255 Hi's Ala Pro Glu Leu 335 Gln Gly Tyr Leu Leu Trp Leu 240 Val Leu Pro Gl y Glu 320 V'al Trp la.
;er 0000 6@ SB 0 0 000000
S
00 00 *0 0
S
0000 0 @0 0 0
S@
0 000000 0 p000 .0 *000
OSOOOS
S
Set...
50 0 5e 00 INFORMATION FOR SEQ ID Wi SEQUENCE CHARACTERTSTICS: LENGTH: 336 amino acids; TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:l0: Glu Arg Phe Lys Glu Leu Thr Pro Asn Tyr Asn Pro Asp Ile Ile Phe 1 5 10 Lys Asp Glu Glu Asn Thr Gly Ala Asp Arg Leu Met Thr Gin Arg Cys 20 25 Lys Asp Arg Leu Asn Ser Leu Ala Ile Ser Val Met Asn Gin Trp Pro 40 Gly Val Lys Leu Arg Val Thr Glu Gly Arg Asp Glu Asp Gly His His 55 Ser Glu Glu Ser Leu His Tyr Glu Gly Arg Ala Val Asp Ile Thr Thr 70 75 Ser Asp Arg Asp Arg Asn Lys Tyr Gly Leu Leu Ala Arg Leu Ala Val 90 Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu Ser Lys Ala His Val His 100 105 110 Cys Ser Val Lys Ser Glu His Ser Ala Ala Ala Lys Thr Gly Gly Cys 115 120 125 Phe Pro Ala Gly Ala Gln Val Arg Leu Glu Asn G'.y Glu Arg Val Ala 130 135 140 Leu Ser.Ala Val Lys Pro Gly Asp Arg Val Leu Ala Met Gly Glu Asp 145 150 155 160 30 Gly Thr Pro Thr Phe Ser Asp Val Leu Ile Phe Leu Asp Arg Glu Pro 165 170 175 3 Asn Arg Leu Arg Ala Phe Gin Val Ile Glu Thr Gin Asp Pro Pro Arg 180 185 190 Arg Leu Ala Leu Thr Pro Ala His Leu Leu Phe Ile Ala Asp Asn His 195 200 205 S 40 Thr Glu Pro Ala Ala His Phe Arg Ala Thr Phe Ala Ser His Val Gln *210 215 220 Pro Gly Gln Tyr Val Leu Val Ser Gly Val Pro Gly Leu Gin Pro Ala 225 230 235 240 S Arg Val Ala Ala Val Ser Thr His Val Ala Leu Gly Ser Tyr Ala Pro 245 250 255 Leu Thr Arg His Gly Thr Leu Val Val Glu Asp Val Val Ala Ser Cys 260 265 270 Phe Ala Ala Val Ala Asp His His Leu Ala Gin Leu Ala Phe Trp Pro 275 280 285 Leu Arg Leu Phe Pro Ser Leu Ala Trp Gly Ser Trp Thr Pro Ser Glu 290 295 300 Gly Val. His Ser TPyr Pro Gin Met Leu Tyr Arg Leu Gly Arg Leu Leu 305 310 315 320 Leu Glu Giu Ser Thr Phe His Pro Leu Gly Met Ser Gly Ala Gly Ser 325 330 335 INFORMATION FOR SEQ ID NO:ii: SEQUENCE CHARACTERISTICS: LENGTH: 437 amino acids TYPE: amino acid TOPOLOGY- linear (ii) MOLE=UE TYPE: protein (xi) SEQUENCE DESC!PIPTZON: SEQ ID NO:11: Met Leu-T.eu Leu Leu Ala.Arg Cys Phe Leu Val Ile Leu Ala Ser Ser Leu Lys Ile Gly Tyr Arg Ser Trp Arg 145 Met Giu Leu .Arg Pro s0 Lys Asn Leu Val Asp 130 Ala Leu Ser Val Arg Asn Ile Pro Met Met 115 Glu Val Ala Lys Cys His Val Thr Asp Thr 100 Asn Asp Asp Arg Ala 180 Pro Pro Ala Arg Ile Gin GIn Gly Ile Leu 3.65 His Gly Lys.
Glu Asn 70 le Arg Trp His Thr 150 Ala Ile ,.eu Oer Phe (2ys Pro Fis 235 Thr Val His Ala I ieu 40 Thr Glu Lys Lys C-iy 1.20 Ser Sar Glu Cys Cys 25 Thr Leu Arg Asp Asp 105 Val Glu Asp Ala Ser 185 Gly Pro Gly Phe Giu 90 Lys Arg Giu Arg Gly 170 Val Pro Leu Ala Lys 75 Glu Leu Leu Ser Asp 155 Phe Lys Gly Ala Glu Asn Asn Arg Leu 140 Arg Asp Al a Arg TyIr Gly Leu Thr Ala Val 125 His Ser Trp, Glu Gly Lys Arg Thr Gly Leu 110 Thr Tyr Lys Val Asn 190 Phe Gin Tyr Pro Ala Ala Glu Glu Tyr Tyr 175 Ser Gly Phe Glu Asn Asp Ile Gly Gly Gly 160 Tyr Val Ala Ala Lys Ser Gly Gly Cys Phe Pro Gly Ser Ala Thr Vai His Leu Glu Gin Gly Gly Thr Lys Leu Val Lys Asp Leu 210 Arg Pro Gly Asp Arg 220 215 Va1 225 Thr Glu Leu Ala Ala 305 Val His Ile Ala Gly 35 385 Ala Leu Phe Thr Phe Leu 290 Glu Thr Gly Glu His 370 21 y 3hz Ala Leu Leu Val 275 Phe Arg Leu Thr Glu 355 Ala Gly Pro Alz Asp Glu 260 Ala Ala Gly Arg Ile 340 His Leu Ser Thr I Asp Arg 245 Pro Pro Ser Gly Glu 325 Leu Ser Leu Ile Ala 405 Asp 230 Asp Arg His Arg Asp 310 Glu Ile Trp Ala Pro 390 Gly Gir Glu Glu Asn Val 295 Arg Glu Asn Ala kla 375 kla Ile Gly Gly Arg Asp 280 Arg Arg Ala Arg His 360 Leu Ala His Arg Ala Leu 265 Ser Pro Leu Gly Val 345 Arg Ila rrp Leu Lys 250 Leu Gly Gly Leu Ala 330 Leu Ala Pro I Ser 3 Tyr S 410 Let 231 Lys Leu Pro Gin Pro 315 Tyr Ala Phe Ua '95 ier 1 Tyr Val Thr Thr Arg 300 Ala Ala Ser C Ala I Arg *1 380 Thr G Sei Phe Ala Pro 285 Val Ala Pro Cy s ?ro 165 'hr lu r As Alz 27C IGl) Tyr Val Leu
T
yr 350 Phe Asp Ala p Phe r Val 255 His Pro Val His Thr 335 Ala Arg Gly Arg Leu 240 Ile Leu Ser Val Ser 320 Ala Val Leu Gly Gly 400 0 .0 Gin Leu Leu Tyr His 415 S 40 Ile Giy Thr T-p Leu Leu Asp Ser Giu Thr Met His Pro n 420 425 Ala Val Lys Ser Ser 435 INFORMATION FOR SEQ ID NO:12: SEQUENCE CHARACTERISTICS: LENGTH: 418 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12: Leu Gly Met 430 /Al Met Arg Leu Leu Thr Arg Val Leu Leu Val Ser Leu Leu Thr Leu Ser Leu Arg Pro Lys Asn Leu Val Asp Ala 145 Leu Ser 35 Ala Asp Leu 225 Phe Thr Phe Val Arg Asn Ile Pro Met Met Glu 130 Val Ser Lys Lys Gly 210 Ala Thr Gin Val Val His Val Thr Asp Thr Asn 115 Asp Asp Arg Ala Ser 195 Gly Ala Asp Glu Leu 275 Ser 20 Pro Ala Arg Ile Gin 100 His Gly Ile Leu His 180 Gly Gin Asp Arg Pro 260 Asp Gly Lys Glu Asn Ile Arg Trp His Thr Ala 165 Ile Gly Lys Ser Asp 245 Val Asn Leu Lys Lys Ser 70 Phe Cys Pro His Thr 150 Val His Cys Ala Ala 230 Ser Glu Ser Ala Leu Thr 55 Glu Lys Lys Gly Phe 135 Ser Glu Cys Phe Val 215 Gly Thr Lys Thr Cys Thr 40 Leu Arg Asp Asp Val 120 Glu Asp Ala Ser Pro 200 Lys Asn Thr Ile Glu 280 Gly 25 Pro Gly Phe Glu Lys 105 Lys Glu Arg Gly Val 185 ly Asp Leu krg rhr 265 Asp Pro Leu Ala Lys Glu 90 Leu Leu Ser Asp Phe 170 Lys Ser Leu Vai Arg 250 Leu Leu Gly Arg Gly Tyr Gly Arg Ala Ser Glu 75 Asn Asn Arg Leu Lys 155 Asp Ala Ala Asn Phe 235 Val Thr His Tyr Gly Leu Thr Ser Val His 140 Ser Trp Glu Leu Pro 220 Ser Phe Ala Thr Lys Arg Thr Gly Leu Thr 125 Tyr Lys Val Asn Val 205 Gly Asp Tyr Ala I Met I 285 Gin Tyr Pro Ala Ala 110 Glu Glu Tyr Tyr Ser Ser Usp ?he lal is 170 rhr Phe Glu Asn Asp Ile Gly Gly Gly Tyr 175 Val Leu Lys Ile Ile 255 Leu Ala Ile Gly Tyr Arg Ser Trp Arg Thr 160 Glu Ala Gin Val Met 240 Glu Leu Ala Tyr Ala Ser Ser Val Arg Ala Gly Gin Lys Val Met Val Val Asp Asp Ser 305 Gin Asp Ala Phe Arg 385 Trp Ser Gly Arg Arg His Leu 370 Arg Leu Ser Lys Phe 325 Ala Phe Lys Thr Ser 405 Ser 310 Al a Ser Al a Thr Gly 390 Asn Val Thr Ala 345 Arg Val Gly His Gin Arg 315 Ala His 330 Val Ile Leu Tyr Gly Pro Ser Cys 395 Pro Leu 410 Ile Gly Giu Tyr Met 380 His Gly Thr Giu Ile Val 335 Gin Gly 350 Val Ser Leu Tyr Met Gly Ser Val 415 Giu 320 Val1 Leu Ser As n Thr 400 Asn 50 INFORMATION FOR SEQ ID NO:i3: Wi SEOUENCE CHARACTE!1,117 .uS.: WA LENGTHik: 475 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID M4et Leu Leu Leu Ala Arg Cys Leu Leu Leu 1 5 Leu Val Cys Ser Gly Leu Ala Cys Gly Pro 20 Arg Arg His Pro Lys Lys Leu Thr Pro Leu1 40 Pro Asn Val Ala Glu Lys Thr Leu Gly Ala 55 Lays Ile Ser Arg Asn Ser Glu Arg Phe LysC 65 70 Asn Pro Asp Ile Ile Phe Lys Asp Giu Glu Leu Met Thr Gin Arg Cys Lys Asp Lys Leu2 100 105 0:13.
