AU2004200576A1 - Human homolog of the drosophila protein "fused" - Google Patents

Human homolog of the drosophila protein "fused" Download PDF

Info

Publication number
AU2004200576A1
AU2004200576A1 AU2004200576A AU2004200576A AU2004200576A1 AU 2004200576 A1 AU2004200576 A1 AU 2004200576A1 AU 2004200576 A AU2004200576 A AU 2004200576A AU 2004200576 A AU2004200576 A AU 2004200576A AU 2004200576 A1 AU2004200576 A1 AU 2004200576A1
Authority
AU
Australia
Prior art keywords
fused
nucleic acid
antibody
disease
seq
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.)
Granted
Application number
AU2004200576A
Other versions
AU2004200576B2 (en
Inventor
Frederic J. De Sauvage
Arnon Rosenthal
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.)
Genentech Inc
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU28779/99A external-priority patent/AU767491B2/en
Application filed by Genentech Inc filed Critical Genentech Inc
Publication of AU2004200576A1 publication Critical patent/AU2004200576A1/en
Application granted granted Critical
Publication of AU2004200576B2 publication Critical patent/AU2004200576B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1205Phosphotransferases with an alcohol group as acceptor (2.7.1), e.g. protein kinases
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Description

AUSTRALIA
Patents Act 1990 COMPLETE SPECIFICATION STANDARD PATENT Applicant(s): GENENTECH, INC.
Invention Title: HUMAN HOMOLOG OF THE DROSOPHILA PROTEIN "FUSED" The following statement is a full description of this invention, including the best method of performing it known to me/us: 1
FUSED
The entire disclosure in the complete specification of our Australian Patent Application No.
28779/99 is by this cross-reference incorporated into the present specification.
FIELD OF THE INVENTION The present invention relates generally to signaling molecules, specifically to signaling and mediator molecules in the hedgehog (Hh) cascade which are involved in cell proliferation and differentiation.
All references, including any patents or patent applications, cited in this specification are hereby incorporated by reference. No admission is made that any reference constitutes prior art. The discussion of the references states what their authors assert, and the applicants reserve the right to challenge the accuracy and pertinency of the cited documents. It will be clearly understood that, although a number of prior art publications are referred to herein, this reference does not constitute an admission that any of these documents forms part of the common general knowledge in the art, in Australia or in any other country.
BACKGROUND OF THE INVENTION Development of multicellular organisms depends, at least in part, on mechanisms which specify, direct or maintain positional information to pattern cells, tissues, or organs. Various secreted signaling molecules, such as members of the transforming growth factor-beta (TGF-P), Wnt, fibroblast growth factors and hedgehog families have been associated with patterning activity of different cells and structures in Drosophila as well as in vertebrates. Perrimon, Cell: 80: 517-520 (1995).
Hedgehog (Hh) was first identified as a segmentpolarity gene by a genetic screen in Drosophila lamelanogaster, Nusslein-Volhard et al., Roux. Arch. Dev.
Biol. 193: 267-282 (1984), that plays a wide variety of developmental functions. Perrimon, supra. Although only one Drosophila Hh gene has been identified, three mammalian Hh homologues have been isolated: Sonic Hh (SHh), Desert Hh (DHh) and Indian Hh (IHh), Echelard et al., Cell 75: 1417-30 (1993); Riddle et al., Cell 1401-16 (1993). SHh is expressed at high level in the notochord and floor plate of developing vertebrate embryos. In vitro explant assays as well as ectopic expression of SHh in transgenic animals show that SHh plays a key role in neuronal tube patterning, Echelard et al., supra., Krauss et al., Cell 75, 1431-44 (1993), Riddle et al., Cell 75: 1401-16 (1993), Roelink et al, Cell 81: 445-55 (1995). In vitro explant assays as well as ectopic expression of SHh in transgenic animals show that SHh plays a key role in neural tube patterning, Echelard et al. (1993), supra.; Ericson et al., Cell 81: 747-56 (1995); Marti et al., Nature 375: 322-5 (1995); Roelink et al. (1995), supra; Hynes et al., Neuron 19: 26 (1997). Hh also plays a role in the development of .limbs (Krauss et al., Cell 75: 1431-44 (1993); Laufer et al., Cell 79, 993-1003 (1994)), somites (Fan and Tessier- Lavigne, Cell 79, 1175-86 (1994); Johnson et al., Cell 79: 1165-73 (1994)), lungs (Bellusci et al., Develop. 124: 53- 63 (1997) and skin (Oro et al., Science 276: 817-21 (1997). Likewise, IHh and DHh are involved in bone, gut and germinal cell development, Apelqvist et al., Curr.
Biol. 7: 801-4 (1997); Bellusci et al., Development. 124: 53-63 (1997); Bitgood et al., Curr. Biol. 6: 298-304 (1996); Roberts et al., Development 121: 3163-74 (1995).
SHh knockout mice further strengthened the notion that SHh is critical to many aspect of vertebrate development, Chiang et al., Nature 383: 407-13 (1996). These mice show defects in midline structures such as the notochord and the floor plate, absence of ventral cell types in neural tube, absence of distal limb structures, cyclopia, and ibabsence of the spinal column and most of the ribs.
At the cell surface, the Hh signals is thought to be relayed by the 12 transmembrane domain protein Patched (Ptch) [Hooper and Scott, Cell 59: 751-65 (1989); Nakano et al., Nature 341: 508-13 (1989)] and the G-protein coupled like receptor Smoothened (Smo) [Alcedo et al., Cell 86: 221-232 (1996); van den Heuvel and Ingham, Nature 382: 547-551 (1996)]. Both genetic and biochemical evidence support a receptor model where Ptch and Smo are part of a multicomponent receptor complex, Chen and Struhl, Cell 553-63 (1996); Marigo et al., Nature 384: 176-9 (1996); Stone et al., Nature 384: 129-34 (1996). Upon binding of Hh to Ptch, the normal inhibitory effect of Ptch on Smo is relieved, allowing Smo to transduce the Hh signal across the plasma membrane. Loss of function mutations in the Ptch gene have been identified in patients with the basal cell nevus syndrome (BCNS), a hereditary disease characterized by multiple basal cell carcinomas (BCCs). Disfunctional Ptch gene mutations have also been associated with a large percentage of sporadic basal cell carcinoma tumors, Chidambaram et al., Cancer Research 56: 4599-601 (1996); Gailani et al., Nature Genet. 14: 78-81 (1996); Hahn et al., Cell B5: 841-51 (1996); Johnson et al., Science 272: 1668-71 (1996); Unden et al., Cancer Res. 56: 4562-5 (1996); Wicking et al., Am. J. Hum. Genet. fiQ: 21-6 (1997). Loss of Ptch function is thought to cause an uncontrolled Smo signaling in basal cell carcinoma. Similarly, activating Smo mutations have been identified in sporatic BCC tumors (Xie et al., Nature 391: 90-2 (1998)), emphasizing the role of Smo as the signaling subunit in the receptor complex for SHh. However, the exact mechanism by which Ptch controls Smo activity still has yet to be clarified and the signaling mechanisms by which the Hh signal is transmitted from the receptor to downstream targets also remain to be elucidated. Genetic epistatic analysis in Drosophila has identified several segment-polarity genes which appear to function as components of the Hh signal transduction pathway, Ingham, Curr. Opin. Genet. Dev. 5: 492-8 (1995); Perrimon, supra. These include a kinesin-like molecule, Costal-2 (Cos-2) [Robbins et al., Cell 90: 225-34 (1997); Sisson et al., Cell 90: 235-45 (1997)], a protein designated fused [Preat et al., Genetics 135: 1047-62 (1993); Therond et al., Proc. Natl Acad Sci. USA 93: 4224-8 (1996)], a novel molecule with unknown function designated Suppressor offused [Pham et al., Genetics 140: 587-98 (1995); Preat, Genetics 132: 725-36 (1992)] and a zinc finger protein Ci. [Alexandre et al., Genes Dev. 10: 2003-13 (1996); Dominguez et al., Science 272: 1621-5 (1996); Orenic et al, Genes Dev. 4: 1053-67 (1990)]. Additional elements implicated in Hh signaling include the transcription factor CBP [Akimaru et al., Nature 386: 735-738 (1997)], the negative regulator slimb [Jiang and Struhl, Nature 391: 493-496 (1998)] and the SHh response element COUP-TFII [Krishnan et al., Science 228: 1947-1950 (1997)].
Mutants in Cos-2 are embryonicly lethal and display a phenotype similar to Hh over expression, including duplications of the central component of each segment and expansion domain of Hh responsive genes.
In contrast, mutant embryos for fused and Ci show a phenotype similar to Hh loss of function including deletion of the posterior part of each segment and replacement of a mirror-like image duplication of the anterior part or each segment and replacement of a mirror-like duplication of the anterior part, Busson et al., Roux. Arch. Dev.
Biol. 197: 221-230 (1988). Molecular characterizations of Ci suggested that it is a transcription factor which directly activates Hh responsive genes such as Wingless and Dpp, Alexandre et al., (1996) supra, Dominguez et al., (1996) supra. Likewise, molecular analysis of fused reveals that it is structurally related to serine threonine kinases and that both intact N-terminal kinase domain and a C-terminal regulatory region are required for its proper function, Preat et al., Nature 342: 87-9 (1990); Robbins et al., (1997), supra; Therond et al., Proc. Natl.
Acad. Sci. USA 93: 4224-8 (1996). Consistent with the putative opposing functions of Cos-2 and fused, fused mutations are suppressed by Cos-2 mutants and also by Suppressor offused mutants, Preat et al., Genetics 135: 2 \\melb...files\home\uzannet\Keep\speci\28779-99.1 SPEjCI.doc 10/05/01 3 1047-62 (1993). However, whereas fused null mutations and N-terminal kinase domain mutations can be fully suppressed by Suppressor of fused mutations, C-terminus mutations of fused display a strong Cos-2 phenotype in a Suppressor of fused background. This suggests that the fused kinase domain can act as a constitutive activator of SHh signaling when Suppressor of Fused is not present. Recent studies have shown that the 92 kDa Drosophila fused, Cos-2 and Ci are present in a microtubule associated multiprotein complex and that Hh signaling leads to dissociation of this complex from microtubules, Robbins et al., Cell 90: 225-34 (1997); Sisson et al., Cell 90: 235- (1997). Both fused and Cos-2 become phosphorylated in response to Hh treatment, Robbins et al., supra; Therond et al., Genetics 142: 1181-98 (1996), but the kinase(s) responsible for this activity(ies) remain to be characterized. To date, the only known vertebrate homologues for these components are members of the Gli protein family Gli-1, Gli-2 and Gli-3). These are zinc finger putative transcription factors that are structurally related to Ci. Among these, Gli-1 was shown to be a candidate mediator of the SHh signal [Hynes et al., Neuron 15: 35-44 (1995); Lee et al., Development 124: 2537-52 (1997); Alexandre et al., Genes Dev. 10: 2003-13 (1996)] suggesting that the mechanism of gene activation in response to Hh may be conserved between fly and vertebrates. To determine whether other signaling components in the Hh cascade are evolutionarily conserved and to examine the function of fused in the Hh signaling cascade on the biochemical level, Applicants have isolated and characterized the human fused cDNA. Tissue distribution on the mouse indicates that fused is expressed in SHh responsive tissues. Biochemical studies demonstrate that fused is a functional kinase. Functional studies provide evidence that fused is an activator of Gli and that a dominant negative form of fused is capable of blocking SHh signaling in Xenopus embryos. Together this 3a data demonstrated that fused is directly involved in Hh signaling.
Applicants have identified a cDNA encoding a human fused (hfused) polypeptide and thus have provided for the first time a vertebrate fused molecule.
For the purposes of this specification it will be clearly understood that the word "comprising" means "including but not limited to", and that the word "comprises" has a corresponding meaning.
SUMMARY OF THE INVENTION The present invention provides an isolated nucleic acid molecule, which is antisense to a nucleic acid molecule having at least an 80% nucleic acid sequence identity to a DNA molecule which encodes a vertebrate fused polypeptide comprising the sequence of amino acids 1 to about 260 of Figure 1 (SEQ ID NO:24) and which has fused biological activity.
The present invention further provides an isolated nucleic acid, which is antisense to a nucleic acid molecule having at least an 80% nucleic acid sequence identity to a DNA molecule encoding the same mature polypeptide as that encoded by the cDNA in ATCC Deposit No. 209637.
The present invention further provides an antibody which specifically binds to an isolated vertebrate fused polypeptide, the polypeptide having at least 80% amino acid sequence identity to amino acid residues 1 to 1315 of Figure 1 (SEQ ID NO:2), and having fused biological activity.
The present invention further provides an antibody which specifically binds to an isolated, native sequence vertebrate fused polypeptide, the polypeptide having at least 80% amino acid sequence identity to the human fused polypeptide encoded by the cDNA deposited under accession number ATCC 209637, and having fused biological activity.
3b In one embodiment, the invention provides an isolated nucleic acid molecule having at least about sequence identity to a DNA molecule encoding a fused polypeptide comprising the sequence of amino acids 1 to 260 of Fig. 1 (SEQ ID NO. or the complement of the DNA molecule of and encoding a polypeptide having fused biological activity. The sequence identity preferably is about 85%, more preferably about 90%, most preferably about 95%. In one aspect, the isolated nucleic acid has at least about 80%, preferably at least about more preferably at least about 90%, and most preferably at least about 95% sequence identity with a polypeptide having amino acid residues 1 to about 1315 of Fig. 1 (SEQ ID NO. Preferably, the highest degree of sequence identity occurs within the kinase domain (amino acids 1 to about 260 (SEQ ID NO:24 as shown in Fig. 1).
Especially preferred are those nucleic acid molecules containing a coding sequence for a lysine at amino acid position 33. In a further aspect, the isolated nucleic acid molecule comprises DNA encoding a human fused polypeptide having amino acid residues 1 to about 260 (SEQ ID NO:24 as shown in Fig. In yet another aspect, the invention provides for an isolated nucleic acid comprising DNA having at least a 95% sequence identity to a DNA molecule encoding the same mature polypeptide encoded by the cDNA in ATCC Deposit No. 209637 (designation: pRK5tkneo.hFused-1272), alternatively the coding sequence of clone pRK5tkneo.hFused-1272, deposited under accession number ATCC 209637. In a still further aspect, the invention provides for a nucleic acid comprising human fused encoding sequence of the cDNA in ATCC deposit No. 209637 (designation: pRK5tkneo.hFused-1272) or a sequence which hybridizes thereto under stringent conditions.
In another embodiment, the invention provides a vector comprising DNA encoding a vertebrate fused polypeptide. A host cell comprising such a vector is also provided. By way of example, the host cells may be mammalian cells, CHO cells), prokaryotic cells E. colt) or yeast cells Saccharomyces cerevisiae). A process for producing vertebrate fused polypeptides is further provided and comprises culturing host cells under conditions suitable for expression of vertebrate fused and recovering the same from the cell culture.
In yet another embodiment, the invention provides an isolated vertebrate fused polypeptide. In particular, the invention provides isolated native sequence vertebrate fused polypeptide, which in one embodiment is a human fused including an amino acid sequence comprising residues 1 to about 1315 of Figure 1 (SEQ ID NO. Human and other native vertebrate fused polypeptides with or without the initiating methionine are specifically included. Alternatively, the invention provides a vertebrate fused polypeptide encoded by the nucleic acid deposited under accession number ATCC 209637.
In yet another embodiment, the invention provides chimeric molecules comprising a vertebrate fused polypeptide fused to a heterologous polypeptide or amino acid sequence. An example of such a chimeric molecule comprises a vertebrate fused polypeptide fused to an epitope tag sequence or a constant region of an immunoglobulin.
In yet another embodiment, the invention provides an expressed sequence tag (EST) comprising the nucleotide sequences identified in Fig. 2 as 2515662 (SEQ ID NO. 3).
In yet another embodiment, the invention provides for compounds and methods for developing antagonists against and agonist promoting fused modulation of Hedgehog signaling. In particular, an antagonist of vertebrate fused which blocks, prevents, inhibits and/or neutralized the normal functioning of fused in the SHh signaling pathway, including both small bioorganic molecules and antisense nucleotides.
In yet another embodiment, the invention provides for an agonist of vertebrate fused which stimulates or enhances the normal functioning of vertebrate fused in the Hh signalling pathway.
In yet another embodiment, the invention provides for alternatively spliced variants of human fused.
In still yet a further embodiment, the invention provides a method of screening or assaying for identifying molecules that modulate the fused activation of hedgehog signaling. Preferably, the molecules either prevent interaction of fused with its associative complexing proteins or prevent or inhibit dissociation of complexes. The assay comprises the incubation of a mixture comprising fused and a substrate Gli, COUP-TFII, slimb, CBP, MBP) with a candidate molecule, and detection of the ability of the candidate molecule to modulate fused phosphorylation of its substrate. The screened molecules preferably are small molecule drug candidates. In particular, the method relates to a technique for screening for antagonists or agonists of fused biological activity, comprising: exposing the fused expressing target cells in culture to a candidate compound; and analyzing cell lysates to asses the level and/or identity of phosphorylation; or scoring phenotypic or functional changes in treated cells; and comparing the results to control cells which were not exposed to the candidate compound.
4 H:\suzannet\Keep\speci\28779-99.1 SPECIdoc 10/05/01 In yet another embodiment, the method relates to a technique of diagnosing to determine whether a particular disorder is modulated by hedgehog signaling, comprising: culturing test cells or tissues; administering a compound which can inhibit fused modulated hedgehog signaling; and measuring the degree of kinase attenuation on the fused substrate in cell lysates or 4a H:\suzannet\Keep\Speci\28779-99.1 SPECI.doc 10/05/01 5 hedgehog-mediated phenotypic effects in the test cells.
BRIEF DESCRIPTION OF THE DRAWINGS Figures 1A-1F show the nucleotide (SEQ ID NO. 1) and derived amino acid sequence (SEQ ID NO. 2) of a native sequence of human fused polypeptide. Included are the kinase domain (residues 1 to about 260)(SEQ ID NO. 24) and the ATP binding site at about amino acid position 33.
Figure 2 shows the EST 2515662 (SEQ ID NO. 3) that was used in the cloning of the human full-length sequence.
Figures 3A-3E show a comparison between human and Drosophila fused (SEQ ID NOs. 2 and 23, respectively).
Gaps introduced for optimal alignment are indicated by dashes. Identical amino acids are boxed. The lysine residue mutated in fused-DN (dominant negative, lysine at amino acid position 33) is shown with a star.
Figures 4A-4F show the sequence of DNA28494 (SEQ ID NO. 6) that was an incorrectly spliced variant of human fused isolated from a fetal lung library. This clone contains a potential initiation methionine at position 116 followed by an open reading frame of 1944 bp. A second open reading frame is present from about position 2295 to 4349. There is one nucleotide difference between clone DNA28495 (SEQ ID NO. 4) and clone DNA28494 (SEQ ID NO. 6) located in the first ORF at position 1863 of clone 28495 (SEQ ID NO.4) (A vs. G) which changes the coding sequence from a Gln to an Arg at position 583. The first open reading frame of DNA28494 (SEQ ID NO. 6) starts at residue 115 and is followed by a 630 amino acid long open reading frame.
Figures 5A-5F show the sequence of DNA28495 (SEQ ID NO. 4) that was another incorrectly spliced variant of human fused isolated from a fetal lung library.
Figure 6 is a western blot of the PCR product of an epitope tag of DNA28495 (SEQ ID NOs. 5 and 21) and DNA28494 (SEQ ID NOs. 7 and 22). A specific band of H.\RBell\Keep\28779-99.doc 15/09/03 5a 150 kDa was detected in the cell pellet of cells transfected with the construct corresponding to clone DNA28494 (SEQ ID NO. 6) and a specific band of approximately 100 kDa could be detected for clone DNA28495 (SEQ ID NO. 4) (Fig. These bands were not present in the mock transfected control. The presence of the 100 kDa band suggests the two open reading frames of DNA28494 (SEQ ID NO. 6) can be spliced together to direct the synthesis of a large protein of 150 kDa. The absence of this band for DNA28495 (SEQ ID NO. 4) suggested that this clone apparently cannot be correctly spiced.
Figure 7 is a northern blot analysis of human fused (SEQ ID NO. Multiple human fetal and adult tissue northern blots were probed with a human fused cDNA probe.
Figures 8A-8F is a photograph showing in situhybridisation of embryonic and adult tissues with fused (SEQ ID NO. Sagittal sections of Ell.5 (Fig. 8A) and E13.5 (Fig. 8B) mouse embryos. Coronal section through the spinal cord of Ell.5 (Fig. 8C) and E.13.5 (Fig. 8D) mouse embryo. Sagittal section through P1 (Fig. 8E) and adult (Fig. 8F) mouse. Cp, choroid plexus; hb, hindbrain; hip, hippocampal formation; ht, heart; hy, hypothalamus; kd, kidney; Ig, lung; mb, midbrain; md, midgut; mnd, mandibular component of first branchial arch; sc, spinal cord; st, stomach; tec, midbrain tectum; vh, ventral horn of spinal cord; vm, ventral midbrain. Scale bars: Fig. 8A, mm; Fig. 8B, 1.62 mm; Fig. 8C, 0.14 mm; Fig. 8D, 0.17 mm; Fig. 8E, 2.00 mm; and Fig. 8F, 3.1 mm.
H.\RBe11\Keep\28779-99.doc 15/09/03 P1272 Figures 9A-9C are a photograph showing in situ hybridization showing the presence of fused mRNA in high levels in the adult mouse testes (Fig. 9A). High magnification reveals differences in levels of expression within seminiferous tubules (Fig. 9C). Hybridization of the testis with a sense strand control probe to fused gave no hybridization (Fig. 9B).
Figures 10A-10B are a bar graph representing the activation of Gli by fused. (Fig. C3H10TI/2 cells were cotransfected with a p9XGliLus, ptkRenilla luciferase and fused or various fused mutants. Cells were harvested 48h after transfection and the luciferase activity was assayed as described in Example 7. (Fig. 10B): Fused transactivation of a Gli reporter construct. C3HIOTI/2 cells were cotransfected with a p9XGliLuc reporter construct, ptkRenilla luciferase and a CMV driven expression vector for fused or various fused mutants. Cells were harvested 48 hours after transfection and the luciferase activity was assayed as described in the Examples. The data represents the mean of duplicative determinations.
Figures 1 IA-I IE are a photograph showing thatfused-DN (SEQ ID NO 25) inhibits SHh signaling in early Xenopus development. Depicted are: (Fig. 11I A) Dorsal view of tadpole stage embryos. Top embryo is fused-DN (SEQ ID NO 25) injection and bottom embryo is the control; (Fig. 11 B) Side view of tadpole stage embryo. Top embryo isfused-DN (SEQ ID NO 25) injection and bottom embryo is the control; (Figs.
I IC I ID) Pax-6 staining of stage 16 neurula embryos injected with control DNA andfused-DN (SEQ ID NO 25), respectively; (Fig. 1 IE) SHh expression in the floor plate of neurula stage control embryo (left) or fused-DN (SEQ ID NO 25) injected embryo (right).
Figure 12 is a photograph which confirms the kinase activity of fused (SEQ ID NO 2) and its activation of Gli. Depicted are 293 cells transfected with HA tagged fused constructs as indicated in Example 10 and immunoprecipitated with anti-HA antibodies and protein A sepharose. Protein A beads were subjected to in vitro kinase assay as described in Example 10 in the presence of MBP.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS I. Definitions The terms "vertebrate fused" and "vertebrate fused polypeptide" when used herein encompass native sequence vertebrate fused and vertebrate fused variants (which are further defined herein) having fused biological activity. Fused may be isolated from a variety of sources, such as from human tissue types or from another source, or prepared by recombinant or synthetic methods.
A "native sequence vertebrate fused" comprises a polypeptide having the same amino acid sequence as a vertebrate fused derived from nature. Such native sequence vertebrate fused can be isolated from nature or can be produced by recombinant and/or synthetic means. The term "native sequence vertebrate fused' specifically encompasses naturally occurring truncated forms of vertebrate fused, naturally occurring variant forms alternatively spliced forms) and naturally-occurring allelic variants of vertebrate fused. Native vertebrate fused includes fused in mammals such as human, murine, bovine, porcine, equine, feline, canine, etc., and preferably refers to human. Thus, one embodiment of the invention, the native sequence human vertebrate fused is a mature or full-length native human vertebrate fused comprising amino acids I to 1315 of SEQ ID NO: 2 as shown in Fig. I with or without the initiating methionine at position 1.
"Vertebrate fused variant" means an active vertebrate fused as defined below having at least about PI272 amino acid sequence identity to a DNA molecule encoding a vertebrate fused polypeptide, or the complement of the DNA molecule of In a particular embodiment, the vertebrate fused variant has at least about 80% amino acid sequence homology with the vertebrate fused having the deduced amino acid sequence (SEQ ID NO:2) shown in Fig. I for a full-length native sequence vertebrate fused. Such vertebrate fused variants include, without limitation, vertebrate fused polypeptides wherein one or more amino acid residues are added, or deleted, at the N- or C-terminus of the sequence of Fig. 1 (SEQ ID NO Preferably, the nucleic acid or amino acid sequence identity is at least about 85%, more preferably at least about and even more preferably at least about "Percent amino acid sequence identity" with respect to the vertebrate fused sequences identified herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the vertebrate fused sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
"Percent nucleic acid sequence identity" with respect to the vertebrate fused sequences identified herein is defined as the percentage of nucleotides in a candidate sequence that are identical with the nucleotides in the vertebrate fused sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent nucleic acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
The term "epitope tagged" when used herein refers to a chimeric polypeptide comprising vertebrate fused polypeptide, or a portion thereof, fused to a "tag polypeptide". The tag polypeptide has enough residues to provide an epitope against which an antibody can be made, yet is short enough such that it does not interfere with activity of the vertebrate fused polypeptide. The tag polypeptide preferably also is fairly unique so that the antibody does not substantially cross-react with other epitopes. Suitable tag polypeptides generally have at least six amino acid residues and usually between about 8 to about 50 amino acid residues (preferably, between about 10 to about 20 residues).
As used herein, the term "immunoadhesin" designates antibody-like molecules which combine the binding specificity of a heterologous protein (an "adhesin") with the effector functions of immunoglobulin constant domains. Structurally, the immunoadhesin comprise a fusion of an amino acid sequence with the desired binding specificity which is other than the antigen recognition and binding site of an antibody is "heterologous"), and an immunoglobulin constant domain sequence. The adhesin part of an immunoadhesin molecule typically is a contiguous amino acid sequence comprising at least the binding site of a receptor or a ligand. The immunoglobulin constant domain sequence in the immunoadhesins may be obtained from any P1272 immunoglobulin, such as IgG-1, IgG-2, IgG-3 or IgG-4 subtypes, IgA (including IgA-1 and IgA-2, IgE, IgD or IgM. Immunoadhesion reported in the literature include fusions of the T cell receptor* [Gascoigne et al., Proc. Natl. Acad. Sci. USA 84: 2936-2940 (1987)]; CD4* [Capron et al., Nature 337: 525-531 (1989); Traunecker et al., Nature 339: 68-70 (1989); Zettmeissl et al., DNA Cell Biol. USA 9: 347-353 (1990); Byrn et al., Nature 344, 667-670 (1990)]; L-selectin (homing receptor) [Watson et al., J. Cell. Biol. 110, 2221-2229 (1990); Watson et al., Nature 349, 164-167 (1991)]; CD44* [Aruffo et al., Cell 61, 1303-1313 (1990)]; CD28' and B7* [Linsley et al., J. Exp. Med. 173, 721-730 (1991)]; CTLA-4' [Lisley et al., J. Exp. Med. 174, 561-569 (1991)]; CD22' [Stamenkovic et al., Cell 66. 1133-1144 (1991)]; TNF receptor [Ashkenazi et al., Proc. Natl.
Acad. Sci. USA 88, 10535-10539(1991); Lesslauer et al., Eur. J. Immunol. 27, 2883-2886 (1991); Peppel et al., J. Exp. Med. 174, 1483-1489 (1991)]; NP receptors [Bennett et al., J. Biol. Chem. 266, 23060-23067 (1991)]; IgE receptor a-chain' [Ridgway and Gorman, J. Cell. Biol. 115, abstr. 1448 (1991)]; HGF receptor [Mark, M.R.
et al., 1992, submitted], where the asterisk indicates that the receptor is member of the immunoglobulin superfamily.
"Stringency" of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization generally depends upon the ability of denatured DNA to reanneal when complementary strands are present in an environment near but below their Tm (melting temperature): The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature which can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. Moreover, stringency is also inversely proportional to salt concentrations. For additional details and explanation of stringency of hybridization reactions, see Ausubel et al., Current Protocols in Molecular Biology (1995).
"Stringent conditions," as defined herein may be identified by those that: employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/O.1% sodium dodecyl sulfate at 50 0 C; employ during hybridization a denaturing agent, such as formamide, for example, 50% (vol/vol) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42 0 C; employ 50% formamide, 5 x SSC (0.75 M NaCI, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 jig/ml), 0.1% SDS, and 10% dextran sulfate at 42°C, with washes at 42°C in 0.2 x SSC (sodium chloride/sodium citrate) and 50% formamide at 55 0 C, followed by a high-stringency wash consisting of 0.1 x SSC containing EDTA at 55 0
C.
"Moderately stringent conditions" may be identified as described by Sambrook et al, Molecular Cloning: A Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), and include the use of a washing solution and hybridization conditions temperature, ionic strength and %SDS) less stringent than described above. An example of moderately stringent conditions is a condition such as overnight incubation at 37°C in a solution comprising: 20% formamide, 5 x SSC (150 mM NaCI, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 5 x Denhardt's solution, 10% dextran sulfate, and mg/mL denatured sheared salmon sperm DNA, followed by washing the filters in 1 x SSC at about 37-50 0
C.
The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like.
"Isolated," when used to describe the various polypeptides disclosed herein, means polypeptide that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would typically interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes. In preferred embodiments, the polypeptide will be purified to a degree sufficient to obtain at least residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or to homogeneity by SDS-PAGE under non-reducing or reducing conditions using Coomassie blue or, preferably, silver stain. Isolated polypeptide includes polypeptide in situ within recombinant cells, since at least one component of the vertebrate fused natural environment will not be present. Ordinarily, however, isolated polypeptide will be prepared by at least one purification step.
An "isolated" vertebrate fused nucleic acid molecule is a nucleic acid molecule that is identified and separated from at least one contaminant nucleic acid molecule with which it is ordinarily associated in the natural source of the vertebrate fused nucleic acid. An isolated vertebrate fused nucleic acid molecule is other than in the form or setting in which it is found in nature. Isolated vertebrate fused nucleic acid molecules therefore are distinguished from the corresponding native vertebrate fused nucleic acid molecule as it exists in natural cells.
The term "control sequences" refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism. The control sequences that are suitable for prokaryotes, for example, include a promoter, optionally an operator sequence, and a ribosome binding site. Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
Nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. For example, DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Generally, "operably linked" means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
The term "antibody" is used in the broadest sense and specifically covers single monoclonal antibodies (including agonist and antagonist antibodies), antibody compositions with polyepitopic specificity, as well as antibody fragments Fab, F(ab') 2 and Fv), so long as they exhibit the desired biological activity.
The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, the individual antibodies comprising the population are identical 9 \\melbfiles\homeS\suzannet\Keep\Speci\28779-99.1 SPECI.doc 10/05/01 P1272 except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler Milstein, Nature 256:495 (1975), or may be made by recombinant DNA methods [see, e.g. U.S. Patent No. 4,816,567 (Cabilly et The monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity Patent No. 4,816,567; Cabilly et al.; Morrison et Proc. Natl. Acad. Sci. USA 81, 6851-6855 (1984)].
"Humanized" forms of non-human murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab') 2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity. In some instances, corresponding non-human residues replace Fv framework residues of the human immunoglobulin. Furthermore, humanized antibody may comprise residues that are found neither in.
the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region typically that of a human immunoglobulin. For further details see: Jones et al., Nature 321, 522-525 (1986); Reichmann et al., Nature 332, 323-329 (1988); Presta, Curr. Op. Struct. Biol. 2 593-596 (1992) and U.S. Patent No. 5,225,539 (Winter) issued July 6, 1993.
"Active" or "activity" for the purposes herein refers to form(s) of vertebrate fused which retain the biologic and/or immunologic activities of native or naturally occurring vertebrate fused. A preferred activity is the ability to bind to and affect, block or otherwise modulate, hedgehog signaling. The activity preferably involves the regulation of the pathogenesis of Basal cell carcinoma. Another preferred biological P1272 activity is the ability to phosphorylate or modulate the phosphorylation of Gli.
The term "antagonist" is used herein in the broadest sense to include any molecule which blocks, prevents, inhibits, neutralizes the normal functioning of fused in the Hh signaling pathway. One particular form of antagonist includes a molecule that interferes with the interaction between fused and its binding or complexing proteins. In a similar manner, the term "agonist" is used herein to include any molecule which promotes, enhances or stimulates the normal functioning of fused in the Hh signaling pathway. Suitable molecules that affect the protein-protein interaction of fused and its binding proteins include fragments of the latter or small bioorganic molecules, peptidomimetics, which will prevent or enhance, as the case may be, the interaction of proper complex formation. Non-limiting examples include proteins, peptides, glycoproteins, glycopeptides, glycolipids, polysaccharides, oligosaccharides, nucleic acids, bioorganic molecules, peptidomimetics, pharmacological agents and their metabolites, transcriptional and translation control sequences, and the like. Another preferred form of antagonist includes antisense nucleotides that inhibit proper transcription of wild type fused. Preferred forms of antagonists and are small molecules, which specifically bind to or block binding of the ATP binding site of fused.