ITal Leu Ily Arg Ua Tyr ;er Gly ;1u Leu %msn Thr Isn Ala Val Gly Lys Arg Thr Gly Leu is' Val Met Asn Gln Trp Pro Gly Val Lys Leu Arg Val Thr Glu Gly Trp 115 120 125 Asp Glu Asp Gly His His Ser Glu Glu Ser Leu His Tyr Glu Gly Arg 130 135 140 Ala Val Asp Ile Thr Thr Ser Asp Arg Asp Arg Ser Lys Tyr Gly Met 145 150 155 160 Leu Ala Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu 165 170 175 Ser Lys Ala His Ile His Cys Ser Val Lys Ala Glu Asn Ser Val Ala 180 185 190 Ala Lys Ser Gly Gly Cys Phe Pro Gly Ser Ala Thr Val His Leu Glu 195 200 205 Gin Gly Gly Thr Lys Leu Val Lys Asp Leu Ser Pro Gly Asp Arg Val 210 215 220 Leu Ala Ala Asp Asp Gin Gly Arg Leu Leu Tyr Ser Asp Phe Leu Thr 225 230 235 240 Phe Leu Asp Arg Asp Asp Gly Ala Lys Lys Val Phe Tyr Val Ile Glu 245 250 255 Thr Arg Glu Pro Arg Glu Arg Leu Leu Leu Thr Ala Ala His Leu Leu 260 265 270 Phe Val Ala Pro His Asn Asp Ser Ala Thr Gly Glu Pro Glu Ala Ser 275 280 285 Ser Gly Ser Gly Pro Pro Ser Giy Gly Ala Leu Gly Pro Arg Ala Leu 35 290 295 300 Phe Ala Ser Arg Val Arg Pro Gly Gin Arg Val Tyr Val Val Ala Glu 305 310 315 320 40 Arg Asp Gly Asp Arg Arg Leu Leu Pro Ala Ala Val His Ser Val Thr 325 330 335 Leu Ser Glu Glu Ala Ala Gly Ala Tyr Ala Pro Leu Thr Ala Gin Gly 340 345 350 Thr Ile Leu Ile Asn Arg Val Leu Ala Ser Cys Tyr Ala Val Ile Glu 355 360 365 Glu His Ser Trp Ala His Arg Ala Phe Ala Pro Phe Arg Leu Ala His 50 370 375 380 Ala Leu Leu Ala Ala Leu Ala Pro Ala Arg Thr Asp Arg Gly Gly Asp 385 390 395 400 Ser Gly Gly Gly Asp Arg Gly Gly Gly Gly Gly Arg Val Ala Leu Thr 405 410 415 Al a His Ser Arg 465 (2) /3-7 Pro Gly Ala Ala Asp Ala Pro Gly Ala Gly Ala Thr Ala Gly Ile 420 425 430 Trp Tyr Ser Gin Leu Leu Tyr Gin Ile Gly Thr Trp Leu Leu Asp 435 440 445 Glu Ala Leu His Pro Leu Gly Met Ala Val Lys Ser Ser Xaa Ser 450 455 460 Gly Ala Gly Gly Gly Ala Arg Glu Gly Ala 470 475 INFORMATION FOR SEQ ID NO:14: SEQUENCE CHARACTERISTICS: LENGTH: 313 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:14: Arg Arg Leu Met Thr Gln Arg Cys Lys Asp A-rg Leu Asn Ser Leu Ala Ile Gly Gly 35 Gly Tyr Ala Leu Arg Leu 145 Ile Ser Trp, Arg 50 Leu Giu Ala Glu Val 130 Ile Giti V-~11 Asp Ala Leu Ser Ala Ser 115 Leu Phe Thr Met Glu Val Ala Lys Lys 100 Gly Ala Leu Gin Asn Asp Asp Arg Ala Thr Al a Met Asp Asp 165 Gin Gly Ile Leu 70 His Gly Arg Gly Arg 150 Pro Pro His 40 Thr Val His Cy s Ala 120 Asp Pro Arg Gly 25 Ser Ser Glu Cys Phe 105 Leu Gly His Arg Val Glu Asp Ala Ser Pro Ser Ser Arg Leu 170 Lys Leu Glu Ser Arg Asp Gly Phe Val Lys Ala Gly Ala Val Pro Thr 140 Leu Arg 155 Ala Leu Arg Leu Arg Asp Ser Ala Arg 125 Phe Ala Thr Val His Asn Trp, Glu Gin 110 Pro Ser Phe Pro Thr Glu Tyr Glu Lys Tyr Val Tyr His Ser Val Arg Gly Asp Asp Val Gin Val 160 Ala His 175 Leu Al a Gly Val.
225 Val.
Leu Trp Leu Leu 305 Leu Phe Thr Phie 195 Val Pro 210 Ala Leu Giu Asp Ala Gin Gly Ser 275 Tyr Arg 290, Gly Met Thr 180 Al a Gly Gly Val.
Leu 260 Trp Leu Ser Ala Asp Ser His Leu Gin Ala Tyr 230 Vai Ala 245 Ala Phe Thr Pro Gly Arg Gly Ala 310 Asn Val Pro 21i5 Ala Ser Trp Gly Leu 295 Gly His Gin 200 Ala Pro Cys Pro Glu 280 Leu Ser Thr 185 Pro Arg Leu Phe Leu 265 Gly Leu Xa~a Giu Pro Gly Gin Vai Ala Thr Lys 235 Ala Ala 250 Arg Leu Val His Glu Glu Al a Tyr Aila 220 His Val Phe Trp Gly 300 Al a Vali 205 Vai Gly Ala His Tyr 28S Ser Arg 190 Leu Ser Thr Asp Ser 270 Pro Phe Phe Val1 Thr Leu His 255 Leu Gin His Arg Al a His Val.
240 His Ala Leu Pro 45 50 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 64 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: peptide FRAGMENT TYPE: internal (xi) Gin 1 His Gly Ile SEQUENCE DESCRIPTION: SEQ ID NO:iS: Aig Cys Lys Asp Lys Leu Asn Set Leu Ala Ile Ser Val Met Asn 5 10 is Tip Pro Gly Val Lys Leu Arg Val Thr Giu Gly Trp Asp Glu Asp 25 His His Phe Glu Glu Ser Leu His Tyr Glu Gly Arg Ala Val Asp 40 Thr Thr Ser Asp Arg Asp Lys Ser Lys Tyr Gly Thr Leu Ser Arg 5560 INFORMATION' FOR SEQ ID NO:16: Wi SEQUENCE CHARACTERISTICS: LENGTH: 65 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: peptide FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ I] Gin Arg Cys Lys Glu Lys Leu Asn 1 5 Met Trp Pro Gly Val Lys Leu Arg Gly Asn His Phe Glu Asp Ser Leu 40 Ile Thr Thr Ser Ser Asp Arg Asp 55 Arg INFORMATION FOR SEQ ID NO:17: SEQUENCE CHARACTERISTICS: LENGTH: 64 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: peptide FRAGMENT TYPE: internal D NO:l16: Ser Leu Ala Ile Ser Val met Asri 10 Val Thr Glu Gly Trp Asp Glu Asp 25 His Tyr Glu Gly Arg Ala Val Asp Arg Asn Lys Tyr Giy Met Phe Ala *aa..a a a (xi) Gin 1 Leu Gly Ile SEQUENCE DESCRIPTION: SEQ ID NO:17: Arg Cys Lys Asp Lys Leu Asn Ser Leu Ala Ile Ser Val Met Asn 5 1.0 Trp Pro Gly Val. Lys Leu Arg Val Thr Giu Gly Trp, Asp Glu Asp 25 Leu His Ser Glu Glu Ser Leu His Tyr Giu Gly Arg Ala Val Asp 40 Thr Thr Ser Asp Arg Asp Arg Asn Lys Tyr Arg Met Leu Ala Arg so 55 INFORMATION FOR SEQ ID NO:lB: Ci) SEQUENCE CHARACTERISTICS: LENGTH: 38 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:18: GGAATTCCCA GCAGNTGCTA AAGGAAGCAA GNGCTNAA 38 INFORMATION FOR SEQ ID NO:19: SEQUENCE CHARACTERISTICS: CA) LENGTH: 33 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:19: TCATCGATGG ACCCAGATCG AAANCONGCT CTC 33 35 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 29 base pairs TYPE: nucleic acid 40 STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID *:GCTCTAGAGC TCNACNGCNA GANCGTNGC 29 INFORMATION FOR SEQ ID, NO:21: Ci) SEQUENCE CHARACTERISTICS: CA) LENGTH: 50 base pairs TYPE: nucleic acid sTR.ANDEDNESS: single (ii) MOLECULE TYPE: cDNA/(I (xi) SEQUENCE DESCRIPTION: SEQ ID NO:21: AGCTGTCGAC GCGGCCGCTA CGTAGGTTAC CGACGTCAAG CTTAGATCTC INFORMATION FOR SEQ ID NO:22: Wi SEQUENCE CHARACTERISTICS: LENGTH: S0 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:22: AGCTGAGATC TAAGCTTGAC GTCGGTAACC TACGTAGCGG CCGCGTCGAC INFORMATION FOR SEQ ID NO:23: Wi SEQUENCE CHARACTERISTICS: LENGTH: 45 base pairs TYPE: nucleic acid STR.ANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:23: GATCGGCCAG GCAGGCCTCG CGATATCGTC ACCGCGGTAT TCGAA INFORMATION FOR SEQ ID NO:24: SEQUENCE CHARACTERISTICS: LENGTH: 30 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: CDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:24: AGTGCCAGTC GGGGCCCCCA GGGCCGCGCc INFORMATION FOR SEQ ID Wi SEQUENCE CHARACTERISTICS: LENGTH: 20 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO: TACCACAGCG GATGGTTCGG INFORMATION FOR SEQ ID NO:26: Ci) SEQUENCE CHARACTERISTICS: CA) LENGTH: 20 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:26: GTGGTGGTTA TGCCGATCGC INFORMATION FOR SEQ ID NO:27: Wi SEQUENCE CHARACTERISTICS: LENGTH: 21 base pairs TYPE: nucleic acid CC)*STRIANDEDNESS: single TOPOLOGY: linear 45 (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:27: TAAGAGGCCT ATAAGAGGCG G 2 INFORMATION FOR SEQ ID NO:28: Ci) SEQUENCE CHARACTERISTICS: LENGTH: 20 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:28: AAGTCAGCCC AGAGGAGACT INFORMATION FOR SEQ ID NO:29: SEQUENCE CHARACTERISTICS: LENGTH: 6 amino acids TYPE: amino aciLd TOPOLOGY: linear (ii) MOLECULE TYPE: peptide FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ ID NO:29: Cys Gly Pro Gly Arg Gly