The term "modulation" or "modulating" means upregulation or downregulation of a signaling pathway. Cellular processes under the control of signal transduction may include, but are not limited to, transcription of specific genes; normal cellular functions, such as metabolism, proliferation, differentiation, adhesion, apoptosis and survival, as well as abnormal processes, such as transformztion, blocking of differentiation and metastasis.
The techniques of "polymerase chain reaction," or "PCR", as used herein generally refers to a procedure wherein minute amounts of a specific piece of nucleic acid, RNA and/or DNA are amplified as described in U.S. Pat. No. 4,683,195 issued 28 July 1987. Generally, sequence information from the ends of the region of interest or beyond needs to be available, such that oligonucleotide primers can be designed; these primer will be identical or similar in sequence to opposite strands of the template to be amplified. The 5' terminal nucleotides of the two primers may coincide with the ends of the amplified material. PCR sequences form total genomic DNA, and cDNA transcribed from total cellular RNA, bacteriophage, or plasmid sequences, etc. See generally Mullis et al., Cold Spring Harbor Symp. Quant. Biol. 51: 263 (1987); Erlich, Ed., PCR Technology, (Stockton Press, NY, 1989). As used herein, PCR is considered to be one, but not the only, example of a nucleic acid test sample comprising the use of a known nucleic acid as a primer and a nucleic acid polymerase to amplify or generate a specific piece of nucleic acid.
II1. Compositions and Methods of the Invention A. Full-length vertebrate fused The present invention provides newly identified and isolated nucleotide sequences encoding polypeptides referred to in the present application as human and vertebrate fused. In particular, Applicants have identified and isolated cDNA encoding a vertebrate fused polypeptide, as disclosed in further detail in the Examples below. Using BLAST, BLAST-2 and FastA sequence alignment computer programs, Applicants found that a full-length native sequence human fused (shown in Figure 3 (SEQ ID NO has 28% amino acid sequence identity with Drosophila fused (SEQ ID NO 23). Accordingly, it is presently believed that the human fused disclosed in the present application is a newly identified member of the P1272 hedgehog signaling cascade.
The full-length native sequence of human vertebratefused gene, or portions thereof, may be used as hybridization probes for a cDNA library to isolate the full-length gene or to isolate still other vertebrate homolog genes (for instance, those encoding naturally-occurring variants of vertebrate fused or vertebrate fused from other species) which have a desired sequence identity to the vertebrate fused sequence disclosed in Fig.] (SEQ ID NO Optionally, the length of the probes will be about 20 to about 50 bases. The hybridization probes may be derived from the nucleotide sequence of Fig. 1 (SEQ ID NO 1) or from genomic sequences including promoters, enhancer elements and introns of native sequence vertebrate fused.
.By way of example, a screening method will comprise isolating the coding region of the vertebrate fused gene using the known DNA sequence to synthesize a selected probe of about 40 bases. Hybridization probes may be labeled by a variety of labels, including radionucleotides such as 32P or 35S, or enzymatic labels such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems. Labeled probes having a sequence complementary to that of the vertebrate fused gene of the present invention can be used to screen libraries of human cDNA, genomic DNA or mRNA to determine which members of such libraries the probe hybridizes to.
B. Vertebrate fused Variants In addition to the full-length native sequence vertebrate fused described herein, it is contemplated that vertebrate fused variants can be prepared. Vertebrate fused variants can be prepared by introducing appropriate nucleotide changes into a known vertebrate fused DNA, or by synthesis of the desired vertebrate fused polypeptides. Those skilled in the art will appreciate that amino acid changes may alter posttranslational processes of the vertebrate fused.
Variations in the native full-length sequence vertebratefusedor in various domains of the vertebrate fused described herein, can be made, for example, using any of the techniques and guidelines for conservative and non-conservative mutations set forth, for instance, in U.S. Patent No. 5,364,934. Variations may be a substitution, deletion or insertion of one or more codons encoding the vertebratefused that results in a change in the amino acid sequence of the vertebrate fused as compared with the native sequence vertebrate fused. Optionally the variation is by substitution of at least one amino acid with any other amino acid in one or more of the domains of the vertebrate fused. Guidance in determining which amino acid residue may be inserted, substituted or deleted without adversely affecting the desired activity may be found by comparing the sequence of the vertebrate fused with that of homologous known protein molecules and minimizing the number of amino acid sequence changes made in regions of high homology. Amino acid substitutions can be the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, such as the replacement of a leucine with a serine, conservative amino acid replacements. Insertions or deletions may optionally be in the range of I to 5 amino acids. The variation allowed may be determined by systematically making insertions, deletions or substitutions of amino acids in the sequence and testing the resulting variants for activity in the in vitro assay described in the Examples below.
The variations can be made using methods known in the art such as oligonucleotide-mediated (site- P1272 directed) mutagenesis, alanine scanning, and PCR mutagenesis. Site-directed mutagenesis [Carter et al., Nucl. Acids Res., 13:4331 (1986); Zoller et al., Nucl. Acids Res., 10:6487 (1987)], cassette mutagenesis [Wells et al., Gene, 34:315 (1985)], restriction selection mutagenesis [Wells et al., Philos. Trans. R. Soc.
London SerA, 317:415 (1986)] or other known techniques can be performed on the cloned DNA to produce the vertebrate fused variant DNA.
Scanning amino acid analysis can also be employed to identify one or more amino acids along a contiguous sequence. Among the preferred scanning amino acids are relatively small, neutral amino acids.
Such amino acids include alanine, glycine, serine, and cysteine. Alanine is typically a preferred scanning amino acid among this group because it eliminates the side-chain beyond the beta-carbon and is less likely to alter the main-chain conformation of the variant. Alanine is also typically preferred because it is the most common amino acid. Further, it is frequently found in both buried and exposed positions [Creighton, The Proteins, Freeman Co., Chothia, J. Mol. Biol., 150:1 (1976)]. If alanine substitution does not yield adequate amounts of variant, an isoteric amino acid can be used.
In the human fused sequence depicted in Figure 1, the kinase domain is represented by amino acid residues 1-260 (SEQ ID NO 24) of which position lysine 33 appears to be necessary for ATP binding and thus enzymatic activity.
C. Modifications of vertebrate fused Covalent modifications of vertebrate fused are included within the scope of this invention. One type of covalent modification includes reacting targeted amino acid residues of the vertebrate fused with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C- terminal residues of the vertebrate fused. Derivatization with bifunctional agents is useful, for instance, for crosslinking vertebrate fused to a water-insoluble support matrix or surface for use in the method for purifying anti-vertebrate fused antibodies, and vice-versa. Commonly used crosslinking agents include, e.g., 1,1-bis(diazo-acetyl)-2-phenylethane, glutaraldehyde, N-hydroxy-succinimide esters, for example, esters with 4-azido-salicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3'dithiobis-(succinimidyl-propionate), bifunctional maleimides such as bis-N-maleimido-1,8-octane and agents such as methyl-3-[(p-azidophenyl)-dithio]pro-pioimi-date.
Other modifications include deamidation of glutaminyl and asparaginyl residues to the corresponding glutamyl and aspartyl residues, respectively, hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the a-amino groups of lysine, arginine, and histidine side chains Creighton, Proteins: Structure and Molecular Properties, W.H. Freeman Co., San Francisco, pp. 79-86 (1983)], acetylation of the N-terminal amine, and amidation of any C-terminal carboxyl group.
Another type of covalent modification of vertebrate fused comprises linking the vertebrate fused polypeptide to one of a variety of nonproteinaceous polymers, polyethylene glycol, polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Patent Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192 or 4,179,337. Such modifications would be expected in increase the half-life of the molecules in circulation in a mammalian system; Extended half-life of fused molecules might be useful under certain circumstances, such as where the fused variant is administered as a therapeutic agent.
P1272 The vertebrate fused of the present invention may also be modified in a way to form a chimeric molecule comprising vertebrate fused bonded to another, heterologous polypeptide or amino acid sequence.
In one embodiment, such a chimeric molecule comprises a fusion of the vertebrate fused with a tag polypeptide, which provides an epitope to which an anti-tag antibody can selectively bind. The epitope tag is generally placed at the amino- or carboxyl- terminus of the vertebrate fused. The presence of such epitopetagged forms of the vertebrate fused can be detected using an antibody against the tag polypeptide. Also, provision of the epitope tag enables the vertebrate fused to be readily purified by affinity purification using an anti-tag antibody or another type of affinity matrix that binds to the epitope tag. In an alternative embodiment, the chimeric molecule may comprise a fusion of the vertebrate fused with an immunoglobulin or a particular region of an immunoglobulin. For a bivalent form of the chimeric molecule, such a fusion could be to the Fc region of an IgG molecule.
Ordinarily, the C-terminus of a contiguous amino acid sequence of a ligand- (IFN-y-) binding domain of an IFNy receptor is fused to the N-terminus of a contiguous amino acid sequence of an immunoglobulin constant region, in place of the variable region(s), however N-terminal fusions are also possible.
Typically, such fusions retain at least functionally active hinge, CH2 and CH3 domains of the constant region of an immunoglobulin heavy chain. Fusions are also made to the C-terminus of the Fc portion of a constant domain, or immediately N-terminal to the CH I of the heavy chain or the corresponding region of the light chain. This ordinarily is accomplished by constructing the appropriate DNA sequence and expressing it in recombinant cell culture. Alternatively, immunoadhesinsmay be synthesized accordingto known methods.
The precise site at which the fusion is made is not critical; particular sites are well known and may be selected in order to optimize the biological activity, secretion or binding characteristicsof the immunoadhesins.
In a preferred embodiment, the C-terminus of a contiguous amino acid sequence which comprises the binding site(s) for IFN-y is fused, at the N-terminal end, to the C-terminal portion of an antibody (in particular the Fc domain), containing the effector functions of an immunoglobulin, e.g. immunoglobulin G, (IgG- As hereinabove mentioned, it is possible to fuse the entire heavy chain constant region to the sequence containing the binding site(s). However, more preferably, a sequence beginning in the hinge region just upstream of the papain cleavage site (which defines IgG Fc chemically; residue 216, taking the first residue of heavy chain constant region to be 114 [Kobet et al., supra], or analogous sites of other immunoglobulins) is used in the fusion. Although it was earlier thought that in immunoadhesins the immunoglobulin light chain would be required for efficient secretion of the heterologous protein-heavy chain fusion proteins, it has been found that even the immunoadhesins containing the whole IgG 1 heavy chain are efficiently secreted in the absence of light chain. Since the light chain is unnecessary, the immunoglobulin heavy chain constant domain sequence used in the construction of the immunoadhesins of the present invention may be devoid of a light chain binding site.
This can be achieved by removing or sufficiently altering immunoglobulin heavy chain sequence elements to which the light chain is ordinarily linked so that such binding is no longer possible. Thus, the CH I domain can be entirely removed in certain embodiments of the IFN-y receptor-immunoglobulinchimeras.
In a particularly preferred embodiment, the amino acid sequence containing the extracellulardomain of an IFN-y receptor is fused to the hinge region and CH2, CH3; or CH 1, hinge, CH2 and CH3 domains of an IgG- 1, lgG-2, lgG-3, or IgG-4 heavy chain. The construction of a typical structure is disclosed in Example 1.
P1272 In some embodiments, the IFN-y receptor-immunoglobulin molecules (immunoadhesins) are assembled as monomers, dimers or multimers, and particularly as dimers or tetramers. Generally, these assembled immunoadhesins will have known unit structures similar to those of the corresponding immunoglobulins. A basic four chain structural unit (a dimer of two immunoglobulin heavy chain-light chain pairs) is the form in which IgG, IgA and IgE exist. A four chain unit is repeated in the high molecular weight immunoglobulins;IgM generally exists as a pentamer of basic four-chain units held together by disulfide bonds.
IgA globulin, and occasionally IgG globulin, may also exist in a multimeric form in serum. In the case of multimers, each four chain unit may be the same or different.
It is not necessary that the entire immunoglobulin portion of the IFN-y receptor-immunoglobulin chimeras be from the same immunoglobulin. Various portions of different immunoglobulinsmay be combined, and variants and derivatives of native immunoglobulins can be made as hereinabove described with respect to IFN-y, in order to optimize the properties of the immunoadhesin molecules. For example, immunoadhesin constructs in which the hinge of IgG- I1 was replaced with that of IgG-3 were found to be functional and showed pharmacokineticscomparableto those of immunoadhesinscomprisingthe entire IgG-I heavy chain.
Various tag polypeptides and their respective antibodies are well known in the art. Examples include poly-histidine (poly-his) or poly-histidine-glycine (poly-his-gly) tags; the flu HA tag polypeptide and its antibody 12CA5 [Field et al., Mol. Cell. Biol., 8:2159-2165 (1988)]; the c-myc tag and the 8F9, 3C7, 6E10, G4, B7 and 9E10 antibodies thereto [Evan et al., Molecular and Cellular Biology, 5:3610-3616 (1985)]; and the Herpes Simplex virus glycoprotein D (gD) tag and its antibody [Paborsky et al., Protein Engineering, 3(6):547-553 (1990)]. Other tag polypeptides include the Flag-peptide [Hopp et al., BioTechnology, 6:1204-1210 (1988)]; the KT3 epitope peptide [Martin et al., Science, 255:192-194 (1992)]; an a-tubulin epitope peptide [Skinner et al., J. Biol. Chem., 266:15163-15166 (1991)]; and the T7 gene protein peptide tag [Lutz-Freyermuth et al., Proc. Natl. Acad. Sci. USA, 87:6393-6397 (1990)]. A preferred tag is the influenza HA tag.
D. Preparation of vertebrate fused The description below relates primarily to production of a particular vertebrate fused by culturing cells transformed or transfected with a vector containing vertebrate fused nucleic acid. It is, of course, contemplated that alternative methods, which are well known in the art, may be employed to prepare vertebrate fused. For instance, the vertebrate fused sequence, or portions thereof, may be produced by direct peptide synthesis using solid-phase techniques [see, Stewart et al., Solid-Phase Peptide Synthesis, W.H.
Freeman Co., San Francisco, CA (1969); Merrifield, J. Am. Chem. Soc., 85:2149-2154 (1963)]. In vitro protein synthesis may be performed using manual techniques or by automation. Automated synthesis may be accomplished, for instance, using an Applied Biosystems Peptide Synthesizer (Foster City, CA) using manufacturer's instructions. Various portions of the vertebrate fused may be chemically synthesized separately and combined using chemical or enzymatic methods to produce the full-length vertebrate fused.
I. Isolation of DNA Encoding vertebrate fused DNA encoding vertebrate fused may be obtained from a cDNA library prepared from tissue believed to possess the vertebrate fused mRNA and to express it at a detectable level. Accordingly, human vertebrate fused DNA can be conveniently obtained from a cDNA library prepared from human tissue, such P1272 as described in the Examples. The vertebrate fused-encoding gene may also be obtained from a genomic library or by oligonucleotide synthesis.
Libraries can be screened with probes (such as antibodies to the vertebrate fused or oligonucleotides of at least about 20-80.bases) designed to identify the gene of interest or the protein encoded by it. Screening the cDNA or genomic library with the selected probe may be conducted using standard procedures, such as described in Sambrook et al., Molecular Cloning: A Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989). An alternative means to isolate the gene encoding vertebrate fused is to use PCR methodology [Sambrook et al., supra; Dieffenbach et al., PCR Primer: A Laboratory Manual (Cold Spring Harbor Laboratory Press, 1995)].
The Examples below describe techniques for screening a cDNA library. The oligonucleotide sequences selected as probes should be of sufficient length and sufficiently unambiguous that false positives are minimized. The oligonucleotide is preferably labeled such that it can be detected upon hybridization to DNA in the library being screened. Methods of labeling are well known in the art, and include the use of radiolabels like "P-labeled ATP, biotinylation or enzyme labeling. Hybridization conditions, including moderate stringency and high stringency, are provided in Sambrook et al., supra.
Sequences identified in such library screening methods can be compared and aligned to other known sequences deposited and available in public databases such as GenBank or other private sequence databases. Sequence identity (at either the amino acid or nucleotide level) within defined regions of the molecule or across the full-length sequence can be determined through sequence alignment using computer software programs such as BLAST, BLAST-2, ALIGN, DNAstar, and INHERIT which employ various algorithms to measure homology.