I
INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: 9 35 LENGTH: 29 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear 40 (ii) MOLECULE TYPE: CDNA *too *V4 (xi) SEQUENCE DESCRIPTION: SEQ ID 0.:AGCAGNTGCT AAAGGAAGCA AGNGCTNAA 29 0*9: C2) INFORMATION FOR SEQ ID N0:31: **so Ci) SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs CB) TYPE: nucleic acid STP.ANDEDNESS: single CD) TOPOLOGY: linear (ii) MOLECULE TYPE: CDNA &6, (xi) SEQUENCE DESCRIPTION: SEQ ID NO:31: CTCNACNGCN AGANCIONGTN GCNA 24 INFORMATION FOR SEQ ID NO:32: SEQUENCE CHARACTERISTICS: LENGTH: 32 base pairs TYPE: nucleic acid STRA2NDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:32: CTGCAGGGAT CCACCATGCG GCTTTTGACG AG '32 INFORMATION FOR SEQ ID NO:33: SEQUENCE CHARACTERISTICS: LENGTH: 31 base pairs TYPE: nucleic acid STRANDEDNE3.i: single TOPOLOGY: linear 66. (ii) MOLECULE TYPE: cDNA (xi) SEQUE'NCE DESCRIPTION: SEQ ID NO:33: 0*3oeCTGCAGGGAT CCT7ATTCCA CACGAGGGAT T 31 40 INFORMATIO14 FOR SEQ ID NO:34: SEQUEKCE CHARACTERISTICS: LENGTH: 471 amino acids 'TYPE: amino acid 45 7oPOLOGY: linear (ii) MOLECULE TYPE: peptide FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ ID NO:34: Met Asp Asn His Ser Ser Val Pro Trp Ala Ser Ala Ala Ser Val Thr 1 5 10 Cys Leu Ser Leu Asp Ala Lys Cys His Ser Ser Ser Ser Ser Ser Ser Ser Gin Thr Pro Arg Glu Asp Leu 145 Arg Trp His Ala Ala 225 Ile Gly Lys Ala Lys Thr 1 Ser Ala Asn Tyr Ser 130 Phe Cys Pro His Thr 210 Val Tyr Cys Pro Asn 290 Ser let Leu His Leu Thr 115 Pro Arg Lys Gly Gly 195 Ser Glu Cys Phe Leu 275 Gly Ala 2 Arg I Val Ser Tyr 100 Asn Lys Asp Glu Ile 180 Gin Asp Ala Ser Thr 260 Gly Gin kla lis la :ys Pro Ser Phe Glu Lys 165 Arg Glu Arg Gly Val 245 Pro Glu Ala Ser Ser Ile Ala 55 Leu Leu 70 Gly Pro Leu Val Ala Ser Lys Asp 135 Glu Gly 150 Leu Asn Leu Leu Ser Leu Asp Gin 215 Phe Asp 230 Lys Ser Glu Ser Leu Ser Val Tyr Ile 40 His Leu Gly Leu Gly 120 Leu Thr Val Val His 200 Ser Trp Asp Thr Ile 280 Ser 25 Ser Thr Ile Arg Lys 105 Pro Val Gly Leu Thr 185 Tyr Lys Val Ser Ala 265 Gly Glu 9i a Gln Val 31y 90 ln Leu Pro Ala Ala 170 Glu Glu Tyr Ser Ser 250 Leu Asp Val Ile Arg Leu 75 Leu Thr Glu Asn Asp 155 Tyr Ser Gly Gly Tyr 235 Ile Leu Arg Ile Pro Cys Pro Gly Ile Gly Tyr 140 Arg Ser Trp Arg Met 220 Val Ser Glu Val Leu 300 Gin Leu Met Arg Pro Val 125 Asn Leu Val Asp Ala 205 Leu Ser Ser Ser Leu 285 Phe Glu Ser Val His Asn 110 Ile Arg Met Met Glu 190 Val Ala Arg His Gly 270 Ser Met Glu Arg Phe Arg Leu Arg Asp Ser Asn 175 Asp Thr Arg Arg Val 255 Val Met Asp Thr Leu Ser Ala Ser Arg Ile Lys 160 Glu Tyr Ile Leu His 240 His Arg Thr Arg 295 Leu Glu Gin Met Asn Phe Val Gin Leu 315 His Thr Asp Gly Gly 320 Ala Val Leu Thr Val Thi- Pro Ala His Leu Val Ser Val Trp Gin Pro 325 330 335 Glu Ser Gin Lys Leu Thi- Phe Val Phe Ala Asp Arg Ile Giu Glu Lys 340 345 350 Asn Gin Val Leu Val Arg Asp Val Glu Thr Gly Giu Leu Arg Pro Gin 355 360 365 Arg Val Val. Lys Val Gly Ser Val Arg Ser Lys Gly Val Val Ala Pro 370 375 380 Leu Thr Arg Glu Gly Th- Ile Val Val Asn Ser Val Ala Ala Ser Cys 385 390 395 400 Tyr Ala Vai Ile Asn Ser G'in Ser Leu Ala His Ti-p Gly Leu Ala Pro 405 410 415 Met Arg Leu Leu Ser Thr Leu Glu Ala Trp Leu Pro Ala Lys Glu Gln 420 425 430 Leu His Ser Ser Pro Lys Val Val Ser Ser Ala Gin Gin Gin Asn Gly 435 440 445 His Trp Tyr Ala Asn Ala Leu Tyr Lys Val Lys Asp Tyr Val Leu 450 455 460 Pro Gin Ser Tip Arg His Asp 30465 470 INFORMATION FOR SEQ ID 110:35: SEQUENCE CHARACTERISTICS: 35(A) LENGTH: 73 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: peptide FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ ID Arg Cys Lys Glu Arg Val Asn Ser Leu Ala Ile Ala Val. Met His Met 1 5 10 is Ti-p Pro Gly Val. Axg Leu Arg Val Thi- Glu Gly Ti-p Asp Giu Asp Gly 25 His His Leu Pro Asp Ser Leu His Tyr Glu Gly Arg Ala Leu Asp Ile 40 Thi- Th- Ser Asp Arg Asp Arg His Lys Tyr Gly Met Leu Ala Arg Leu 5560 167 Ala Val Glu Ala Gly Phe Asp Trp Val INFORMATION FOR SEQ ID NO:36: SEQUENCE
CHARACTERISTICS:
LENGTH: 73 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: peptide FRAGMENT TYPE: internal 25 (xi) Arg 1 Trp His Thr SEQUENCE DESCRIPTION: SEQ ID NO:36: Cys Lys Asp Lys Leu Asn Ala Leu Ala Ile Ser Val Met Asn Gln 5 10 Pro Gly Val Lys Leu Arg Val Thr Glu Gly Trp Asp Glu Asp Gly 25 His Ser Glu Glu Ser Leu His Tyr Glu Gly Arg Ala Val Asp Ile 35 40 Thr Ser Asp Arg Asp Arg Ser Lys Tyr Gly Met Leu Ala Arg Leu n 55 Ala Val Glu Ala Gly Phe Asp Trp Val 40 45 INFORMATION FOR SEQ IL NO:37: SEQUENCE
CHARACTERISTICS:
LENGTH: 64 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: peptide FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ ID NO:37: Lys Arg Cys Lys Glu Lys Leu Asn Val Leu Ala Tyr Ser Val Met Asn 1 5 10 Glu Trp Pro Gly Ile Arg Leu Val Val Thr Glu Ser Trp Asp Glu Asp 20 25 Tyr His His Gly Gln Glu Ser Leu His Tyr Glu Gly Arg Ala Val Thr A 40 168 Ile Ala Thr Ser Asp Arg Asp Gin Ser Lys Tyr Giy Met Leu Ala Arg 55 INFORMATION FOR SEQ ID NO:38: SEQUENCE CHARACTERISTICS: LENGTH: 28 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:38: AAAAGCTTTA YTGYTAYGTN GGNATHGG 28 INFORMATION FOR SEQ ID NO:39: Ci) SEQUENCE CHARACTERISTICS: LENGTH: 28 base pairs TYPE: nucleic acid STR.ANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cD NA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:39: AAGAATTCTA NGCRTTRTAR TTRTTNGG 28 INFORMATION FOR SEQ ID Ci) SEQUENCE CHARACTERISTICS: LENGTH: 165 amino acids TYPE: amino acid CD) TOPOLOGY: linear ii) MOLECULE TYPE- peptide FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ ID Cys Gly Pro Giy Arg Gly Xaa Gly Xaa Arg Arg His Pro Lys Lys Leu 1 5 10 Thr Pro Leu Ala Tyr Lys Gin Phe Ile Pro Asn Val Al~a Glu Lys Thr 25 Leu Gly Arg Phe Asp Glu Asp LYS Val Xaa Glu Glu Asp Arg 130 Ala Gly 145 Ser Val.
Al a Lys Glu Leu Leu Ser Asp Phe Lys Ser Glu Asn Asn Arg 100 Leu Xaa Asp Ala Gly Leu Thr Xaa Val His Ser Trp Glu Arg Thr Gay 70 Lieu Thr Tyr Lys Val 2.50 Tyr Pro 55 Al a Ala Glu Glu Tyr 135 Tyr Glu 40 Asn Asp Ile Gly Gly 120 Gly Tyr Gly Lys Tyr Asfl Arg Leu Ser Val 90 Trp Asp 105 Arg Al a Xaa Leu Glu Ser Ile Pro Met 75 Met Glu Val Xaa Lys 155 Xaa Asp Thr Asn Asp Asp Arg 140 Ala Arg Ile Gin Xaa Gly Ile 125 Leu His Asn Ile Arg Trp His 110 Thr Ala Ile Ser Phe Cys Pro His Thr Va.