Nucleic acid having protein coding sequence may be obtained by screening selected cDNA or genomic libraries using the deduced amino acid sequence disclosed herein for the first time, and, if necessary, using conventional primer extension procedures as described in Sambrook et al., supra, to detect precursors and processing intermediates of mRNA that may not have been reverse-transcribed into cDNA.
2. Selection and Transformation of Host Cells Host cells are transfected or transformed with expression or cloning vectors described herein for vertebrate fused production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
The culture conditions, such as media, temperature, pH and the like, can be selected by the skilled artisan without undue experimentation. In general, principles, protocols, and practical techniques for maximizing the productivity of cell cultures can be found in Mammalian Cell Biotechnology: A Practical Approach, M.
Butler, ed. (IRL Press, 1991) and Sambrook et al., supra.
Methods of transfection are known to the ordinarily skilled artisan, for example, CaPO, and electroporation. Depending on the host cell used, transformation is performed using standard techniques appropriate to such cells. The calcium treatment employing calcium chloride, as described in Sambrook et al., supra, or electroporation is generally used for prokaryotes or other cells that contain substantial cell-wall barriers. Infection with Agrobacterium tumefaciens is used for transformation of certain plant cells, as described by Shaw et al., Gene, 23:315 (1983) and WO 89/05859 published 29 June 1989. For mammalian P1272 cells without such cell walls, the calcium phosphate precipitation method of Graham and van der Eb, Virology 52:456-457 (1978) can be employed. General aspects of mammalian cell host system transformations have been described in U.S. Patent No. 4,399,216. Transformations into yeast are typically carried out according to the method of Van Solingen et al., J. Bact., 130:946 (1977) and Hsiao et al., Proc.
Natl. Acad Sci. (USA), 76:3829 (1979). However, other methods for introducing DNA into cells, such as by nuclear microinjection, electroporation, bacterial protoplast fusion with intact cells, or polycations, e.g., polybrene, polyornithine, may also be used. For various techniques for transforming mammalian cells, see Keown et al., Methods in Enzymology, 185:527-537 (1990) and Mansour et al., Nature, 336:348-352 (1988).
Suitable host cells for cloning or expressing the DNA in the vectors herein include prokaryote, yeast, or higher eukaryote cells. Suitable prokaryotes include but are not limited to eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriae such as E. coli. Various E. coli strains are publicly available, such as E. coli K12 strain MM294 (ATCC 31,446); E. coli X1776 (ATCC 31,537); E. coli strain W3110 (ATCC 27,325) and K5 772 (ATCC 53,635).
In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for vertebrate fused-encoding vectors. Saccharomyces cerevisiae is a commonly used lower eukaryotic host microorganism.
Suitable host cells for the expression of vertebrate fused are derived from multicellular organisms.
Examples of invertebrate cells include insect cells such as Drosophila S2 and Spodoptera Sf9, as well as plant cells. Examples of useful mammalian host cell lines include Chinese hamster ovary (CHO) and COS cells. More specific examples include monkey kidney CVI line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol., 36:59 (1977)); Chinese hamster ovary cells/-DHFR (CHO, Urlaub and Chasin, Proc.
Natl. Acad. Sci. USA, 77:4216 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod 23:243-251 (1980)); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); and mouse mammary tumor (MMT 060562, ATCC CCL51). The selection of the appropriate host cell is deemed to be within the skill in the art.
3. Selection and Use of a Replicable Vector The nucleic acid cDNA or genomic DNA) encoding vertebrate fused may be inserted into a replicable vector for cloning (amplification of the DNA) or for expression. Various vectors are publicly available. The vector may, for example, be in the form of a plasmid, cosmid, viral particle, or phage. The appropriate nucleic acid sequence may be inserted into the vector by a variety of procedures. In general, DNA is inserted into an appropriate restriction endonuclease site(s) using techniques known in the art. Vector components generally include, but are not limited to, one or more of a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence. Construction of suitable vectors containing one or more of these components employs standard ligation techniques, which are known to the skilled artisan.
Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Such sequences are well known for a variety of bacteria, yeast, and viruses. The origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, P1272 the 2: plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells. A preferred replicable expression vector is the plasmid is pRK5. Holmes et al., Science, 253:1278-1280 (1991).
Expression and cloning vectors will typically contain a selection gene, also termed a selectable marker. Typical selection genes encode proteins that confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, complement auxotrophic deficiencies, or supply critical nutrients not available from complex media, the gene encoding D-alanine racemase for Bacilli.
An example of suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the vertebrate fused nucleic acid, such as DHFR or thymidine kinase. An appropriate host cell when wild-type DHFR is employed is the CHO cell line deficient in DHFR activity, prepared and propagated as described by Urlaub et al., Proc. Natl. Acad. Sci. USA, 77:4216 (1980).
A suitable selection gene for use in yeast is the trpl gene present in the yeast plasmid YRp7 [Stinchcomb et al., Nature, 282:39 (1979); Kingsman et al., Gene, 2:141 (1979); Tschemper et al., Gene, 10:157 (1980)].
The trpl gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1 [Jones, Genetics, 85:12 (1977)].
Expression and cloning vectors usually contain a promoter operably linked to the vertebrate fused nucleic acid sequence to direct mRNA synthesis. Promoters recognized by a variety of potential host cells are well known. Promoters suitable for use with prokaryotic hosts include the p-lactamase and lactose promoter systems [Chang et al., Nature, 275:615 (1978); Goeddel et al., Nature, 281:544 (1979)], alkaline phosphatase, a tryptophan (trp) promoter system [Goeddel, Nucleic Acids Res., 8:4057 (1980); EP 36,776], and hybrid promoters such as the tac promoter [deBoer et al., Proc. Natl. Acad. Sci. USA, 80:21-25 (1983)].
Promoters for use in bacterial systems also will contain a Shine-Dalgarno sequence operably linked to the DNA encoding vertebrate fused.
Examples of suitable promoting sequences for use with yeast hosts include the promoters for 3phosphoglycerate kinase [Hitzeman et al., J. Biol. Chem_, 255:2073 (1980)] or other glycolytic enzymes [Hess et al., J. Adv. Enzyme Reg., 7:149 (1968); Holland, Biochemistry, 17:4900 (1978)], such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phospho-fructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
Other yeast promoters, which are inducible promoters having the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Suitable vectors and promoters for use in yeast expression are further described in EP 73,657.
Vertebrate fused transcription from vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus (UK 2,211,504 published 5 July 1989), adenovirus (such as Adenovirus bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, the actin promoter or an immunoglobulin promoter, and from heat-shock promoters, provided such promoters are compatible with the host cell systems.
Inserting an enhancer sequence into the vector may increase transcription of a DNA encoding the vertebrate fused by higher eukaryotes. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription. Many enhancer sequences are now known from mammalian genes (globin, elastase, albumin, a-fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus. Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers. The enhancer may be spliced into the vector at a position 5' or 3' to the vertebrate fused coding sequence, but is preferably located at a site 5' from the promoter.
Expression vectors used in eukaryotic host cells (yeast, fungi, insect, plant, animal, human, or nucleated cells from other multicellular organisms) will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5' and, occasionally 3, untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding vertebrate fused.
Still other methods, vectors, and host cells suitable for adaptation to the synthesis of vertebrate fused in recombinant vertebrate cell culture are described in Gething et al., Nature, 223:620-625 (1981); Mantei et al., Nature, 281:40-46 (1979); EP 117,060; and EP 117,058.
For the purposes of this specification it will be clearly understood that the word "comprising" means "including but not limited to", and that the word "comprises" has a corresponding meaning.
1. Detecting Gene Amplification/Expression Gene amplification and/or expression may be measured in a sample directly, for example, by conventional Southern blotting, Northern blotting to quantitate the transcription of mRNA [Thomas, Proc, Natl.
Acad. Sci. USA, 12:5201-5205 (1980)], dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein. Alternatively, antibodies may be employed that can recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. The antibodies in turn may be labeled and the assay may be carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.
Gene expression, alternatively, may be measured by immunological methods, such as immunohistochemical staining of cells or tissue sections and assay of cell culture or body fluids, to quantitate directly the expression of gene product. Antibodies useful for immunohistochemical staining and/or assay of sample fluids may be either monoclonal or polyclonal, and may be prepared in any mammal. Conveniently, the antibodies may be prepared against a native sequence vertebrate fused polypeptide or against a synthetic peptide based on the DNA sequences provided herein or against exogenous sequence fused to vertebrate fused DNA and encoding a specific antibody epitope.
2. Purification of Polypeptide 19 \\nlelb-.files\homes\suzannet\Keep\Speci\28779-99 .1 SPECI .doc 10/05/01 Forms of vertebratefused may be recovered from host cell lysates. If membrane-bound, it can be released from the membrane using a suitable detergent solution Triton-X 100) or by enzymatic 1 9a \\melbfiles\hone$\euzannet\Keep\speci\28779..99.1 SPECI .doc 10/05/01 P1272 cleavage. Cells employed in expression of vertebrate fused can be disrupted by various physical or chemical means, such as freeze-thaw cycling, sonication, mechanical disruption, or cell lysing agents.
It may be desired to purify vertebrate fused from recombinant cell proteins or polypeptides. The following procedures are exemplary of suitable purification procedures: by fractionation on an ion-exchange column; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation-exchange resin such as DEAE; chromatofocusing; SDS-PAGE; ammonium sulfate precipitation; gel filtration using, for example, Sephadex G-75; protein A Sepharose columns to remove contaminants such as IgG; and metal chelating columns to bind epitope-tagged forms of the vertebrate fused. Various' methods of protein purification may be employed and such methods are known in the art and described for example in Deutscher, Methods in Enzymology, 182 (1990); Scopes, Protein Purification: Principles and Practice, Springer-Verlag, New York (1982). The purification step(s) selected will depend, for example, on the nature of the production process used and the particular vertebrate fused produced.
E. Uses for vertebrate fused Fused is universal mediator ofHh signaling The human fused full length molecule of (Fig. I (SEQ ID NO encodes a protein with a predicted molecular weight of 150 kDa, which is significantly larger that Drosophila fused (100 kDa, dfused (SEQ ID NO Human fused (hfused) shows notable homology to the Drosophila homologue in the kinase domain, but little 'homology with dfused or any other known protein over the remaining =1000 amino acids. The kinase domain extends from residue I to about residue 260, as is represented in Fig. I (SEQ ID NOS. 24 This divergence at the C-terminus of the molecules is unexpected given that the C-terminus of the Drosophila molecule is required for its activity, Preat et al., Nature 347: 87-9 (1990). An ATP binding site is at about amino acid position 33 and is required for kinase activity.
Prior studies in Drosophila indicate that dfused is necessary for Hh signal to occur but have not addressed the issue whetherfused is sufficient to activate this signaling system. As depicted in the Examples, applicants have herein used a Gli DNA binding element present in the HNF3P promoter, in front of a luciferase mediator of the Hh cascade, which clearly demonstrates that fused alone is capable of activating Gli mediated transcription in this system. It is further apparent that both an intact kinase domain and an intact C-terminal non-catalytic domain are required for this activation, which supports the notion that fused functions as a kinase and that the C-terminus may play a role in the substrate recognition or in regulating the kinase activity.
Applicants have shown in the present application that hfused is a kinase which is capable of phosphorylating artificial substrates such as MBP. However, the identity of the physiological substrate for hfused remains to be determined. One obvious candidate is Gli-I itself, as Gli-I phosphorylation by hfused can be detected in vitro.
To determine if human fused is essential for Hh signaling in vertebrates, a mutant was constructed by altering a conserved lysine in the ATP binding site (about amino acid residue 33). Typically, such mutants act as inhibitor of the corresponding wild type kinase by blocking access to substrate and/or regulatory factors, He et al., Nature 374, 617-22 (1995). When overexpressed in 2-cell stage Xenopus embryos, the most remarkable phenotype was the presence of fused eyes in about 30% of the injected P1272 embryos. Several lines of evidence indicate that this phenotype is likely to result from the inhibition of Hh signaling. First, SHh knockouts display a cyclopia phenotype attributed recently to mutations in the SHh gene, Chiang et al., Nature 383: 407-13 (1996). Second, zebrafish embryos (cyclops) with reduced expression of SHh or injected with constitutively active form of PKA, a negative regulator of the Hh pathway are cyclops. Third, SHh, emanating from prechordal plate, has been shown to inhibit expression of Pax-6, a key transcription factor required for eye development, in the center of a continuous eyefield, Ekker et al., Curr. Biol. 5: 944-55 (1995); Li et al., Development 124: 603-15 (1997); Macdonald et al., Development 121: 3267-78 (1995). Finally, staining for Pax-6 embryos injected with fused-DN revealed a single field of expression suggesting a failure of SHh emanating from the prechordal plate to downregulate the expression of Pax-6 at the center of the eyefield.
To confirm the position of fused in the Hh signaling pathway, expression of SHh in the floor plate of Xenopus embryos injected with hfused-DN could be rescued by coinjection of Gli-1. This suggests that fused acts in association with Gli in the SHh signaling pathway.
The tissue distribution of fused shows that it is expressed in all SHh responsive cells. In particular, its expression pattern overlaps well with Ptch, the binding component of the Hh receptor which is itself a target gene of the SHh signaling pathway. These data suggest that fused is involved in mediating a wide variety of effect SHh has on different tissues. Functionally, this was observed again in frog embryos where, fused-DN inhibited eye development as well as SHh expression in the floor plate.
hFused-DN also appears to affect normal development of tissues such as the frog gut which is regulated by Indian Hh. This, combined with the fact that fused is expressed in the gut and testis, sites of IHh and DHh action respectively, suggest that fused may be a universal mediator of signaling for all members of the Hh protein family.
Very high levels of fused mRNA was found on germ cell, the development of which appears to be regulated by DHh. Homozygous mutant mice for DHh fail to develop germ cells and are viable but sterile (Bitgood et al., Curr. Biol. 6: 298-304 (1996). However, Patched, a Hedgehog receptor is expressed on interstitial Leydig cells and not on germ cells where fused is expressed, Bitgood et al, supra. I his discrepancy suggests that there may be additional hedgehog receptors.
Applicants have shown in the Examples that wild type hfused is capable of activating Gli in a reporter assay. Furthermore, expression of SHh in the floor plate of frog embryos injected with hfused-DN could be rescued by coinjection of Gli-1. Taken together- these observations are consistent with the assertion that fused acts downstream of Smo and upstream of Gli in this signaling pathway, which is consistent with the genetic evidence in Drosophila to date.
General uses for vertebrate fused Nucleotide sequences (or their complement) encoding vertebrate fused have various applications in the art of molecular biology, including uses as hybridization probes, in chromosome and gene mapping and in the generation of anti-sense RNA and DNA. Vertebratefused nucleic acid will also be useful for the preparation of vertebratefused polypeptides by the recombinant techniques described herein.
The full-length native sequence vertebrate fused gene, or portions thereof, may be used as hybridization probes for a cDNA library to isolate the full-length gene or to isolate still other genes (for instance, those encoding naturally-occurring variants of vertebrate fused or vertebrate fused from other species) which have a desired sequence identity to the vertebrate fused sequence disclosed in Fig.1 (SEQ ID NO 1).
Optionally, the length of the probes will be about 20 to about 50 bases. The hybridization probes may be derived from the nucleotide sequence of Fig. 1 (SEQ ID NO 1) or from genomic sequences including promoters, enhancer elements and introns of native sequence vertebrate fused. By way of example, a screening method will comprise isolating the coding region of the vertebrate fused gene using the known DNA sequence to synthesize a selected probe of about 40 bases. Hybridization probes may be labeled by a variety of labels, including radionucleotides such as 32P or 35S, or enzymatic labels such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems. Labeled probes having a sequence complementary to that of the vertebrate fused gene of the present invention can be used to screen libraries of human cDNA, genomic DNA or mRNA to determine to which members of such libraries the probe hybridizes. Hybridization techniques are described in further detail in the Examples below.
The probes may also be employed in PCR techniques to generate a pool of sequences for identification of closely related vertebrate fused sequences.
Nucleotide sequences encoding a vertebrate fused can also be used to construct hybridization probes for mapping the gene, which encodes vertebrate fused and for the genetic analysis of individuals with genetic disorders. The nucleotide sequences provided herein may be mapped to a chromosome and specific regions of a chromosome using known techniques, such as in situ hybridization, linkage analysis against known chromosomal markers, and hybridization screening with libraries.
Vertebrate fused polypeptides can be used in assays to identify the other proteins or molecules involved in complexing with fused which ultimately results in the modulation of hedgehog signaling. Alternatively, these molecules can modulate the fused kinase phosphorylation of its substrate. By such methods, inhibitors of the binding interaction can be identified. Proteins involved in such binding interactions can also be used to screen for peptide or small molecule inhibitors or agonists of the binding interaction. Also, the substrate of vertebrate fused can be used to isolate correlative complexing proteins. Screening assays can be designed to find lead compounds that mimic the biological activity of a native vertebrate fused or to find those that act as a substrate for vertebrate fused. Such screening assays will include assays amenable to high-throughput screening of chemical libraries, making them particularly suitable for identifying small molecule drug candidates. Such small molecule inhibitors could block the enzymatic action of fused, and thereby inhibit hedgehog signaling. Small molecules contemplated include synthetic organic or inorganic compounds. The assays can be performed in a variety of formats, including protein-protein binding assays, biochemical screening assays, immunoassays and cell based assays, which are well characterized in the art.
Nucleic acids which encode vertebrate fused or its modified forms can also be used to generate either transgenic animals or "knock out" animals which, in turn, are useful in the development and screening of therapeutically useful reagents. A transgenic animal a mouse or rat) is an animal having cells that contain a transgene, which transgene was introduced into the animal or an ancestor of the animal at a prenatal, an embryonic stage. A transgene is a DNA that is integrated into the genome of a cell from 22 \\melb....iles\home$\suzannet\Keep\Speci\28779-99.1 SPECI.doc 10/05/01 P1272 which a transgenic animal develops. In one embodiment, cDNA encoding vertebrate fused can be used to clone genomic DNA encoding vertebrate fused in accordance with established techniques and the genomic sequences used to generate transgenic animals that contain cells which express DNA encoding vertebrate fused. Methods for generating transgenic animals, particularly animals such as mice or rats, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866 and 4,870,009.
Typically, particular cells would be targeted for vertebrate fused transgene incorporation with tissue-specific enhancers. Transgenic animals that include a copy of a transgene encoding vertebrate fused introduced into the germ line of the animal at an embryonic stage can be used to examine the effect of increased expression of DNA encoding vertebrate fused. Such animals can be used as tester animals for reagents thought to confer protection from, for example, pathological conditions associated with its overexpression. For example, for basal cell carcinoma, fused can be overexpressed in the basal cell layer of the skin using a Keratin 5 or 14 promoter. In accordance with this facet of the invention, an animal is treated with the reagent and a reduced incidence of the pathological condition, compared to untreated animals bearing the transgene, would indicate a potential therapeutic intervention for the pathological condition.
Non-human homologues of vertebrate fused can be used to construct a vertebrate fused "knock out" animal which has a defective or altered gene encoding vertebrate fused as a result of homologous recombination between the endogenous gene encoding vertebrate fused and altered genomic DNA encoding vertebrate fused introduced into an embryonic cell of the animal. For example, cDNA encod;ng.vertebrate fused can be used to clone genomic DNA encoding vertebrate fused in accordance with, established techniques. A portion of the genomic DNA encoding vertebrate fused can be deleted or replaced with another gene, such as a gene encoding a selectable marker that can be used to monitor integration. Typically, several kilobases of unaltered flanking DNA (both at the 5' and 3' ends) are included in the vector [see e.g., Thomas and Capecchi, Cell, 51:503 (1987) for a description of homologous recombination vectors]. The vector is introduced into an embryonic stem cell line by electroporation) and cells in which the introduced DNA has homologously recombined with the endogenous DNA are selected [see Li et al., Cell, 69:915 (1992)]. The selected cells are then injected into a blastocyst of an animal a mouse or rat) to form aggregation chimeras [see Bradley, in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E. J. Robertson, ed. (IRL, Oxford, 1987), pp. 113-152]. A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term to create a "knock out" animal. Progeny harboring the homologously recombined DNA in their germ cells can be identified by standard techniques and used to breed animals in which all cells of the animal contain the homologously recombined DNA. Knockout animals can be characterized for instance, for their ability to defend against certain pathological conditions and for their development of pathological conditions due to absence of the vertebrate fused polypeptide.
As fused has been implicated as a universal mediator for all members of the Hh family (SHh, IHh, DHh), disease states or disorders which are associated with general Hh signaling, would also be treatable with fused and antagonists and agonists thereof. For example, SHh activation fused agonists) has recently been promoted as a treatment for various degenerative disorders of the nervous system, e.g., Parkinson's disease; memory deficits, Alzheimer's disease, Lou Gehrig's disease, Huntington's disease, P1272 schizophrenia, stroke and drug addiction. Recent studies suggest that Dhh mutant males are infertile due to the failure of spermatocytes to complete their differentiation into mature sperm, Bitgood et al., Curr. Biol. 6: 298-304 (1996); Bitgood et al., Dev. Biol. 172: 126-138 (1995). Additionally, fused agonists could be used to great gut diseases, bone diseases, skin diseases, diseases of the testis, ulcers, lung diseases, diseases of the pancreas, diabetes, osteoporosis.
The presence of the protein kinase domain suggests that fused may act similarly as members of the protein kinase family in the modulation of Hh signaling. Protein kinases are essential elements of regulatory circuits in differentiated as well as growing cells; Preat et al., Nature 347: 87-89 (1990). Many of these enzyme are involved in transduction of extracellular signals and operate through a cascade of phosphorylation events that amplify and disseminate the effects of a primary signal. As described earlier, Drosophila fused bears significant homology to other intracellular serine/threonine kinases. Many serine/threonine kinases are implicated in cell-cycle control in yeasts and in mammals, Hunter, Cell 50: 823- 829 (1987); Dunphy Newport, Cell 55: 925-928 (1988); Lee Nurse, Trend. Genet. 4: 287-290 (1988).
Suppression or inhibition of 1Hh signaling is also an objective of therapeutic strategies. Since inactive fused has been shown to inhibit Hh signaling, it follows that a fused antagonist would also be expected to be antagonistic to Hh signaling. Limiting Hh signaling would be useful in disease states or disorders characterized by Hh signaling. For example, SHh is known to be active in Basal Cell Carcinoma; Pfh is known to be active in spermatogenesis. Inhibitor or antagonist of Hh signaling would be effective therapeutics in the treatment of Basal Cell Carcinoma or male contraception, respectively..
The stimulation of Hh signaling is also an objective of therapeutic strategies. Activating Hh signaling would be useful in disease states or disorders characterized by inactive or insufficient Hh signaling.
For example, degenerative disorders of the nervous system, Parkinson's disease, memory deficits, Alzheimer's disease, Lou Gehrig's disease, Huntington's disease, schizophrenia, stroke and drug addiction.
Additionally, fused agonists could be used to great gut diseases, bone diseases, skin diseases, diseases of the testis (including infertility), ulcers, lung diseases, diseases of the pancreas, diabetes, osteoporosis.
F. Anti-vertebrate fused Antibodies The present invention further provides anti- vertebrate fused antibodies. Exemplary antibodies include polyclonal, monoclonal, humanized, bispecific, and heteroconjugate antibodies.
I. Polyclonal Antibodies The anti-vertebrate fused antibodies may comprise polyclonal antibodies. Methods of preparing polyclonal antibodies are known to the skilled artisan. Polyclonal antibodies can be raised in a mammal, for example, by one or more injections of an immunizing agent and, if desired, an adjuvant.
Typically, the immunizing agent and/or adjuvant will be injected in the mammal by multiple subcutaneous or intraperitoneal injections.. The immunizing agent may include the vertebrate fused polypeptide or a fusion protein thereof. It may be useful to conjugate the immunizing agent to a protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include but are not limited to keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. Examples of adjuvants that may be employed include Freund's complete adjuvant and MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate). The immunization protocol may be P1272 selected by one skilled in the art without undue experimentation.
2. Monoclonal Antibodies The anti-vertebrate fused antibodies may, alternatively, be monoclonal antibodies.
Monoclonal antibodies may be prepared using hybridoma methods, such as those described by Kohler and Milstein, Nature, 256:495 (1975). In a hybridoma method, a mouse, hamster, or other appropriate host animal, is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent. Alternatively, the lymphocytes may be immunized in vitro.
The immunizing agent will typically include the vertebrate fused polypeptide or a fusion protein thereof. Generally, either peripheral blood lymphocytes ("PBLs") are used if cells of human origin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired. The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell [Goding, Monoclonal Antibodies: Principles and Practice, Academic Press, (1986) pp. 59-103]. Immortalized cell lines are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine and human origin. Usually, rat or mouse myeloma cell lines are employed. The hybridoma cells may be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells. For example, if the parental cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine ("HAT medium"), which substances prevent the growth of HGPRT-deficient cells.
Preferred immortalized cell lines are those that fuse efficiently, support stable high level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium.
More preferred immortalized cell lines are murine myeloma lines, which can be obtained, for instance, from the Salk Institute Cell Distribution Center, San Diego, California and the American Type Culture Collection, Rockville, Maryland. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies [Kozbor, J. Immunol., 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, Marcel Dekker, Inc., New York, (1987) pp. 51-63].
The culture medium in which the hybridoma cells are cultured can then be assayed for the presence of monoclonal antibodies directed against vertebrate fused. Preferably, the binding specificity of monoclonal antibodies produced by the hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). Such techniques and assays are known in the art. The binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson and Pollard, Anal. Biochem., 107:220 (1980).
After the desired hybridoma cells are identified, the clones may be subcloned by limiting dilution procedures and grown by standard methods [Goding, supra]. Suitable culture media for this purpose include, for example, Dulbecco's Modified Eagle's Medium and RPMI-1640 medium. Alternatively, the hybridoma cells may be grown in vivo as ascites in a mammal.
The monoclonal antibodies secreted by the subclones may be isolated or purified from the culture P1272 medium or ascites fluid by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
The monoclonal antibodies may also be made by recombinant DNA methods, such as those described in U.S. Patent No. 4,816,567. DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells of the invention serve as a preferred source of such DNA. Once isolated, the DNA may be placed into expression vectors, which are then transfected into host cells such as simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. The DNA also may be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences Patent No. 4,816,567; Morrison et al., supra] or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a nonimmunoglobulin polypeptide. Such a non-immunoglobulin polypeptide can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigencombining site of an antibody of the invention to create a chimeric bivalent antibody.
The antibodies may be monovalent antibodies. Methods for prepa-ing monovalent antibodies are well known in the art. For example, one method involves recombinant expression of immunoglobulin light chain and modified heavy chain. The heavy chain is truncated generally at any point in the Fc region so as to prevent heavy chain crosslinking. Alternatively, the relevant cysteine residues are substituted with another amino acid residue or are deleted so as to prevent crosslinking.
In vitro methods are also suitable for preparing monovalent antibodies. Digestion of antibodies to produce fragments thereof, particularly, Fab fragments, can be accomplished using routine techniques known in the art.
3. Humanized Antibodies The anti-vertebrate fused antibodies of the invention may further comprise humanized antibodies or human antibodies. Humanized forms of non-human murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab') 2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity. In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies may also comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and P1272 all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region typically that of a human immunoglobulin [Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol., 2:593-596 (1992)].
Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source that is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import" variable domain. Humanization can be essentially performed following the method of Winter and co-workers [Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)], by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized" antibodies are chimeric antibodies Patent No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
Human antibodies can also be produced using various techniques known in the art, including phage display libraries [Hoogenboom and Winter, J. Mol. Biol., 227:381 (1991); Marks et al., J. Mol. Biol., 222:581 (1991)]. The techniques of Cole et al. and Boerner et al. are also available for the preparation of human monoclonal antibodies (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985) and Boerner et al., J. Immunol., 147(1):86-95 (1991)].
4. Bispecific Antibodies Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens. In the present case, one of the binding specificities is for the vertebrate fused, the other one is for any other antigen, and preferably for a cell-surface protein or receptor or receptor subunit.
Methods for making bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy-chain/lightchain pairs, where the two heavy chains have different specificities [Milstein and Cuello, Nature, 305:537- 539 (1983)]. Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of ten different antibody molecules, of which only one has the correct bispecific structure. The purification of the correct molecule is usually accomplished by affinity chromatography steps. Similar procedures are disclosed in WO 93/08829, published 13 May 1993, and in Traunecker et al., EMBO 10:3655-3659 (1991).
Antibody variable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, C-12, and CH3 regions.
It is preferred to have the first heavy-chain constant region (CHI) containing the site necessary for lightchain binding present in at least one of the fusions. DNAs encoding the immunoglobulin heavy-chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are P1272 co-transfected into a suitable host organism. For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986).
Heteroconiugate Antibodies Heteroconjugate antibodies are also within the scope of the present invention.
Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells Patent No. 4,676,980], and for treatment of HIV infection [WO 91/00360; WO 92/200373; EP 03089]. It is contemplated that the antibodies may be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate and those disclosed, for example, in U.S. Patent No. 4,676,980.
G. Uses for anti-vertebrate fused Antibodies The anti-vertebrate fused antibodies of the invention have various utilities. For example, antivertebrate fused antibodies may be used in diagnostic assays for vertebrate fused, detecting its expression in specific cells, tissues, or serum. Various diagnostic assay techniques known in the art may be used, such as competitive binding assays, direct or indirect sandwich assays and immunoprecipitation assays conducted in either heterogeneous or homogeneous phases [Zola, Monoclonal Antibodies: A Manual of Techniques, CRC Press, Inc. (1987) pp. 147-158]. The antibodies used in the diagnostic assays can be labeled with a detectable moiety. The detectable moiety should be capable of producing, either directly or indirectly, a detectable signal. For example, the detectable moiety may be a radioisotope, such as 3H, 14C, 32P, 35S, or 1251, a fluorescent or chemiluminescent compound, such as fluorescein isothiocyanate, rhodamine, or luciferin, or an enzyme, such as alkaline phosphatase, beta-galactosidase or horseradish peroxidase. Any method known in the art for conjugating the antibody to the detectable moiety may be employed, including those methods described by Hunter et al., Nature, 144:945 (1962); David et al., Biochemistry, 13:1014 (1974); Pain et al., J. Immunol. Meth., '0:219 (1981); and Nygren, J. Histochem. and Cytochem., 30:407 (1982).
Anti-vertebrate fused antibodies also are useful for the affinity purification of vertebrate fused from recombinant cell culture or natural sources. In this process, the antibodies against vertebrate fused are immobilized on a suitable support, such a Sephadex resin or filter paper, using methods well known in the art. The immobilized antibody then is contacted with a sample containing the vertebrate fused to be purified, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the vertebrate fused, which is bound to the immobilized antibody. Finally, the support is washed with another suitable solvent that will release the vertebrate fused from the antibody.
H. Fused Antagonists Several approaches may be suitably employed to create the fused antagonist and agonist compounds of the present invention. Any approach where the antagonist molecule can be targeted to the interior of the cell, which interferes or prevents wild type fused from normal operation is suitable. For example, competitive inhibitors, including mutant fused such as dominant negative mutant identified in the Examples, P1272 which prevent fused from properly binding with other proteins necessary for Hh signaling. Additional properties of such antagonist or agonist molecules are readily determinable by one of ordinary skill, such as size, charge and hydrophobicity suitable for transmembrane transport.
Where mimics or other mammalian homologues of fused are to be identified or evaluated, the cells are exposed to the test compound and compared to positive controls which are exposed only to human fused, and to negative controls which were not exposed to either the compound or the natural ligand. Where antagonists or agonists of fused signal modulation are to be identified or evaluated, the cells are exposed to the compound of the invention in the presence of the natural ligand and compared to controls which are not exposed to the test compound.
Detection assays may by employed as a primary screen to evaluate the phosphatase inhibition/enhancing activity of the antagonist/agonist compounds of the invention. The assays may also be used to assess the relative potency of a compound by testing a range of concentrations, in a range from 100 mM to 1 pM, for example, and computing the concentration at which the amount of phosphorylation or signal transduction is reduced or increased by 50% (IC 5 0 compared to controls.
Assays can be performed to identify compounds that affect phosphorylation of fused substrates.
Specifically, assays can be performed to identify compounds that increase the phosphorylation activity of fused or assays can be performed to identify compounds that decrease the phosphorylation of fused substrates. These assays can be performed either on whole cells themselves or on cell extracts. The assays can be performed in a variety of formats, including protein-protein binding assays, biochemical screening assays, immunoassays, cell based assays, etc. Such assay formats are well known in the art.
The screening assays of the present invention are amenable to high-throughput screening of chemical libraries, and are particularly suitable for identifying small molecule drug candidates.
Antagonist and agonist molecules To screen for antagonists and/or agonists of fused signaling, the assay mixture is incubated under conditions whereby, but for the presence of the candidate pharmacological agent, fused induces hedgehog signaling with a reference activity. The mixture components can be added in any order that provides for the requisite hedgehog activity. Incubation may be performed at any temperature that facilitates optimal binding, typically between about 40 and 40 0 C, more commonly between about 150 and 40 0 C. Incubation periods are likewise selected for optimal binding but also minimized to facilitate rapid, high-throughput screening, and are typically between about 0.1 and 10 hours, preferably less than 5 hours, more preferably less than 2 hours. After incubation, the effect of the candidate pharmacological agent on the fused signaling is determined in any convenient way. For cell-free binding-type assays, a separation step is often used to separate bound and unbound components. Separation may, for example, be effected by precipitation (e.g.