His Glu Lys Lys Gly Xaa Ser Giu Cys 160 INFORMIATION FOR SEQ ID NO:41: Wi SEQUENCE CHARACTERISTICS: LENGTH: .167 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: peptide FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ ID NO:41: Cys Gly Pro Gly Arg Gly Xaa Xaa Xaa Arg 15 10 Xaa Leu Xaa Pro Leu Xaa Tyr Lys Ginx Phe 25 Xaa Thr Lieu Gly Ala Ser Gly Xaa Xaa Glu 35 40 Ser Glu Arg Phe Xaa Xaa lieu Thr Pro Asn s0 55 Phe Lys Asp Glu Glu Asn Xaa Gly Ala Asp 70 Arg Xaa Xaa Xaa Pro Xaa Gly Xaa Xaa Tyr Asn Pro Arg Leu Met 75 Xaa Pro Lys Xaa Xaa Glu Xaa Arg Xaa Asp Ile Ile Thr Xaa Arg s0 Cys Lys Xaa Xaa Xaa Asn Xaa Leu 1 76 Ala Ile 90 Ser Val Met Asn Xaa Trp Pro Gly Val His Xaa Xaa 115 Leu Arg Val Thr Giu 105 Gly Xaa Asp Glu Asp Gly His 110 Asp Ile Thr Xaa Ser Leu His Tyr Glu Gly Arg Ala Xaa 125 Thr Ser 130 Asp Arg Asp Xaa Xaa 135 Lys Tyr C-iy Xaa Leu 140 Xaa Arg Leu Ala Glu Ala Gly Phe Trp 1Val Tyr Tyr Giu 155 Ser Xaa Xaa His His Xaa Ser Val Lys Xaa Xaa INFORMATION FOR SEQ ID NO:42: SEQUENCE CHARACTERISTICS: LENGTH: 3900 base pairs TYPE: nucleic acid STRANDEDNESS: both TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
ATG
Met 40 1 (ix) FEATURE: NAME/KEY: CDS LOCATION: 1. .3897 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:42: GAC CGC GAC AGC CTC CCA CGC GTT Asp Arg Asp Ser Leu Pro Arg Val CCG GAC Pro Asp 10 ACA' CAC GOC Thr His (fly GAT GTG Asp Val GTC GAT GAG Val Asp Glu TTA TTC TCG Leu Phe Ser GAT CTT Asp Leu 25 TAC ATA CGC ACC Tyr Ile Arg Thr GAC GCC CAA GTG GCG CTC CAT CAG ATA CAT AAG GGC AAA Asp Ala Gin Val Ala Leu Asp Gin Ile Asp Lys Gly Lys 40 AGC CGC ACG GCG ATC TAT CTG CGA TCA GTA TTC CAG TCC Ser Arg Thr Ala Ile Tyr Leu Arg Ser Val Phe Gin Ser s0 55 AGC J'GG GTG Ser Trp Val GCG CGT GGC Aia Arg Gly CAC CTC GAA His Leu Giu CTA TTC GTG Leu Phe Val 192 ACC CTC Thr Leu GGC AGC TCC GTG CAA AAG CAC OCG GGC AAG GTG Gly Ser Ser Vai Gin Lys His Ala Gly Lys Val 70 75 240 GCT ATC CTG GTG Ala Ile Leu Val AGO ACC TTC TGC Ser Thr Phe Cys '71
GTC
Val GGC CTG AAG AGC Gly Leu Lys Ser GCC CAG Ala Gin ATC CAC TCC Ile His Ser GAG GCG GAA Glu Ala Glu 115
AAG
Lys 100 GTG CAC CAG CTG Val His Gin Leu
TGG
Trp 105 ATC CAG GAG Ile Gin Giu CTG GCC TAO ACA CAG AAG ACG ATC GGC Leu Ala Tyr Thr Gin Lys Tbr Ilie Gly 120 GGC GGC GGG CTG Gly Gly Gly Leu 110 GAG GAC GAG TCG Giu Asp 0-lu Ser 125 CCG AAC GCC TCC Pro Asn Ala Ser 384 GCC ACG Ala Thr 130 CAT CAG CTG CTC His Gin Leu Leu CAG ACG ACC CAC Gin Thr Thr His 432 0-TO Val 145 CTG CAT CCG CAG GCG CTG CTT GCC CAC Leu His Pro Gin Ala Leu Leu Ala His 150
OTG
Leu 155 GAG GTC CTG 0-TO Giu Val Leu Val
AAG
Lys 160 GCC ACC GCC GTC Ala Thr Ala Val GTG CAC 070 TAC Vai His Leu Tyr
GAC
Asp 170 ACC GAA TGG GGG Thr Glu Trp, Gly CTG CGC Leu Arg 175 GAC ATG TGC Asp Met Cys ATC GAG CAG 30 Ile Glu Gin 195 ATG CCG AGC ACG Met Pro Ser Thr TCC TTC GAG GG0C Ser Phe Glu Gly ATC TA.C TAC Ile Tyr Tyr 190 ATC ACG CCG Ile Thr Pro 576 ATC CTG CGC CAC Ile Leu Arg His
CTC
Leu 200 ATT CCG TGC TCG Ile Pro'Cys Ser CTG GAO Leu Asp 210 TGT TTC TGG GAG Cys Phe Trp Giu
GGA
Gly 215 AGO CAG CTG TTG Ser G1.n Leu Leu COG GAA 'rCA GO Pro Giu Ser Ala 555...
555.
GTC
Val 225
CCC
Pro GTT ATA COA GGC Val Ile Pro Gly
CTC
Leu 230 AAC CAA CGA O-TZ1 Asn Gin Arg Leu
CTG
LeU 235 TGG ACC ACA CTG Trp Thr Thr Leu
AAT
Asn 240 GCC TOT GTG Ala Ser Val
ATG
Met 245 CAG TAT ATO AAG Gin Tyr Met Lys
CAG
Gin 250 AAG ATG TOO GAG Lys Met Ser 0-lu GAA AAG Giu Lys 255 ATO AGO T70 Ile Scr Phe ATT CG AGT Ile Aia Ser 275 TTC GAG ACC GTG Phe Glu Thr Val CAG TAO ATG AAG Gin Tyr Met Lys CT GOG 0-00 Arg Ala Ala 270 CTG AAT CCC Leu Asn Pro 0GOC TAO ATG GAG Gly Tyr Met 0-lu
AAG
Lys 280 COO TGC OTO AAC Pro Cys Leu Asn AAT TGC Asn Cys 290 COG GAO ACG 0-CA Pro Asp Thr Ala
CG
Pro 295 AAO AAG AAC AGO Asn Lys Asn Ser CAG COG COG GAT Gin Pro Pro Asp 912 GGA GCC ATC CTG Gly Ala Ile Leu
TCC
Ser 310 GGA GGC TGC Gly Gly Cys I7X
TAC
Tyr TAT GCC GCG AAG CAC 960 Tyr Ala Ala Lys His 320 ATG CAC TGG CCG Met His Trp Pro
GAG
Giu 325 GAG CTG ATT GTG GGC GGA GCG AAG AGG Giu Leu Ile Val Gly Giy Ala Lys Arg 330 AiX CGC As~n Arg 3 1.008 AGC GGA CAC Ser Gly His ATG ACC GAG Met. Thr 0 u 355
TTG
Lieu 340 AGG AAG GCC CAG Arg Lys Ala Gin
GCC
Al a 345 CTG CAG TCG GTG Leu Gin Ser Val GTG CAG CTG Val O:.n Leu 350 TAC AhG GTG Tyr Lys Val i1056 1104. AAG GAA ATG TAC Lys Glu Met Tyr
GAC
Asp 360 CAG TOG CAG GAC Gin Trp Gin Asp CAC. CAT His His 370
TGCAG
Trp.Gin CTT GGA TOG ACG Lieu Gly Trp Thr GAG AAG GCA GCG Giu Lys Ala Ala
GAG
Giu 380 GTT TTG AP.C GCC Val Lieu Avn Ala COC AAC TTT bArg Azn Phe ATT GCC ACC Ile Ala Thr 405 CGG GAG GTC GAA CAG Arg Giu Vai Oiu Gin CTG CTA COT Al-A Leu--Leu Arg Lys-
CAG
Gi-n 400 2.15;~: 120Ot.
1248 TCG.AGA, Ser JArg AAC TAC GAT ATC Asn Tyr Asp Ile
TAC
Tyr 410 .GTG TTC7 AGC TCG Vai Phe Ser Ser GCT GCA A" a Ala CTrG OAT GAC 30 ieu;, Asp Asp GTC. A'C GOC Val Tie Gly 435 CTG 0CC AAG TTC Lieu Ala Lys Phe
TCC
Ser 425 CAT ,CCC AOC GCC His Pro Ser Ala TTG TCIC ATT Lieu Ser Ile 430 ACG CTC Thr Lieu Lieu 1296 1344 GTG 0CC GTC ACC Val Ala Val Thr
OTT
Vai 440 TTO TAT-GCC TTC Leu Tyr Ala.Phe
TGC
Cys 445 COC TG Arg. Trp 450 AOG GXC CCC GTC Arg Asp Pro Val
COT
Arg 455 OGA CAG AGC AGT GTC GGC. GTG OC 7GG Gly Gin Ser Ser Vai -Gly Val Alai Gly 1392 1440 *0 0 0
CTT
Val 465 CTG CTC ATG TGC Leu Leu Met Cys
TTT
Phe 470 AGT ACC 0CC 0CC Ser Thr Ala Ala
GGA
Gly 475 TTG GGA TTG-TCA' Lieu Giy Lieu Ser:
GCC
Ala 480 45 CTG CTC GOT ATC OTT TTC AAT 0CC Lieu Lieu Oly Ile Val Phe Asn Ala 485 GCC AGC Ala Ser 490 ACC CAG GTG OTT Thr Gin Val Val CCG TTT Pro Phe 495 1488 TTO 0CC CTT Lieu Ala Lieu CTO GOC GTC GAT Lieu Oly Val Asp ATC TTC ATG CTG Ile Phe Met Lieu ACC OCT 0CC Thr Ala Ala 510 CTC AAG AAA Lieu Lys Lys 1536 1584 TAT OCG GAG AGC AAT CGO COG GAG Tyr Ala Glu Ser Asn Arg Arg Oiu 515 520 CAG ACC AAO CTG Gin Thr Lys Lieu
ATT
Ile 525 /73 GTG GGA CCG AGC ATC CTG TTC AGT GCC TGC Val Gly 530 Pro Ser Ile Leu Phe Ser Ala Cys 535 AGC ACC Ser Thr 540 GCA GGA TCC TTC Ala Gay Ser Phe 1632
TTT
Phe 54-5 GCG GCC GCC TTT Ala Ala Ala Phe ATT CCG GTG Ile Pro Val 550 CCG GCT TTG AAG GTA TTC Pro Ala Leu Lys Val Phe 555 TGT CTG Cys Leu 560 1680 CAG GCT GCC ATC Glh Ala Ala Ile
GTA
Val 565 ATG TGC TCC AAT Met Cys Ser Asn
TTG
Leu 570 GCA GCG GCT Ala Ala Ala TTT CCG GCC Phe Pro Ala GO GAC ATC Al a Asp Ile 595 ATr TCG TTG OAT Ile Ser Leu Asp
CTA
Leu 585 COG AGA CGT ACC Arg Arg Arg Thr CTA TTG GTT ILeu Leu Val 575 .GCC GGC AOG Ala Gly Arg 590 CAG CCG AAG Gin Pro Lys TTC tGC TGC TGT Phe Cys Cys Cys
TTT
Phe 600 CCG GTG.TGG AAG Pro Val'Trp, Lys AAC AAC AAC AAC Asn Asn Asn Asn 620
GAA
Glu 605 GTG GCA Vai Ala 610 CCA CCG GTG CTG Pro Pro Val Leu CCG CTG Pro Leu 61is AAC AAC Asn Asn GGG CGC GGG GCC Gly Arg Gly Ala
C.
C
CG
Arg 62' i CAT CCG AAG AGC His Pro Lys Ser
TGC
Cys 630
AAC
Asn AGG GTG Arg Val 635 GCG CTG CCC GCC Ala Leu Pro Ala
CAG
Gin 640 ATCCT CTG CTG 30 Asni Pro Leu Leu
OAA
G iu 645 CAG AGG GCA GAC Gin Arg Ala Asp
ATC
Ile 650 CCT GGG AGC AGT Pro Gly Ser Ser CAC TCA His Ser 655 1728 1776 1824 1872 1920 1968 201i6 2064' 2112 2160 2208 2256 CTG OCO TCC Leui Ala Ser