TCA precipitation, immunoprecipitation, etc.), immobilization on a solid substrate), followed by washing. The bound protein is conveniently detected by taking advantage of a detectable label attached to it, e.g. by measuring radioactive emission, optical or electron density, or by indirect detection using, e.g.
antibody conjugates.
For example, a method of screening for suitable fused antagonists and/or agonists could involve the application of agents present in the fused activating Gli reporter assay described in the Examples. Such a screening assay could compare in situ hybridization in the presence and absence of the candidate antagonist and/or agonist in a fused expressing tissue as well as confirmation or absence of fused modulated cellular development. Typically these methods involve exposing an immobilized fused to a molecule suspected of binding thereto and determining binding or phosphorylation of the molecule to the immobilized fused and/or evaluating whether or not the molecule activates (or blocks activation of) fused. In order to identify such fused binding ligands,fused can be expressed on the surface of a cell and used to screen libraries of synthetic candidate compounds or naturally-occurring compounds from endogenous sources such as serum or cells).
Suitable molecules that affect the protein-protein interaction of fused and its binding proteins include fragments of the latter or small molecules, peptidomimetics, which will prevent interaction and proper complex formation. Such small molecules, which are usually less than 10 K molecular weight, are preferable as therapeutics since they are more likely to be permeable to cells, are less susceptible to degradation by various cellular mechanisms, and are not as apt to elicit an immune response as proteins. Small molecules include but are not limited to synthetic organic or inorganic compounds. Many pharmaceutical companies have extensive libraries of such molecules, which can be conveniently screened by using the assays of the present invention.
Non-limiting examples include proteins, peptides, glycoproteins, glycopeptides, glycolipids, polysaccharides, oligosacchardies, nucleic acids, bioorganic molecules, peptidomimetics, pharmacological agents and their metabolites, transcriptional and translation control sequences, and the like.
A preferred technique for identifying molecules which bind to fused utilizes a chimeric substrate epitope-taggedfused or fused immunoadhesin) attached to a solid phase, such as the well of an assay plate. The binding of the candidate molecules, which are optionally labeled radiolabeled), to the immobilized receptor can be measured. Alternatively, competition for activation of Gli can be measured. In screening for antagonists and/or agonists, fused can be exposed to a fused substrate followed by the putative antagonist and/or agonist, or the fused binding protein and antagonist and/or agonist can be added simultaneously, and the ability of the antagonist and/or agonist to block fused activation can be evaluated.
Detection assays The fused polypeptides are useful in assays for identifying lead compounds for therapeutically active agents that modulate fused hedgehog signaling. Specifically, lead compounds that either prevent the formation of fused signaling complexes or prevent or attenuate fused modulated hedgehog signaling binding to fused itself or to a substrate) can be conveniently identified.
Various procedures known in the art may be used for identifying, evaluating or assaying the inhibition of activity of the fused proteins of the invention. As fused is believed to operate in a similar manner as other kinases, techniques known for use with identifying kinase/phosphatase modulators may also be employed with the present invention. In general, such assays involve exposing target cells in culture to the compounds and a) biochemically analyzing cell lysates to assess the level and/or identity of phosphorylation; or scoring phenotypic or functional changes in treated cells as compared to control cells that were not exposed to the test substance. Such screening assays are described in U.S.P. 5,602171, U.S.P. 5,710,173, WO 96/35124 and WO 96/40276.
\\melb_files\home$\suzannet\Keep\Speci\28779-99.1 SPECI.doc 10/05/01 Biochemical detection techniques Biochemical analysis techniques can be evaluated by a variety of techniques. One typical assay mixture which can be used with the present invention contains fused and a protein with which fused is normally associated Gli), usually in an isolated, partially pure or pure form. One or both of these components may be fused to another peptide or polypeptide, which may, for example, provide or enhance protein-protein binding, improve stability under assay conditions, etc. In addition, one of the components usually comprises or is coupled to a detectable label. The label may provide for direct detection by measuring radioactivity, luminescence, optical or electron density, etc., or indirect detection such as an epitope tag, an enzyme, etc. The assay mixture can additionally comprise a candidate pharmacological agent, and optionally a variety of other components, such as salts, buffers, carrier proteins, e.g. albumin, detergents, protease inhibitors, nuclease inhibitors, antimicrobial agents, etc., which facilitate binding, increase stability, reduce non-specific or background interactions, or otherwise improve the efficiency or sensitivity of the assay.
The following detection methods may also be used in a cell-free system wherein cell lysate containing the signal transducing substrate molecule and fused is mixed with a compound of the invention. The substrate is phosphorylated by initiating the kinase reaction by the addition of adenosine triphosphate (ATP). To assess the activity of the compound, the reaction mixture may be analyzed by the SDS-PAGE technique or it may be added to substrate-specific anchoring antibody bound to a solid support, and a detection procedure as described above is performed on the separated or captured substrate to assess the presence or absence of pSer/Thr. The results are compared to those obtained with reaction mixtures to which the compound is not added. The cell-free system does not require the natural ligand or knowledge of its identity. The cell-free system does not require mixtures to which the compound is not added. The cell-free system does not require the natural ligand or knowledge of its identity. For example, Posner et al. 5,155,031 describes the use of insulin receptor as a substrate and rat adipocytes as target cells to demonstrate the ability of pervanadate to inhibit ATP activity. Another example, Burke et al., Biochem. Biophys. Res. Comm. 204: 129-134 (1994) describes the use of autophosphorylated insulin receptor and recombinant PTP1B in assessing the inhibitory activity of a phophotyrosyl mimetic.
Whole cell detection A common technique involves incubating cells with vertebrate fused and radiolabeled phosphate, lysing the cells, separating cellular protein components of the lysate using an SDSpolyacrylamide gel (SDS-PAGE) technique, in either one or two dimensions, and detecting the presence of phosphorylated proteins by exposing X-ray film. Detection can also be effected without using radioactive labeling. In such a technique, the protein components separated by SDS-PAGE) are transferred to a nitrocellulose membrane where the presence. of phosphorylated serine/threonines is detected using an antiphosphoserine/threonine antibody (anti-pS/T).
Alternatively, the anti-pS/T can be conjugated with an enzyme, such as horseradish peroxidase, and detected by subsequent addition of a colorimetric substrate for the enzyme. A further alternative involves 31 \\melb..files\home$\SUZaninet\Keep\speci\28779-99.1 SPECI.doc 10/05/01 P1272 detecting the anti-PS/T by reacting with a second antibody that recognizes the anti-PS/T, this second antibody being labeled with either a radioactive moiety or an enzyme as previously described. Examples of these and similar techniques are described in Hansen et al., Electrophoresis 14: 112-126 (1993); Campbell et al., J. Biol. Chem. 268: 7427-7434 (1993); Donato et al., Cell Growth Diff. 3: 258-268 (1992); Katagiri et al., J. Immunol. 150: 585-593 (1993). Additionally, the anti-pS/T can be detected by labeling it with a radioactive substance, followed by scanning the labeled nitrocellulose to detect radioactivity or exposure of X-ray film.
(ii) Kinase assays When the screening methods of the present invention for fused antagonists/agonists are carried out as an ex vivo assay, the target kinase fused) can be a substantially purified polypeptide. The kinase substrate MBP, Gli) is a substantially purified substrate, which in the assay is phosphorylated in a reaction with a substantially purified phosphate source that is catalyzed by the kinase. The extent of phosphorylation is determined by measuring the amount of substrate phosphorylated in the reaction. A variety of possible substrates may be used, including the kinase itself in which instance the phosphorylation reaction measured in the assay is autophosphorylation. Exogenous substrates may also be used, including standard protein substrates such as myelin basic protein (MBP); yeast protein substrates; synthetic peptide substrates, and polymer substrates. Of these, MBP and other standard protein substrates may be regarded as preferred (see Example 10). Other substrates may be identified, however, which are superior by way of affinity for the kinase, minimal perturbation of reaction kinetics, possession of single or homogenous reaction sites, ease of handling and post-reaction recover, potential for strong signal generation, and resistance or inertness to test compounds.
Measurement of the amount of substrate phosphorylated in the ex vivo assay of the invention may be carried out by means of immunoassay, radioassay or other well-known methods. In an immunoassay measurement, an antibody (such as a goat or mouse anti-phosphoserine/threonine antibody) may be used which is specific for phosphorylated moieties formed during the reaction. Using well-known ELISA techniques, the phosphoserine/threonine antibody complex would itself be detected by a further antibody linked to a label capable of developing a measurable signal (as for example a fluorescent or radioactive label). Additionally, ELISA-type assays in microtitre plates may be used to test purified substrates. Peraldi et al., J. Biochem. 285: 71-78 (1992); Schraag et al., Anal. Biochem. 211: 233-239 (1993); Cleavland, Anal.
Biochem. 190: 249-253 (1990); Farley, Anal. Biochem. 203: 151-157 (1992) and Lozaro, Anal. Biochem.
192: 257-261 (1991).
For example, detection schemes can measure substrate depletion during the kinase reaction.
Initially, the phosphate source may be radiolabeled with an isotope such as P or P, and the amount of substrate phosphorylation may be measured by determining the amount of radiolabel incorporated into the substrate during the reaction. Detection may be accomplished by: commercially available scintillantcontaining plates and beads using a beta-counter, after adsorption to a filter or a microtitre well surface, or photometric means after binding to a scintillation proximity assay bead or scintillant plate. Weemink and Kijken, J. Biochem. Biophs. Methods 1: 49, 1996; Braunwalder et al., Anal. Biochem. 234: 23 (1996); Kentrup et al., J. Biol. Chem. 271: 3488 (1996) and Rusken et al., Meth. Enzymol. 200: 98 (1991).
Preferably, the substrate is attached to a solid support surface by means of non-specific or, preferably, specific binding. Such attachment permits separation of the phosphorylated substrate from unincorporated, labeled phosphate source (such as adenosine triphosphate prior to signal detection. In one embodiment, the substrate may be physically immobilized prior to reaction, as through the use of NuncTM high protein binding plate (Hanke et al., J. Biol. Chem. 221: 695 (1996)) or Wallac ScintiStrip T M plates (Braunwalder et al., Anal.
Biochem. 234: 23 (1996). Substrate may also be immobilized after reaction by capture on, for example, P81 phophocellulose (for basic peptides), PEI/acidic molybdate resin or DEAE, or TCA precipitation onto Whatman T m 3MM paper, Tiganis et al., Arch. Biochem. Biophys. 325: 289 (1996); Morawetz et al., Mol. Gen.
Genet. 250; 17 (1996); Budde et al, Int J. Pharmacognosy 33: 27 (1995) and Casnellie, Meth. Enz. 200: 115 (1991). Yet another possibility is the attachment of the substrate to the support surface, as by conjugation with binding partners such as glutathione and streptavidin (in the case of GST and biotin), respectively) which have been attached to the support, or via antibodies specific for the tags which are likewise attached to the support.
Further detection methods may be developed which are preferred to those described above. Especially for use in connection with high-throughput screening, it is expected that such methods would exhibit good sensitivity and specificity, extended linear range, low background signal, minimal fluctuation, compatibility with other reagents, and compatibility with automated handling systems.
The in vivo efficacy of the treatment of the present invention can be studied against chemically induced tumors in various rodent models. Tumor cell lines propagated in in vitro cell cultures can be introduced in experimental rodents, e.g. mice by injection, for example by the subcutaneous route. Techniques for chemical inducement of tumors in experimental animals are well known in the art.
Biological detection techniques: The ability of the antagonist/agonist compounds of the invention to modulate the activity fused, which itself modulates hedgehog signaling, may also be measured by scoring for morphological or functional changes associated with ligand binding. Any qualitative or quantitative technique known in the art may be applied for observing and measuring cellular processes which comes under the control of fused. The activity of the compounds of the invention can also be assessed in animals using experimental models of disorders caused by or related to dysfunctional hedgehog signaling. For example, ineffective DHh hedgehog signaling in mice leads to viable but sterile mice. The effects of mutant fused (hfused-DN) also affects gut development, which is regulated by IHh expression. Additionally, proper SHh signaling is critical to murine embryonic development at the notochord and floor plate, neural tube, distal limb structures, spinal column and ribs. Improper SHh signaling, is also correlative with cyclopia. Any of these phenotypic properties could be evaluated and quantified in a screening assay for fused antagonists and/or agonist. Disease states associated with overexpression of hedgehog is associated with basal cell carcinoma while inactive sonic hedgehog signaling leads to improper neural development.
The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosages for use in humans. The dosage of the compounds of the invention should lie within a range 33 14 1--l 1 1-1 PI272 of circulating concentrations with little or no toxicity. The dosage may vary within this range depending on the dosage form employed and the route of administration.
Antisense nucleotides Another preferred class of antagonists involves the use of gene therapy techniques, include the administration of antisense nucleotides. Applicable gene therapy techniques include single or multiple administrations of therapeutically effective DNA or mRNA. Antisense RNAs and DNAs can be used as therapeutic agents for blocking the expression of certain genes in vivo. Short antisense oligonucleotides can be imported into cells where they act as inhibitors, despite their low intracellular concentrations caused by restricted uptake by the cell membrane, Zamecnik et al., Proc. Nail. Acad. Sci. USA 83: 4143-4146 (1986).
The oligonucleotides can be modified to enhance their uptake, by substituting their negatively charged phosphodiester groups by uncharged groups.
There are a variety of techniques known for introducing nucleic acids into viable cells. The techniques vary depending upon whether the nucleic acid is transferred into cultured cells in vitro, ex vivo, or in vivo in the cells of the intended host. Techniques suitable for the transfer of nucleic acid into mammalian cells in vitro include the use of liposomes, electroporation, microinjection, cell fusion, DEAE-dextran, the calcium phosphate precipitation method, etc. The currently preferred in vivo gene transfer techniques include transfection with viral (typically retroviral) vectors and viral coat protein-liposome mediated transfection, Dzau et Trends Biotech. 205-210 (1993). In some situations it is desirable to provide the nucleic acid source with an agent that targets the target cells, such as an antibody specific for a cell surface membrane protein associated with endocytosis may be used for targeting and/or to facilitate uptake, e.g., capsid proteins or fragments thereof tropic for a particular cell type, antibodies for proteins which undergo internalization in cycling, and proteins that target intracellular localization and enhance intracellular half-life.
The technique of receptor-mediated endocytosis is described, for example, by Wu et al., 1. Biol. Chem. 262: 4429-4432 (1987); Wagner et al., Proc. Natl. Acad. Sci. USA 87: 3410-3414 (1990). For a review of known gene marking and gene therapy protocols, see Anderson et al., Science 256: 808-813 (1992).
In one embodiment, fused antagonist and/or agonist molecules may be used to bind endogenous ligand in the cell, thereby causing the cell to be unresponsive to fused wild type, especially when the levels of fused in the cell exceed normal physiological levels. Also, it may be beneficial to bind endogenous fused substrates or complexing agents that are activating undesired cellular responses (such as proliferation of tumor cells).
In a further embodiment of the invention, fused expression may be reduced by providing fusedexpressing cells with an amount of fused antisense RNA or DNA effective to reduce expression of the fused protein.
1. Diagnostic Uses Another use of the compounds of the invention human and vertebrate fused, vertebrate fused variant and anti-vertebrate fused antibodies) described herein is to help diagnose whether a disorder is driven, to some extent, fused or hedgehog signaling. For example, basal cell carcinoma cells are associated with active hedgehog signaling.
A diagnostic assay to determine whether a particular disorder is driven by hedgehog signaling, can be carried out using the following steps: culturing test cells or tissues; administering a compound which can inhibit fused modulated hedgehog signaling; and measuring the degree of kinase attenuation on the fused substrate in cell lysates or hedgehog mediated phenotypic effects in the test cells. The steps can be carried out using standard techniques in light of the present disclosure. For example, standard techniques can be used to isolate cells or tissues and culturing or in vivo.
Compounds of varying degree of selectivity are useful for diagnosing the role of fused. For example, compounds which inhibit fused in addition to another form of kinase can be used as an initial test compound to determine if one of several serine/threonine kinases drive the disorder. The selective compounds can then be used to further eliminate the possible role of the other serine/threonine kinases in driving the disorder. Test compounds should be more potent in inhibiting serine/threonine kinase activity than in exerting a cytotoxic effect an IC 5 0
/LD
50 of greater than one). The IC 50 and LD5 0 can be measured by standard techniques, such as an MTT assay, or by measuring the amount of LDH released. The degree of IC 50
/LD
5 0 of a compound should be taken into account in evaluating the diagnostic assay. Generally, the larger the ratio the more relative the information. Appropriate controls take into account the possible cytotoxic effect of a compound, such as treating cells not associated with a cell proliferative disorder control cells) with a test compound, can also be used as part of the diagnostic assay. The diagnostic methods of the invention involve the screening for agents that modulate the effects of fused upon hedgehog signaling. Exemplary detection techniques include radioactive labeling and immunoprecipitating 5,385,915).
The following examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way.
All patent and literature references cited in the present specification are hereby incorporated by reference in their entirety.
EXAMPLES
Commercially available reagents referred to in the examples were. used according to manufacturer's instructions unless otherwise indicated. The source of those cells identified in the following examples, and throughout the specification, by ATCC accession numbers is the American Type Culture Collection, Rockville, Maryland.
EXAMPLE 1 Isolation of human fused cDNA clones An expressed sequence tag (EST) DNA database (LIFESEQM, Incyte Pharmaceuticals, Palo Alto, CA) was searched for a human homologue of the Drosophila segment polarity gene fused (SEQ ID NO 26) (Preat et al., Nature 347: 87-9 (1990)). The EST Incyte #2515662 (Fig. 2) (SEQ ID NO. 3) was identified as a potential candidate. In order to identify human cDNA libraries containing human fused clones, human cDNA libraries in pRK5 were first screened by PCR using the following primers: h-FUSED.f (SEQ ID NO. 8) 5'-CAATACAATGGTGCTGACATCCATCAAAGGCA-3' h-FUSED.r (SEQ ID NO. 9) 5'-'GAAGGGAGGGGTGCCTACTGCCA-3' A fetal lung library was selected and enriched for fused cDNA clones by extension of single stranded DNA from plasmid libraries grown in dug'/bung' host using the h-FUSED.f primer (SEQ ID NO. 8) in a reaction containing \\melbfiles\home$\suzannet\Keep\Speci\28779-99.1 SPECI.doc 10/05/01 of 10x PCR Buffer (Perkin Elmer), 14tl dNTP (20 mM), 1 p.1 library DNA (200 ng), 0.5 ml primer, 86.5 p1
H
2 0 and 1 pl of Amplitaq® (Perkin Elmer) added after a hot start. The reaction was denatured for 1 min. at 0 C, annealed for 1 min. at 60 0 C then extended for 20 min. at 72 0 C. DNA was extracted with phenol/CHCI3, ethanol precipitated, then transformed by electroporation into DH10B host bacteria. Colonies from each transformation were plated and lifted on nylon membranes and screened with an oligo probe derived from the EST sequence of the following sequence: h-FUSED.p (SEQ ID NO. 10) 5'-CTCCAGCTCTGGAGACATATAGAGTGGTGTGCCTTTGA-3' The oligo probe was labeled with 32P]-ATP and T4 polynucleotide kinase. Filters were hybridized overnight at 42*C in 50% formamide, 5xSSC, 10xDenhardt's, 0.05M sodium phosphate (pH 0.1% sodium pyrophosphate, 50 gpg/ml of sonicated salmon sperm DNA. The filters were then rinsed in 2x SSC and washed in 0.1x SSC, 0.1% SDS then exposed to Kodak® X Ray films. Two positive clones (DNA28494 (SEQ ID NO.
6) and DNA28495 (SEQ ID NO. 4) Figs. 4 5) containing an insert of approximately 5 kb were isolated and sequenced. The sequence of clone DNA28495 (SEQ ID NO..4) contains a potential initiation methionine at position 116 followed by an open reading frame of 1944 bp (Fig. However, this open reading frame (ORF) encodes a protein that is only 648 amino acids long, somewhat shorter than the 795 amino acid sequence of the Drosophila fused. Interestingly, a second open reading frame is present in the 3' region of the cDNA, from nucleotide 2295 to 4349 (Fig. which suggests that the cDNA may have been improperly spliced and that an intron remains between the 2 ORFs, or correspond to an alternatively spliced variant of fused. The sequence of clone DNA28494 (SEQ ID NO. 6) is very similar. There is one nucleotide difference between clone DNA28495 (SEQ ID NO. 4) and clone DNA28494 (SEQ ID NO. 6) located in the first ORF at position 1863 of clone 28495 (SEQ ID NO. 4) (A vs. G) which changes the coding sequence from an Gin to a Arg at position 583. (Fig. This change is likely due to an allelic variation. The first open reading frame of DNA28494 (SEQ ID NO. 6) starts at residue 115 and is followed by a 647 amino acid long open reading frame. The sequences are identical except for the one change described above at position 583 and for the last 9 residues in the first open reading frame.
EXAMPLE 2 Expression of fused clones In order to determine the size of the protein expressed from the cDNA corresponding to DNA28495 (SEQ ID NO. 4) and DNA28494 (SEQ ID NO. an HA epitope tag was inserted at the Nterminus of the protein by PCR using the following primers: Hfus.Cla-HA.F: (SEQ ID NO. 11) 5'-CCATCGATGTACCCATACGACGTCCCAGACTACGCTGAAAAGTACCACGTGTTGGAGATG-3' and hFus.Xba.R: (SEQ ID NO. 12) 5'-GCTCTAGACTAAGGGGCAGGTCCTGTGTTCTG-3'.
The PCR product was purified, digested with ClaI-Smal and subcloned into the pRK5 plasmids containing DNA28494 (SEQ ID NO. 6) and DNA28495 (SEQ ID NO. DNA from each of the constructs was transfected overnight into 293 cells using the CaPO 4 method (Sambrook et al, supra; Ausuble et al., supra).
After about 24 h. to 48 h. after transfection, the cells were harvested and the cell pellet was lysed in 1 ml of lysine 36 \\melbfilee\home$\suzannet\Keep\Speci\28779-99.1 SPECI.doc 10/05/01 buffer (50 mM Tris pH 8.0, 150 mM NaCI, 1 mM EDTA, 1% NP40, Aprotinin, Leupeptin, -PMSF, 1 mM NaF and 1 mM Sodium Vanadate) for 20 min at 4°C. The extract was spun for 10 min at 10K then the supernatant was transferred to a new tube and precleared with 20 p.1 Protein A sepharose for 1 h. The protein A sepharose was spun down and 1 p.l of anti-HA antibody (5 g.g, Boehringer) was added to each tube. After overnight incubation at 4°C, 30 p.1 of Protein G sepharose was added and the tubes incubated at 4°C for 1 hour. The protein G beads were then sun down for 1 min., washed 3 times with lysis buffer, resuspended in 20 p.1 of laemli buffer in the presence of P-mercapto ethanol. Samples were denatured for 5 min. at 100°C then loaded on a 6% polyacrylamide gel. Proteins were then transferred to nitrocellulose and analyzed by Western blot using the same anti-HA antibody overnight at 1 g.g/ml in blocking buffer (PBS, 0.5% Tween 5% non fat dry milk, 3% goat serum followed by an anti-mouse HRP. ECL was used for the detection and the membrane was exposed for seconds to X-Ray films. A specific band of 150 kDa was detected in the cell pellet of cells transfected with the construct with construct corresponding to clone DNA28494 (SEQ ID NO. 6) and a specific band of approximately 100 kDa could be detected for clone DNA28495 (SEQ ID NO. 4) (Fig. These bands were not present in the mock transfected control. The presence of the 150 kDa band suggests the two open reading frames of DNA28494 (SEQ ID NO. 6) can be spliced together to direct the synthesis of a large protein of 150 kDa. The absence of this band for DNA28495 (SEQ ID NO. 4) suggested that this clone apparently cannot be correctly spliced. Alternative splicing of the fused gene seems to lead to the production of several different products and may be a mechanism or regulation of fused activity. Specific regions at the C-terminus of the Drosophila fused protein is known to be required for the activity of the molecule, Therond et al., Genetics 142: 1181-1198 (1996); Robbins et al., Cell 90: 225-234 (1997). Shorter fused molecules truncated at the Cterminus may therefore correspond to inactive or to dominant negative forms of the molecule.
EXAMPLE 3 Northern Blots In order to determine the best tissue source to isolate more fused cDNAs and to identify a transcript encoding a full length 150 kDa fused molecule, human multiple tissue northern blots I, II and fetal blot from Clontech were probed with a 1.6 kb, Clal-AccI fragment derived from clone DNA28494 (SEQ ID NO. 6) labeled by random priming. The blots were hybridized in 50% formamide, 5x SSC, lOx Denhardt's, 0.05M Sodium phosphate (pH 0.1% Sodium pyrophosphate, 50 mg/ml sonicated salmon sperm DNA, all in the presence of xl10 6 cpm/ml 32 P-labeled probe at 42°C overnight. The blots were washed in 2x SSC at RT for minutes and washed in 0.2x SSC/0.1 SDS at 42°C for 30 minutes then exposed to x-ray film overnight. Fig. 7 shows that the fused message is expressed at high levels in testis and at low levels in most other tissues, including fetal tissues. (Fig. 7).
EXAMPLE 4 PCR on different tissues to identify the correct splice form In order to isolate a cDNA where the 2 potential ORFs were spliced together correctly, we designed the following primers flanking the potential intron and amplified various tissues including human fetal brain, brain, keratinocyte, testis, ovary, fetal liver, and lung templates.
F1 (SEQ ID NO. 13) 5'-CTGACGACACAGCAGGTTGTC-3' R4 (SEQ ID NO. 14) 5'-CAGATGCTTCAGGATGGACAT-3' 37 \\meJlbfiles\home$\suzannet\Keep\speci\28779-99.1 spECi.doc 10/0S/01 Two microliters of each cDNA library was used as the template and PCR was done with Klentaq® polymerase. PCR was performed for 45 cycles of amplification with 94 0 C denaturation for 1 min., annealing for 1 min., and 68 0 C extensions for 2 min. One fifth of the reaction was loaded on 1% agarose gel and was Southern blotted. The blot was hybridized overnight with full-length fused probe labeled by random priming as described for the Northern blot.
A 1 kb PCR fragment was identified in fetal brain, testis and ovary. This fragment was gel-purified and subjected to direct PCR sequencing using both the F1 and R4 primers (SEQ ID NOS. 13 and 14) identified above as well as the following primers: hfl6 (SEQ ID NO. 15) 5'-AGAGTAGCAACGTCACTGC-3' hf8 (SEQ ID NO. 16) 5'-CCTCACTGACAAGGCAGCAGG-3' hfl9 (SEQ ID NO. 17) 5'-CCCGAGGAGGCATCTGCACAG-3' The sequence of this I kb fragment revealed that intron sequences were absent and that the 2 ORFs were connected together in the same reading frame. The sequence of the correctly spliced sequence is shown in Fig. 1 (SEQ ID NO. The initiator ATG is present at position 161 and is followed by an ORF of 3945 nucleotides which encodes a 1315 amino acid long protein with a predicted molecular weight of 144 kDa.
The overall similarity with Drosophila fused (SEQ ID NO. 23) is 28% (Fig. The N-terminal 263 amino acid domain of the protein containing the kinase domain is 55% homologous to the Drosophila fused kinase domain. The remaining 1052 amino acids portion of the protein is not appreciably homologous to other known proteins and, interestingly, is not homologous to the corresponding region in Drosophila fused.
Interestingly, this region of non-homology includes the very C-terminus of the fly protein which appears to be required for activity, Robbins et al., Cell 9Q: 225-34 (1997); Therond et al., Genetics 142: 1181-98 (1996). The improperly spliced cDNAs described above may reflect alternative splicing of the fused gene which leads to the production of a molecule with a truncated C-terminus and may be a mechanism to regulate fused activity.
EXAMPLE Reconstitution of the correctly spliced full length human fused The fused clone DNA28495 (SEQ ID NO. 4) was subcloned from the pRK5B plasmid into using ClaI-HindII. PCR was performed using human testis cDNA as a template and the primers hf3 (SEQ ID NO. 18) (CAGAACTTCAGGTCCTAAAGG) and R4 (sequence see above, Example PCR conditions were 45 cycles of (94°C,1 min, 46 0 C to 68 0 C temperature gradient annealing for 1 min, and 68°C, 4 min). The PCR fragment was digested with AccI and ligated in the pRK5.tkneo.fused plasmid cut with AccI in order to replace the region containing the intron with the correct spliced form. Two subclones were sequenced between the two AccI site and had the same correct sequence.
EXAMPLE 6 In situ hybridization El 11.3 and E13.5 mouse embryos were immersion-fixed overnight at 4°C in 4% paraformaldehyde, cryoprotected overnight in 15% sucrose, embedded in O.T.C. and frozen on liquid nitrogen. Adult mouse brains were fresh frozen with powdered dry ice. P1 mouse brains, adult mouse testis and adult rat spinal cords were embedded in O.T.C. and frozen on liquid nitrogen. Sections were cut at 16 mm, and processed for in situ hybridization for fused by the method of Phillips et Science 250: 290-294 (1990). RNA probes were labeled 38 \\melb..files\home$\suzannet\Keep\speci\28779-99.1 SPECI.doe 10/05/01 with 33 P-UTP as described by Melton et al., Nucleic Acids Res. 12: 7035-7052 (1984). Sense and antisense probes were synthesized from a mouse fused DNA fragment using T3 and T7, respectively, corresponding to the region encoding amino acid residues 317-486 of the human sequence.
Figure 8 reveals that the mouse fused mRNA is widely distributed in SHh responsive tissues, including the neural tube, pre-somitic mesoderm, somites, developing limb buds and skin. Transcripts for fused were also found in the embryonic gut, testis, cartilage and muscle Tissues that are exposed to the other members of the Hh protein family, Desert and Indian. In the E11-5 mouse nervous system, high levels of fused transcripts were detected throughout the forebrain, midbrain, hindbrain and spinal cord. These high levels of expression were retained in embryonic day 13.5. In both embryonic days 11.5 and 13.5, fused mRNA was detected mainly in the ventral aspect of the neural tube, in regions that are likely to be exposed to the ventral midline-derived SHh. By post natal day widespread expression of fused is still maintained throughout the brain with high levels of transcripts detected in the cortex, hypocampus, ependima and choroid plexus. In the adult, low levels of fused expression are detected all through the brain with higher levels confined to the ependima.
The tissue distribution of fused and the Hh receptor components, Smo and Ptch show considerable overlap. All of them are initially expressed through the neural tube as well as in other Hh responsive tissues.
However, whereas Smo mRNA was evenly distributed along the dorso-ventral axis, Ptch and fused mRNAs are found at higher levels ventrally, suggesting that they may be upregulated by Hh. In addition while by day E12, expression of both Smo and Ptch is found mainly in cells which are in close proximity to the ventricular zone, fused mRNA is still widely expressed and its levels decline only later. In the adult expression of both Smo and fused is confined to the ependima where neurogenesis continues.
Detailed analysis of fused expression in adult testis was also performed by in situ hybridization (Fig. 9).
fused was found to be expressed at very high levels on stages I and II germ cells in the seminiferous tubules.
Levels of fused vary in different seminiferous tubules, suggesting that its expression is regulated according to the germinal cell state of differentiation.
EXAMPLE 7 Gli Luciferase Assay Given the low homology between dfused and hfused, it was prudent to determine whether in fact the isolated hfused is indeed a mediator of Hh signaling. The following assay was developed to measure the activation of the transcription factor GLI, the mammalian homologue of the Drosophila cubitus interruptus (Ci).
It has been shown that GLI is a transcription factor activated upon SHh stimulation of cells.
Nine copies of a GLI binding site present in the HNF3 enhancer, (Sasaki et al., Development 124: 1313-1322 (1997)), were introduced in front of a thymidine kinase minimal promoter driving the luciferase reporter gene in the pGL3 plasmid (Promega). The sequence of the GLI binding sequence was: TCGACAAGCAGGGAACACCCAAGTAGAAGCTC (p9XGliLuc) (SEQ ID NO. 19), while the negative control sequence was: TCGACAAGCAGG AAGTGGGAAGTAGAAGCTC (p9XmGliLuc) (SEQ ID NO. These constructs were cotransfected with the full length fused construct or with a plasmid encoding sonic hedgehog in C3H10T1/2 cells grown in F12, DMEM (50:50), 10% FCS heat inactivated. The day before transfection 1 x 105 cells per well was inoculated in 6 well plates, in 2 ml of media. The following day, 1 ltg of each construct was cotransfected in duplicate with 0.025 mg ptkRenilla luciferase plasmid using lipofectamine 39 \\melb_files\home$\suzannet\Keep\Speci\28779-99.1 SPECI.doc 10/05/01 (Gibco-BRL) in 100 .l OptiMem (with GlutaMAX) as per manufacturer's instructions for 3 hours at 37*C.