TTC
Phe 660 TCT CTG GCA ACA Ser Leu Ala Thr
TTC
Phe 665 GCC TTT CAG CAC Ala Phe Gin His TAC ACT CCC Tyr Thr Pro 670 OGT :TTC CTG Oly Phe Leu
*CCCCC
TT C CT .C Pkhe Leu GCdG0CC Ala Ala 690
ATG
Met 675 COC AGC TGG OTO Arg Ser Trp Val
AAG
Lys 680 TTC CTG C OTT Phe Leu Thr Val
ATG
Met 685 CTC ATA TCC AGC Leu Ile Ser Ser TTG TAT Leu Tyr 695 0CC TCC ACG Ala Ser Thr
CGC
Arg 700 CTT CAG GAT GGC Leu Gin-Asp Gly
CTG
Leu 705 GAC ATT ATT OAT Asp Ile Ile Asp GTG CCC AAG GAC Val Pro Lys Asp
AGC
Ser 715 AAC GAG CAC AAO Asn Oiu His Lys CTG GAT OCT CAA Leu Asp Ala Gin CGO CTC TTT GOC Arg Leu Phe Gly
TTC
Phe 730 TAC AGC ATG TAT Tyr Ser Met Tyr GCG OTT Ala Val 735 ACC CAG C Thr Gin Gly
AAC
Asn 740 TTT GAP. TAT CCC ACC CAG CAG CAG TTG Phe Oiu Tyr Pro Thr Gin Gin Gin Leu 745 CTC AOG GAC Leu Arg Asp 750 TAC CAT GAT TPyr His Asp 755 TCC TTT GTG CGG Ser Phe Val Arg
GTG
Val 760 CCA CAT GTG ATC Pro His Val Ile
AAG
Lys 765 AAT GAT AAT Asn Asp Asn 2304 2352 GGT GGA Gly Gly 770 CTG CCG GAC TTC Leu Pro Asp Phe CTG CTG CTC TTC Leu Leu Leu Phe
AGC
Ser 780 GAG TGG CTG GGT Glu Trp Leu Gay AAJ7 Asa 78S~ CTG CAA AAG ATA Leu Gin Lys le GAC GAG GAA TAC Asp Giu Glu Tyr GAC GGA CGG CTG Asp Gay Arg. Leu
ACC
Thr 800 2400 2448 AAG GAG TGC TGG Lys Giu Cys Trp TTC CCA AAC GCC AGC AGC Phe Pro Asn Aia Ser Ser 805 5 10 GAT GCC ATC CTG A~sp Ala Ile Leu GCC TAC Ala-Tyr 815 AAG CTA ATC Lys Leu Ile CTG GTG CTC Leu Val Leu 835 cAA P.CC GGC CAT Gin Thr Gly His
GTG
Val 825 GAC 4AC CCC GTG Asp Asn Pro Val GAC AAG GAA Asp Lys 'Glu 830 ATC AAC CAA Ilie Asn Gin 2496 2544 ACC AAT CGC CTG GTC AAC AGC GAT GGC Thr Asn Arg Leu Val Asn Ser Asp Gly 840 CGC GCC Arg Ala 850 TTC TAC AAC TAT Pkie Tyr Asn Tyr
CTG
Leu 855 TCG GCA TGG A'CC Ser Ala Trp, 2la
ACC
Thr 560 AAC GCG, TCT.TCG Asn Ala' Ser. Ser CCT ACG 30 Pro Thr 865 GAG CTT CTC Giu Leu iLeu GCA AAT TGT ATC Ala Azn Cys Ile AAC CGC GCC Asn Arg Ala AAC ,GGA Asri..Gay ;880 GCT TCT CAG GGC Ala Ser Gin Gly
AAA
Lys 885 TTG TAT CCG GAA Leu Tyr Pro Glu
CCG
Pro 890 CGC CAG TAT Arg Gin Tyr *CCC AAC GAG TAC GAT C-TT AAG ATA Pro Asn Giu Tyr Asp Leu Lys Ile 900 AAG AS.T CTG CCA Lys Ser Leu Pro TTTf CAC CAA Phe His.Gin 895 TTG. GTCTAC Leu:-Val Tyr 910 TCG -CAG.ATC Ser Gin'Ile 2592 2640 26,88 2736 2784 2832 GCT CAG ATG Ala Gin Met 915 CCC TTT TAC CTC Pro Phe Tyr Leu GGA CTA ACA GAT Gly Leu Thr Asp 45 AAG ACC Lys Thr 930 CTG ATA GGT CAT Leu Ilie Gly His CGC GAC CTG AGC Arg Asp Leu Ser AAG TAC GAG GGC Lys Tyr Glu Gly
TTC
Phe 945 GGC CTG CCC AAC Giy Leu Pro Asn CCA TCG GGC ATT Pro Ser Gly Ile TTC ATC TTC Phe Ile Phe TGG GAG Trp Giu 960 TGC GTG Cys Val 975 2880 2928 CAG TAC ATG ACC Gin Tyr Met Thr CTG CGC TCC TCA CTG GCC Leu Arg Ser Ser Leu Ala 965 970 ATG ATC CTG GCC Met Ile Leu Ala CTA CTC GCC Leu Leu Ala GCC GCC GTT Ala Ala Val 995 TTT GGG GCC Phe Gly Ala 1010 GCC CTG GTG CTG GTC TCC Ala Leu Val Leu Val Ser 980 985 CTC GTG ATC CTC AGC GTT Leu Val. Ile Leu Ser Val 1000 175.
CTG CTC CTG CTC TCC GTT TG Leu Leu Leu Leu Ser Val Trp 990 CTG GCC TCG CTG, GCC CAG ATC Leu Ala Ser Leu Ala Gin Ile 1005 2976 3024 ATG ACT CTG CTG GGC Met Thr Leu Leu Gly 1015 ATC AAA CTC TCG Ile Lys Leu Ser 1020 GTC ATA Val Ile 1025 CTC ATC CTC Leu Ile Leu AGC GTG Ser Val 1030 GGC ATG ATG Giy Met Met CTG TGC Leu Cys 1035 GCC ATT CCG GCA Ala Ile Pro Ala TTC AAT GTG CTG Phe Asn Val Leu 1040 CAG CGC CGC GTC Gin Arg Arg Val 1055 3072 3120 3168 3216 ATA TCA CTG GGC Ile Ser Leu Gly TTC ATG ACA Phe Met Thr 1045 TCC GTT GGC AAC CGA Ser Val Gly Asn Arg 1050 CAG CTG AGC ATG CAG ATG TCC CTG GGA CCA CTT GTC CAC GGC ATG CTG Gin Leu Ser Met Gin Met Ser Leu Giy Pro Leu Val His Giy Met Leu 1060 1065 1070 ACC TCC GGA GTG GCC GTG TTC ATG CTC TCC ACG TCG CCC TTT GAG TTT Thr Ser Gly Val Ala Val Phe Met Leu Ser Thr Ser Pro Phe Giu Phe 3264 1075 1080 1095 I* GTG ATC CG 30 Val Ile Arg 1090 CAC TTC TGC His Phe Cys TOG CTT Trp Leu 1095 CTG CTG GTG GTC TTA TGC GTT GGC Leu Leu Val Val .Leu Cys Val Gly 1100 3312 3360 GCC TGC Ala Cys 13.05 AAC AGC CTT Asn Ser Leu TTG GTG Leu Val 1110 TTC CCC ATC Phe Pro Ile CTA CTG Leu Leu i115 AGC ATG GTG Ser Met Val
GGA
Gly 1120 CCG GAG GCG GAG Pro Glu Ala Glu CTG GTG Leu Val 1125 CCG CTG GAG Pro Leu Giu CAT CCA His Pro 1130 GAC CGC ATA Asp Arg Ile TCC ACG Ser Thr 1135 CCC TCT CCG CTG CCC GTG CGC AGC Pro Ser Pro Leu Pro Val Arg Ser 1140 AGC AAG AGA Ser Lys Arg 1145 45 GTG GTG CAG GGA TCG Val Val Gin Gly Ser 1155 CAC CAC CAC CAC AAA His His His His Lys 1170 GAG GAG CCG CAG TCG Glu Glu Pro Gin Ser l185 CGA TCC TCG CGA GGC AGC Arg Ser Ser Arg Gly Ser 1160 GAC CTT AAT GAT CCA TCG Asp Leu Asn Asp Pro Ser 1175 TCG GGC AAA TCC TAT Ser Gly Lys Ser Tyr 1150 TOC CAG AAG TCG CAT Cys Gin Lys Ser His 1165 CTG ACO ACG ATC ACC Leu Thr Thr Ile Thr 1180 3408 3456 3504 3552 TGG AAG TCC AGC AAC TCG TCC ATC CAG ATG Trp Lys Ser Ser Asn Ser Ser Ile Gin Met 1190 1195 ccc Pro 1200 3600 76 AAT GAT TGG ACC TAC CAG CCG CGG GAA CAG CGA CCC GCC TCC TAC GCG Asn Asp Trp, Thr Tyr Gin Pro Arg Giu Gin Arg Pro Ala Ser Tyr Ala 1205 1210 1215 3648 GCC CCG CCC CCC GCC Ala Pro Pro Pro Ala 1220 TAT CAC AAG GCC GCC GCC CAG CAG Tyr His Lys Ala Ala Ala Gin Gin 1225 CAC CAC CAG His His Gin 1230 3696 CAT CAG GGC CCG His Gin Gay Pro 1235 CCC ACA ACG Pro Thr Thr CCC CCG Pro Pro 1240 CCG CCC TTC Pro Pro Phe CCG ACG GCC TAT Pro Thr Ala Tyr 1245 3744 CCG CCG GAG Pro Pro GJlu 1250 CTG CAG AGC ATC GTG GTG CAG CCG Leu Gin Ser Ile Val Val Gin Pro 1255 GAG GTG Giu Val 1260 ACG GTG GAG Thr Val. Giu 3792 ACG ACG Thr Thr 1265 CAC TCG GAC His Ser Asp AGC AAC ACC Ser Asn Thr 1270 ACC AAG GTG ACG Thr Lys Val Thr 1275 GCC ACG 0CC Ala Thr Ala
AAC
Asn 1280 3840 ATC AAG GTG GAG CTG GCC ATG CCC GGC Ile Lys Val Giu Leu Ala Met Pro Gly 1285 AGG GCG Arg Ala 1290 GTG CGC AGC Vai Arg Ser TAT AAC Tyr Asn 1295 3888 3900 0 0.
00..
TTT ACG ACT TAG Phe Thx Ser INFORMATION FOR SEQ ID NO:43: Ci) SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:43: ACCGAGGGCT GGGACGAAGA TGGC *5 S 0
S.
S INFORMATION FOR SEQ ID NO:44: SEQUENCE CHARACTERISTICS: LENGTH: 25 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: CDNA /77 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:44: CGCTCGGTCG TACGGCATGA ACGAC INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 27 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID ATGGGGATGT GTGTGTGGTC AAGTGTA 27 INFORMATION FOR SEQ ID NO:46: SEQUENCE CHARACTERISTICS: LENGTH: 25 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:46: TTCACAGACT CTCAAAGTGT ATTTT INFORMATION FOR SEQ ID NO:47: SEQUENCE CHARACTERISTICS: LENGTH: 18 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: peptide FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ ID NO:47: Met Gly Ser Ser His His His His His His Leu Val Pro Arg Gly Ser 1 5 1o His Met

Claims (73)

1. An isolated nucleic acid encoding a polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
2. The nucleic acid of claim 1, wherein the portion thereof is selected from residues
27-425 of SEQ ID No: 8, residues 22-396 of SEQ ID No: 9, residues 1-336 of SEQ ID No: 10, residues 25-437 of SEQ ID No: 11, residues 24-418 of SEQ ID No: 12, residues 24-475 of SEQ ID No: 13, and residues 1-312 of SEQ ID No: 14. 3. The nucleic acid of claim 1, wherein said hedgehog amino acid sequence corresponds to an extracellular fragment of a hedgehog protein having an approximate molecular weight of 19 kD. 4. The nucleic acid of claim 1, wherein said hedgehog amino acid sequence is all •or a portion of a naturally occurring Sonic hedgehog protein. 5. The nucleic acid of claim 4, wherein said Sonic hedgehog protein is a mammalian Sonic hedgehog protein. 6. The nucleic acid of claim 5, wherein said Sonic hedgehog protein is a human Sonic hedgehog protein. 7. The nucleic acid of claim 1, wherein said hedgehog amino acid sequence includes at least 150 amino acid residues of the N-terminal half of a hedgehog protein. P:\OPER\Fas\247389)-clns.doc-02/11/01 179 8. The nucleic acid of claim 1, wherein said hedgehog amino acid sequence is at least 80% identical to residues 1-336 of SEQ ID No: 9. The nucleic acid of claim 1, wherein said polypeptide binds to a patched protein. The nucleic acid of claim 9, wherein said patched protein is a patched protein of a vertebrate organism. 11. The nucleic acid of claim 1, wherein said polypeptide regulates proliferation of chondrocytes. 12. The nucleic acid of claim 1, wherein said hedgehog amino acid sequence is all or a portion of a naturally occurring Indian hedgehog protein. S 10 13. The nucleic acid of claim 12, wherein said Indian hedgehog protein is a mammalian Indian hedgehog protein. 14. The nucleic acid of claim 13, wherein said Indian hedgehog protein is a human Indian hedgehog protein. The nucleic acid of claim 1, wherein said polypeptide induces expression of a BMP-2, BMP-4, Isletl, Paxl or Hoxd gene. 16. The nucleic acid of claim 1, wherein said polypeptide promotes differentiation of neuronal cells or survival of differentiated neuronal cells. o* 17. The nucleic acid of claim 16, wherein said neuronal cell is a dopaminergic neuron. 18. The nucleic acid of claim 16, wherein said neuronal cell is a motor neuron. 19. The nucleic acid of claim 1, wherein the hedgehog amino acid sequence includes at least 150 contiguous amino acids of an extracellular domain of the hedgehog protein. P:\OPER\Fas\2473890-clms.doc-02/1 1/01 180 The nucleic acid of claim 1, wherein the hedgehog amino acid sequence includes at least 100 contiguous amino acids of an extracellular domain of the hedgehog protein. 21. The nucleic acid of claim 1, wherein the hedgehog amino acid sequence includes at least 50 contiguous amino acids of an extracellular domain of the hedgehog protein. 22. An isolated nucleic acid which hybridizes under highly stringent conditions to a nucleic acid sequence selected from SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, which nucleic acid encodes a hedgehog amino acid sequence that binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila. 23. The nucleic acid of claim 22, wherein said hedgehog amino acid sequence is 0* encoded by a nucleic acid which hybridizes under stringent conditions to a sequence 15 selected from residues 310-567 of SEQ ID NO: 1, residues 304-561 of SEQ ID NO: 4, residues 301-558 of SEQ ID NO: 5, and residues 304-561 of SEQ ID NO: 6. 24. The nucleic acid of claim 22, wherein said hedgehog amino acid sequence is encoded by a nucleic acid which hybridizes under stringent conditions to a sequence selected from residues 64-567 of SEQ ID NO: 1, residues 73-561 of SEQ ID NO: 4, residues 70-558 of SEQ ID NO: 5, and residues 73-561 of SEQ ID NO: 6. a.. The nucleic acid of claim 22, wherein said hedgehog amino acid sequence is encoded by a naturally occurring Sonic hedgehog gene. 26. The nucleic acid of claim 25, wherein said Sonic hedgehog gene is a mammalian Sonic hedgehog gene. P:\OPER\Fas2473800-cIms.doc4)2/1 1/1 181 27. The nucleic acid of claim 26, wherein said Sonic hedgehog gene is a human Sonic hedgehog gene.
28. The nucleic acid of claim 26, wherein said hedgehog amino acid sequence is encoded by at least a portion of a Sonic hedgehog gene of vertebrate origin corresponding to residues 64-567 of SEQ ID NO: 1, residues 73-561 of SEQ ID NO: 4, residues 70-558 of SEQ ID NO: 5 and residues 73-561 of SEQ ID NO: 6.
29. The nucleic acid of claim 22, wherein said hedgehog amino acid sequence corresponds to an extracellular fragment of a hedgehog protein having an approximate molecular weight of 19 kD.
30. The nucleic acid of claim 22, wherein said hedgehog amino acid sequence includes at least 150 amino acid residues of the N-terminal half of a hedgehog protein.
31. The nucleic acid of claim 22, wherein said hedgehog amino acid sequence is encoded by a nucleic acid which hybridizes under stringent conditions to residues 81- 594 of SEQ ID NO: 3. 15 32. The nucleic acid of claim 22, wherein said hedgehog amino acid sequence is encoded by a nucleic acid which hybridizes under stringent conditions to residues 1-594 of SEQ ID NO: 3. 5 0
33. The nucleic acid of claim 22, wherein said hedgehog amino acid sequence is encoded by a naturally occurring Indian hedgehog gene. *0
34. The nucleic acid of claim 33, wherein said Indian hedgehog gene is a mammalian Indian hedgehog gene. The nucleic acid of claim 34, wherein said Indian hedgehog gene is a human Indian hedgehog gene. P:\OPER\Fs\2473890-clnms.doc-02/1 l/0 182
36. The nucleic acid of claim 22, wherein said polypeptide binds to a patched protein.
37. The nucleic acid of claim 36, wherein said patched protein is a patched protein of a vertebrate organism.
38. The nucleic acid of claim 22, wherein said polypeptide promotes differentiation of neuronal cells or survival of differentiated neuronal cells.
39. The nucleic acid of claim 38, wherein said neuronal cell is a dopaminergic neuron. The nucleic acid of claim 38, wherein said neuronal cell is a motor neuron.
41. The nucleic acid of claim 22, wherein said polypeptide regulates proliferation of chondrocytes.
42. The nucleic acid of claim 22, wherein said polypeptide induces expression of a BMP-2, BMP-4, Isletl, Paxl or Hoxd gene.
43. The nucleic acid of claim 22, wherein the hedgehog amino acid sequence 15 includes at least 150 contiguous amino acids of an extracellular domain of the hedgehog protein. 9999* S44. The nucleic acid of claim 22, wherein the hedgehog amino acid sequence includes at least 100 contiguous amino acids of an extracellular domain of the hedgehog protein.
45. The nucleic acid of claim 22, wherein the hedgehog amino acid sequence includes at least 50 contiguous amino acids of an extracellular domain of the hedgehog protein. -183-
46. A method for inducing a cell to differentiate to a neuronal cell phenotype, including contacting said cell with a polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to apatched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
47. The method of claim 46, wherein said neuronal cell phenotype is selected from motor neurons, cholinergic neurons, dopaminergic neurons, serotonergic neurons, and peptidergic neurons.
48. A method of modulating skeletogenesis including contacting a target tissue with an amount of a polypeptide effective to cause one or both of chondrogenesis and osteogenesis in the target tissue, said polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated 20 neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
49. The method of claim 48, wherein said target tissue is selected from bone, connective tissue, and a combination thereof. 25 50. A method for treating a degenerative disorder of the nervous system characterized by neuronal cell death, including administering to a patient a therapeutically effective amount of a pharmaceutical preparation of a polypeptide, thereby causing, relative to the absence of treatment, prolonged survival of neural cells 00 5002302061 .Doc/BSW P:\OPER\Fas\2473890-clms.doc4)2/1 1/01 184 in said patient, said polypeptide including a hedgehog amino acid sequence at least identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
51. The method of claim 50, wherein said therapeutically effective amount of hedgehog polypeptide inhibits the de-differentiation of neural cells of said patient.
52. The method of claim 51, wherein said neural cells include glial cells.
53. The method of claim 51, wherein said neural cells include nerve cells.
54. The method of claim 50, wherein said degenerative disorder is a neuromuscular disorder.
55. The method of claim 50, wherein said degenerative disorder is an autonomic disorder.
56. The method of claim 50, wherein said degenerative disorder is a central nervous system disorder. .i 57. The method of claim 50, wherein said degenerative disorder is selected from Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Pick's disease, Huntington's disease, multiple sclerosis, neuronal damage resulting from anoxia- ischemia, neuronal damage resulting from trauma, and neuronal degeneration associated with a natural aging process. P:\OPER\Fas\247389(0-lms.doc-02/1 1/01 185
58. A method of claim 50, further including administering to said patient a therapeutically effective amount of a growth factor having neurotrophic activity.
59. The method of claim 58, wherein said growth factor is selected from a nerve growth factor, cilliary neurotrophic growth factor, schwanoma-derived growth factor, glial growth factor, striatal-derived neuronotrophic factor and platelet-derived growth factor. A method for modulating one or more of growth, differentiation and survival of a neuronal cell, including contacting said cell with an effective amount of a polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of i chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
61. A method for promoting survival of mammalian neuronal cells responsive to hedgehog induction, including treating the cell with an amount of a hedgehog polypeptide effective to increase the rate of survival of the neuronal cells, said 20 polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila. P:\OPER\Fs\2473890-cms,.doc-02/ l]/]0 186
62. A method for promoting growth of mammalian neuronal stem cells, including treating the cell with an amount of a polypeptide effective to increase the rate of growth of the neuronal stem cells, said polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
63. A method for preventing, treating or reducing the severity of a neurodegenerative disorder, including administering to a subject a therapeutically effective amount of a polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ :i 15 ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment Sthereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) 20 functionally replaces drosophila hedgehog in transgenic drosophila. oo* 64. The method of claim 63, wherein said disorder is Parkinson's disease. The method of claim 63, wherein said disorder is selected from Alzheimer's Disease, Huntington's Disease, Pick's Disease, Ballism, Guillain-Barre Syndrome, Amylotrophic Lateral Sclerosis, spinocerebellar degenerations, and peripheral neuropathy.
66. The method of claim 63, wherein said neurodegenerative disorder includes loss of neuronal cells selected from cholinergic neurons, GABAnergic neurons, and striatal neurons. P:\OPER\Fas\2473890-clims.doc-12/11/01 187
67. A method for preventing, treating or reducing the severity of an acute, subacute or chronic injury to the nervous system in a subject, including administering to a subject a amount of a polypeptide therapeutically effective to alter, relative to the absence of treatment, at least one of rate of growth, (ii) differentiation, or (iii) survival of one or more neuronal cell-types in said subject, said polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
68. The method of claim 67, wherein said injury is selected from traumatic injury, chemical injury, vasal injury, vasal deficit, infectious injury, inflammatory injury and 15 tumor-induced injury.
69. The method of claim 67, wherein said injury is a result of a chronic inflammatory disease. The method of claim 69, wherein said inflammatory disease is multiple sclerosis. 20 71. The method of claim 67, wherein said injury includes ischemia of neuronal tissue.
72. The method of claim 71, wherein said injury includes ischemia resulting from a stroke.
73. The method of claim 71, wherein said neuronal cells are introduced into a subject by cerebral grafting. P:\OPER\F;s\2473890-clls.doc4)2/1 1/01 188
74. The method of claim 73, wherein said neuronal cells are derived from fetal or neonatal animals. The method of claim 72, wherein said neuronal cell is a neuronal stem cell.
76. The method of claim 75, wherein said neuronal stem cell is a neural crest cell.
77. The method of claim 60, 61, 62, 63, or 67, wherein said polypeptide is administered in combination with one or more other neurotrophic factors.
78. The method of claim 77, wherein said other neurotrophic factor is selected from CNTF, BNTF and NGF.
79. The method of claim 63, wherein the neurodegenerative disorder includes degeneration of the peripheral nervous system.
80. The method of claim 79, wherein said disorder affects smooth muscle tissue and endocrine tissue, such as glandular tissue.
81. The method of claim 80, wherein said disorder is tachycardia or atrial cardiac arrhythmia.
82. The method of claim 79, wherein said disorder affects sensory or motor neurons.
83. The method of claim 82, wherein said disorder is selected from trauma, infarction, infection, metabolic disease, nutritional deficiency, toxic agents and chromic pain syndrome.
84. The method of claim 60, wherein said neuronal cell is a neural progenitor cell.
85. The method of claim 60, wherein said neuronal cell differentiates into a cell having a particular neural phenotype, such as a neuron or a glia. P:\OPER\FUs\2473891)-cilusdoc)2/1 1/01 189
86. The method of claim 60, wherein said neuronal cell is in the central nervous system or the peripheral nervous system.
87. The method of claim 86, wherein said treatment repairs central or peripheral nerve damage.
88. The method of claim 60, wherein said polypeptide includes a hedgehog amino acid sequence encoded by a naturally occurring vertebrate hedgehog gene.
89. The method of claim 88, wherein said hedgehog gene is a mammalian hedgehog gene. The method of claim 89, wherein said hedgehog gene is a human hedgehog gene.
91. The method of claim 60, wherein said polypeptide includes a hedgehog amino 4 acid sequence which is encoded by at least a portion of a hedgehog gene of vertebrate origin corresponding to residues 64-567 of SEQ ID NO: 1, residues 64-561 of SEQ ID NO: 2, residues 1-348 of SEQ ID NO: 3, residues 73-561 of SEQ ID NO: 4, and residues 70-558 of SEQ ID NO:
92. The method of claim 60, wherein said polypeptide has an approximate molecular weight of 19 kD.
93. The method of claim 60, wherein said polypeptide includes at least 150 amino acid residues of the N-terminal half of a hedgehog protein. 9
94. The method of claim 60, wherein said polypeptide binds to a patched protein. The method of claim 94, wherein said patched protein is a patched protein of a vertebrate organism. P:\OPER\Fas\247389()-climsdoc-)2/I I/01 190
96. The method of claim 60, wherein said neuronal cells are selected from motor neurons, cholinergic neurons, dopaminergic neurons, serotonergic neurons and peptidergic neurons.
97. A method for modulating the growth state of a chondrocytic cell including contacting the cell with a polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
98. The method of claim 97, wherein the polypeptide increases the rate of proliferation of the cell. 15 99. The method of claim 97, wherein the polypeptide decreases the rate of "proliferation of the cell. S100. The method of claim 97, wherein the cell is in culture, and the polypeptide is provided as a cell culture additive.
101. The method of claim 97, wherein the cell is treated in an animal and the polypeptide is administered to the animal as a therapeutic composition.
102. The method of claim 97, wherein said polypeptide has an approximate molecular weight of 19 kD.
103. The method of claim 97, wherein said polypeptide includes at least 150 amino acid residues of the N-terminal half of a hedgehog protein. P:\OPER\Fas\2473890-chns.do-4)2/I 1/I 191
104. The method of claim 97, wherein said polypeptide binds to a patched protein.
105. The method of claim 104, wherein said patched protein is a patched protein of a vertebrate organism.
106. The method of claim 97, wherein said polypeptide includes at least 50 amino acid residues of the N-terminal half of a hedgehog protein.
107. The method of claim 97, wherein said polypeptide includes at least 100 amino acid residues of the N-terminal half of a hedgehog protein.
108. The method of claim 101, wherein animal is being treated prophylactically.
109. A method for treating a skeletogenic disorder, including administering to a patient an amount of a hedgehog polypeptide effective to alleviate conditions of the 0 disorder, said polypeptide including a hedgehog amino acid sequence at least identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to a patched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ 0" lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila.
110. The method of claim 109, wherein the hedgehog polypeptide is coadministered 20 with PTHrP or an analog thereof.
111. The method of claim 109, wherein said polypeptide has an approximate molecular weight of 19 kD.
112. The method of claim 109, wherein said polypeptide includes at least 150 amino acid residues of the N-terminal half of a hedgehog protein.
193- 117. Use of a hedgehog polypeptide said polypeptide including a hedgehog amino acid sequence at least 80% identical to at least one of SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, or a fragment thereof, which hedgehog amino acid sequence binds to apatched receptor, (ii) regulates differentiation of neuronal cells, (iii) regulates survival of differentiated neuronal cells, (iv) regulates proliferation of chondrocytes, regulates proliferation of testicular germ lines cells, (vi) induces expression of a Hoxd gene, and/or (vii) functionally replaces drosophila hedgehog in transgenic drosophila for the manufacture of a medicament for treating a skeletogenic disorder. 118. An isolated nucleic acid encoding a polypeptide, substantially as herein described with reference to one or more of the examples but excluding comparative examples. 119. An isolated nucleic acid which hybridises under stringent conditions to a nucleic acid sequence, substantially as herein described with reference to one or more of the examples but excluding comparative examples. 120. A method for inducing a cell to differentiate to a neuronal cell phenotype, substantially as herein described with reference to one or more of the examples but "excluding comparative examples. V 121. A method of modulating skeletogenesis, substantially as herein described with reference to one or more of the examples but excluding comparative examples. 0 .122. A method for treating a degenerative disorder of the nervous system characterized by neuronal cell death, substantially as herein described with reference to one or more of the examples but excluding comparative examples. 123. A method for modulating one or more of growth, differentiation and survival of a S. 55 25 neuronal cell, substantially as herein described with reference to one or more of the •examples but excluding comparative examples. S 124. A method for promoting survival of mammalian neuronal cells responsive to hedgehog induction, substantially as herein described with reference to one or more of the examples but excluding comparative examples. the examples but excluding comparative examples. 500230206_.DOC/BSW -194- 125. A method for promoting growth of mammalian neuronal stem cells, substantially as herein described with reference to one or more of the examples but excluding comparative examples. 126. A method for preventing, treating or reducing the severity of a neurodegenerative disorder, substantially as herein described with reference to one or more of the examples but excluding comparative examples. 127. A method for preventing, treating or reducing the severity of an acute, subacute or chronic injury to the nervous system in a subject, substantially as herein described with reference to one or more of the examples but excluding comparative examples. 128. A method for modulating the growth state of a chondrocytic cell, substantially as herein described with reference to one or more of the examples but excluding comparative examples. 129. Use of a pharmaceutical preparation of a polypeptide for the manufacture of a medicament, substantially as herein described with reference to one or more of the examples but excluding comparative examples. 130. Use of a polypeptide for the manufacture of a medicament, substantially as herein described with reference to one or more of the examples but excluding comparative examples. 131. Use of a hedgehog polypeptide for the manufacture of a medicament, 20 substantially as herein described with reference to one or more of the examples but excluding comparative examples. DATED this 2 4 th day of September 2003 BALDWIN SHELSTON WATERS Attorneys for: President and Fellows of Harvard College and Imperial Cancer Research Technology Ltd *0 .0 0 @00 S 0 500230206 .Doc/BSW
AU87319/01A 1993-12-30 2001-11-02 Vertebrate embryonic pattern-inducing hedgehog-like proteins Expired AU767640B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU87319/01A AU767640B2 (en) 1993-12-30 2001-11-02 Vertebrate embryonic pattern-inducing hedgehog-like proteins

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US08/176427 1993-12-30
US08/356,060 US5844079A (en) 1993-12-30 1994-12-14 Vertebrate embryonic pattern-inducing proteins, and uses related thereto
US08/356060 1994-12-14
AU16451/99A AU736492C (en) 1993-12-30 1999-02-15 Vertebrate embryonic pattern-inducing hedgehog-like proteins
AU87319/01A AU767640B2 (en) 1993-12-30 2001-11-02 Vertebrate embryonic pattern-inducing hedgehog-like proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU16451/99A Division AU736492C (en) 1993-12-30 1999-02-15 Vertebrate embryonic pattern-inducing hedgehog-like proteins

Publications (2)

Publication Number Publication Date
AU8731901A AU8731901A (en) 2002-05-02
AU767640B2 true AU767640B2 (en) 2003-11-20

Family

ID=29780883

Family Applications (1)

Application Number Title Priority Date Filing Date
AU87319/01A Expired AU767640B2 (en) 1993-12-30 2001-11-02 Vertebrate embryonic pattern-inducing hedgehog-like proteins

Country Status (1)

Country Link
AU (1) AU767640B2 (en)

Also Published As

Publication number Publication date
AU8731901A (en) 2002-05-02

Similar Documents

Publication Publication Date Title
AU704178B2 (en) Vertebrate embryonic pattern-inducing hedgehog-like proteins
US5844079A (en) Vertebrate embryonic pattern-inducing proteins, and uses related thereto
US6576237B1 (en) Vertebrate tissue pattern-inducing proteins, and uses related thereto
US7781215B2 (en) Vertebrate embryonic pattern-inducing proteins, and uses related thereto
US6271363B1 (en) Nucleic acids encoding hedgehog proteins
US7901686B2 (en) Hedgehog antibodies
US6261786B1 (en) Screening assays for hedgehog agonists and antagonists
US6610656B1 (en) Method of promoting chondrocyte differentiation with hedgehog related polypeptides
US5795734A (en) EPH receptor ligands, and uses related thereto
EP1471926B1 (en) Compositions and methods for the therapeutic use of an atonal-associated sequence
JP4083810B2 (en) Hedgehog-interacting protein and related uses
AU726918B2 (en) TGFbeta signal transduction proteins, genes, and uses related thereto
WO1997022697A9 (en) TGFβ SIGNAL TRANSDUCTION PROTEINS, GENES, AND USES RELATED THERETO
AU736492B2 (en) Vertebrate embryonic pattern-inducing hedgehog-like proteins
AU767640B2 (en) Vertebrate embryonic pattern-inducing hedgehog-like proteins
US7196170B2 (en) Aiolos, Helios, Daedalos and Ikaros: genes, polypeptides, regulatory elements and uses thereof
US20080152642A1 (en) Aiolos, Helios, Daedalos and Ikaros: genes, polypeptides, regulatory elements and uses thereof
KAUFMAN RELATED APPLICATIONS
NZ500010A (en) Hedgehog proteins that bind to a patched receptor
US6777536B1 (en) EPH receptor ligands, and uses related thereto
De Robertis Inactivation of mouse Twisted gastrulation reveals its role in promoting Bmp4 activity during forebrain development
JP2001309735A (en) ANIMAL DEFICIENT IN Smg GDS ENCODED GENE

Legal Events

Date Code Title Description
NB Applications allowed - extensions of time section 223(2)

Free format text: THE TIME IN WHICH TO MAKE A FURTHER APPLICATION FOR A DIVISIONAL PATENT HAS BEEN EXTENDED TO 20011102

FGA Letters patent sealed or granted (standard patent)