Serum 1 ml) was then added to each well and the cells were incubated for 3 more hours at 37 0 C. Cells were then washed twice with PBS, then incubated for 48 hours at 37°C in 2 ml of media. Each well was then washed with PBS, and the cells lysed in 0.5 ml Passive Lysis Buffer (Promega) for 15 min. at room temperature on a shaker. The lysate was transferred in eppendorf tubes on ice, spun in a refrigerated centrifuge for seconds and the supernatant saved on ice. For each measure, 20 pl of cell lysate was added to 100 pl of LARH (luciferase assay reagent, Promega) in a polypropylene tube and the luciferase light activity measured. The reaction was stopped by the addition of Stop and Glow buffer (Promega), mixed by pipetting up and down 3 to times and Renilla luciferase lights activity was measured on the luminometer.
As shown in Figure 6, fused can induce GLI activity (9.5 fold) in a similar manner as SHh (5.5 fold) This result suggests that the fused gene isolated is a mediator of SHh signaling. An irrelevant serine-threonine kinase, Akt, was not active in this assay (data not shown). The fused activity is dependent on an intact kinase domain as molecules with deletion of this region (fused C-term) (SEQ ID NO. 27) or mutation of a conserved lysine residue at about amino acid position 33 in the ATP binding site (fused-DN (SEQ ID NO. 25)) were not able to activate GLI. Similarly, the C-terminal tail of the protein is necessary for this activity since the kinase domain alone was not active in this assay (fused KD) (SEQ ID NO. 24). Expression of each protein was verified by Western blot using an HA tag inserted at the N-terminus of the molecule (data not shown). These results substantiate the conclusion that the homologue of the dfused isolated by Applicants is indeed hfused.
Furthermore, these results indicate that fused is capable of and sufficient for the activation of Gli, the major target of SHh signaling and is thus likely to be a direct mediator of the SHh signal in vertebrates.
EXAMPLE 8 Induced cyclopia in frog embryos Introduction: In order to demonstrate that the human fused gene is not only capable of but also required to transduce the SHh signal in vertebrates, a mutant version of fused known as fused-DN (dominant negative) having a mutation of the lysine at position 33 in the ATP binding site was created (SEQ ID NO. 25). This residue is \\uielb-.file\home\suzanrnet\Keep\Speci\28779-99.1 SPECX.doc 10/05/01 P1272 conserved among all kinases and is necessary for kinase activity (Hanks et al., Methods Enzymol. 200: 38-62 (1991) and its conversion to any other residue in most cases results in the creation of dominant negative mutants.
Methods: Plasmid Construction: Wild type fused cDNA (SEQ ID NO. 1) with an HA tag inserted at the carboxy terminus was subcloned into pRK5 and a dominant negative form was generated by conversion of lysine at positive 33 to an arginine. Supercoiled plasmid DNA was prepared by Qiagen and used for injection into Xenopus laevis embryo.
Manipulation of Xenopus embryos: Adult female frogs were boosted with 200 I.U. pregnant mare serum 3 days before use and with 800 I.U. of human chorionic gonadotropin the night before injection. Fresh oocytes were squeezed out from female frogs the next morning and in vitro fertilization of oocytes was performed by mixing oocytes with minced testis from sacrificed male frogs. Developing embryos were maintained and staged according to Nieuwkoop and Faber, Normal Table of Xenopus laevis, P. Co., ed. (Amsterdam, 1967).
Fertilized eggs were dejellied with 2% cysteine (pH 7.8) for 10 minutes, washed once with distilled water and transferred to 0.1 x MBS with 5% Ficoll. Fertilized eggs were lined on injection trays in 0.1 x MBS with 5% Ficoll. Two-cell stage developing Xenopus embryos were injected with 200 pg of either containing wild type fused (WT (SEQ ID NO. or dominant negativefused (DN (SEQ ID NO. Injected embryos were kept on trays for another 6 hours, after which they were transferred to 0.1 x MBS with 50 mg/ml gentamycin for 3 days until reaching Nieukwkoop stage 35 when eye development is complete.
Results: To test whether human fused gene acts as a signal transducer of Hedgehog signaling, we injected wild type or dominant negative form of human fused in developing frog embryos. Embryos injected with 120 pg of DNA divided normally in blastula stage and gastrulate normally. While eye development was normal in wild type,fused (SEQ ID NO. 2) injected and mock injected embryos, about 30% (Table 1) of the embryos that were injected with fused-DN showed fused eye structure or two eyes connected by some pigmented retina tissue (Fig. 11 IA). In Table 1, 200 pg of plasmid DNA was delivered to the animal pole of 2-cell stage embryos. Each sample represents the results of at least 3 independent experiments. Embryos were scored visually for cyclopia defects.
TABLE 1 Fusion-DN Induced Cyclopia in Xenopus Embryos Injected DNA Normal Cyclop n Hu-fused (SEQ ID NO. 2) 45 0 kinase domain (SEQ ID NO. 24) 43 0 43 C-terminus (SEQ ID NO. 27) 53 1 54 fused DN (SEQ ID NO. 25) 32 15 47 uninjected 61 0 61 The observed cyclopia phenotype is strikingly similar to the one of mouse embryos deficient in SHh (Chiang et al., Nature 383: 407-13 (1996) and of zebrafish embryos where SHh signaling has been blocked by overexpression of a constitutive active PKA, Hammerschmidt et al., Genes Dev. D10: 647-58 (1996); Ungar and Moon, Dev. Biol. 178: 186-91 (1996). In addition, both brain (forebrain) and gut development appeared normal at later stages of tadpole development in the fused-DN (SEQ ID NO. 25) injected embryos (Fig. 11B). In contrast, embryos overexpressing either wild type fused (SEQ ID NO. 2) or N or C-terminal terminal truncation mutants (SEQ ID NOS. 27 24, respectively) did not present any abnormalities.
During normal development of the Xenopus eye, the eye primordium starts as a single field expressing transcription factor Pax-6, which is a vertebrate homologue of Drosophila eyeless, Li et al., Development, 124: 603-15 (1997). At the neurula stage, this eye field is separated into two eye primordia due to an inhibiting signal from prechordal mesoderm. It has been further demonstrated that SHh is the prechordal mesoderm derived signal that is responsible for the inhibition of Pax-6 expression in the midline of the eyefield.
To further understand how overexpression of fused-DN (SEQ ID NO: 25) induced a fused eye in Xenopus embryos, whole mount in situ hybridization was performed in order to determine the expression pattern of Pax-6 in injected embryos. As shown in Figure 11C, Pax-6 expression in embryos injected with fused-DN (SEQ ID NO: 25) remains as a single field (Fig. 11D). Thus fused-DN (SEQ ID NO: 25) induces a cyclopia phenotype by most likely preventing SHh from inhibiting Pax-6 expression in the midline of the eyefield.
EXAMPLE 9 Rescue of fused-DN (SEQ ID NO. 25) Injected Xenopus Embryos by Gli SHh expression in early floor plate cells is induced by SHh produced by the notochord. To test whether SHh expression in the floor plate will also be inhibited when SHh signaling is blocked, early neurula stage embryos injected with fused-DN or wild-type constructs were stained for SHh expression (See Example 8 for procedure). SHh expression in floor plate cells or early neurula stage embryos was completely suppressed in 26 out of 28 embryos injected when the mutated fused is overexpressed (Table 2, Figure 11 C, left embryo), while the expression of SHh was unaffected in control embryos (Fig. 6E, right embryo). Table 2 represents scored data from three independent experiments. 100 pg of fused-DN, 100 pg of fused-wt or 50 pg of Gli-1 plasmid were injected in 2-cell stage embryos. Embryos were harvested at early neurula stage for SHh staining.
TABLE I Wild type fused and Gli rescue SHh expression in floor plate when coexpressed with fused-DN SHh staining percentage fused-DN 2/28 7% fused-DN +fused WT 20/24 83% fused-DN Gli 36/36 100 To confirm that this phenotype was due to specific inhibition of the SHh signaling pathway in the floor plate, we attempted to rescue the phenotype by coinjection of wtfused RNA with fused-DN RNA in a 1:1 ratio.
Table 2 shows that more than 80% of the embryos coinjected with wtfused andfused-DN RNAs show normal SHh staining in the floor plate. This demonstrates that SHh expression in fused-DN injected embryos is 42 specifically blocked by inhibition of endogenous fused activity.
To further demonstrate that the observed phenotype of fused-DN are due to disruption of the SHh signal cascade and to confirm that hfused works upstream of Gli in this pathway, we asked whether the overexpression of Gli can also rescue the phenotype of Xenopus embryos injected with fused-DN. As shown in Table 2, the rescue of SiHh expression in the floor plate of fused-DN injected embryos is complete when Gli is overexpressed.
Taken together, these findings are consistent with Applicants hypothesis that vertebrate fused functions in the SHh pathway and that is a necessary mediator in the SHh signal transduction pathway, which acts upstream of Gli.
EXAMPLE Immunoprecipitations and In Vitro Kinase Assay To directly determine whether hfused has kinase activity, fused (SEQ ID NO. fused-DN (SEQ ID NO. 25) and fused-kd (SEQ ID NO. 24) cDNAs were tagged with the influenza HA epitope tag and transiently transfected into 293 cells. Immunoprecipitates were tested for kinase activity in the presence of myelin basic protein (MBP) and 32P]-ATP. The amount of 32 P incorporated into MBP was determined after SDS-PAGE and found to be was about 3 times higher than in fused-KD (SEQ ID NO. 24) and 2 times higher in wt fused (SEQ ID NO. 2) containing extracts compared to controls, while mutation of Lys33 to Arg (fused-DN (SEQ ID NO. 25)) neutralizes the activity (Fig. 12).
For immunoprecipitation experiments human embryonic kidney 293 cells were transiently transfected with the various expression plasmids. After 24 hours, the transfected cells were collected and.lysed for 20 min.
at 4°C in 1 ml of lysis buffer (50 mM Tris, pH 150 mM NaCI, 1 mM EDTA, 1 mM sodium fluoride, 1 mM sodium orthovanadate, 1 mM PMSF and protease inhibitors (Complete, Boehringer Mannheim) containing 1% 0.5% deoxycholic acid. Cell debris was removed by centrifugation for 10 min. at 10,000 rpm and the sodium chloride concentration of the cell lysates was increased to 250 mM. The supernatant was precleared for 1 hour with 20 .tl Protein A Sepharose (Pharmacia). Lysates were immunoprecipitated using anti-HA antibodies followed by Protein A Sepharose. The beads were washed twice with lysis buffer containing 250 mM sodium chloride, twice with lysis buffer containing 1 M sodium chloride, and then twice with kinase assay buffer (20 mM HEPES, pH 1 mM DTT, 1 mM NaF and 1 mM sodium orthovanadate). After the last wash, the beads were resuspended in 20 .l kinase assay buffer supplemented with 10 mCi 32P]-ATP, 20 mM j-glycerophosphate, mM PNPP, 20 mM MgCI 2 1 mM EGTA, 100 gIM cold ATP and 0.5 mg/nl Myelin Basic Protein (Sigma), and incubated for 20 min. at 37 0 C. Reactions were stopped with 20 il SDS-sample buffer, run on a denaturing 4-20% SDS polyacrylamide gel, and analyzed by phosphoimager.
43 PI272 EXAMPLE 11 Expression of fused in E. coli The DNA sequence encoding human fused is initially amplified using selected PCR primers. The primers should contain restriction enzyme sites that correspond to the restriction enzyme sites on the selected expression vector. A variety of expression vectors may be employed. An example of a suitable vector is pBR322 (derived from E. coli; see Bolivar et al., Gene, 2:95 (1977)) which contains genes for ampicillin and tetracycline resistance. The vector is digested with restriction enzyme and dephosphorylated. The PCR amplified sequences are then ligated into the vector. The vector will preferably include sequences that encode for an antibiotic resistance gene, a trp promoter, a polyhis leader (including the first six STII codons, polyhis sequence, and enterokinase cleavage site), the vertebrate fused coding region, lambda transcriptional terminator, and an argU gene.
The ligation mixture is then used to transform a selected E. coli strain using the methods described in Sambrook ct al., supra. Transformants are identified by their ability to grow on LB plates and antibiotic resistant colonies are then selected. Plasmid DNA can be isolated and confirmed by restriction analysis and DNA sequencing.
Selected clones can be grown overnight in liquid culture medium such as LB broth supplemented with antibiotics. The overnight culture may subsequently be used to inoculate a larger scale culture. The cells are then grown to a desired optical density, during which the expression promoter is turned on.
After culturing the cells for several more hours, the cells can be harvested by centrifugation. The cell pellet obtained by the centrifugation can be solubilized using various agents known in the art, and the solubilized vertebratefused protein can then be purified using a metal chelating column under conditions that allow tight binding of the protein.
EXAMPLE 12 Expression of fused in mammalian cells The vector, pRK5 (see EP 307,247, published March 15, 1989), is employed as the expression vector. Optionally, the vertebrate fused DNA is ligated into pRK5 with selected restriction enzymes to allow insertion of the vertebrate fused DNA using ligation methods such as described in Sambrook et al., supra.
The resulting vector is called In one embodiment, the selected host cells may be 293 cells. Human 293 cells (ATCC CCL 1573) are grown to confluence in tissue culture plates in medium such as DMEM supplemented with fetal calf serum and optionally, nutrient components and/or antibiotics. About 10 pg pRK5-fused DNA is mixed with about I pg DNA encoding the VA RNA gene [Thimmappaya et al., Cell, 31:543 (1982)] and dissolved in 500 pl of I mM Tris-HCI, 0.1 mM EDTA, 0.227 M CaCI 2 To this mixture is added, dropwise, 500 pl of mM HEPES (pH 7.35), 280 mM NaCI, 1.5 mM NaPO 4 and a precipitate is allowed to form for 10 minutes at 25°C. The precipitate is suspended and added to the 293 cells and allowed to settle for about four hours at 37°C. The culture medium is aspirated off and 2 ml of 20% glycerol in PBS is added for 30 seconds. The 293 cells are then washed with serum free medium, fresh medium is added and the cells are incubated for about 5 days.
P1272 Approximately 24 hours after the transfections, the culture medium is removed and replaced with culture medium (alone) or culture medium containing 200 pCi/ml 35S-cysteine and 200 pCi/ml methionine. After a 12 hour incubation, the conditioned medium is collected, concentrated on a spin filter, and loaded onto a 15% SDS gel. The processed gel may be dried and exposed to film for a selected period of time to reveal the presence of vertebrate fused polypeptide. The cultures containing transfected cells may undergo further incubation (in serum free medium) and the medium is tested in selected bioassays.
In an alternative technique, vertebrate fused may be introduced into 293 cells transiently using the dextran sulfate method described by Somparyrac et al., Proc. Natl. Acad Sci., 12:7575 (1981). 293 cells are grown to maximal density in a spinner flask and 700 pg pRK5-fused DNA is added. The. cells are first concentrated from the spinner flask by centrifugation and washed with PBS. The DNA-dextran precipitate is incubated on the cell pellet for four hours. The cells are treated with 20% glycerol for 90 seconds, washed with tissue culture medium, and re-introduced into the spinner flask containing tissue culture medium, pg/ml bovine insulin and 0.1 pg/ml bovine transferrin. After about four days, the conditioned media is centrifuged and filtered to remove cells and debris. The sample containing expressed vertebrate fused can then be concentrated and purified by any selected method, such as dialysis and/or column chromatography.
In another embodiment, vertebrate fused can be expressed in CHO cells. The pSUi-fused can be transfected into CHO cells using known reagents such as CaPO 4 or DEAE-dextran. As described above, the cell cultures can be incubated, and the medium replaced with culture medium (alone) or medium containing a radiolabel such as 3 5 S-methionine. After determining the presence of vertebrate fused polypeptide, the culture medium may be replaced with serum free medium. Preferably, the cultures are incubated for about 6 days, and then the conditioned medium is harvested. The medium containing the expressed vertebrate fused can then be concentrated and purified by any selected method.
Epitope-tagged vertebrate fused may also be expressed in host CHO cells. The vertebrate fused may be subcloned out of the pRK5 vector. The subclone insert can undergo PCR to fuse in frame with a selected epitope tag such as a poly-his tag into an expression vector. The poly-his tagged vertebrate fused insert can then be subcloned into a SV40 driven vector containing a selection marker such as DHFR for selection of stable clones. Finally, the CHO cells can be transfected (as described above) with the SV40 driven vector.
Labeling may be performed, as described above, to verify expression. The culture medium containing the expressed poly-His tagged vertebrate fused can then be concentrated and purified by any selected method, such as by Ni -chelate affinity chromatography.
EXAMPLE 13 Expression of vertebrate fused in Yeast The following method describes recombinant expression of vertebrate fused in yeast.
First, yeast expression vectors are constructed for intracellular production or secretion of vertebrate fused from the ADH2/GAPDH promoter. DNA encoding vertebrate fused, a selected signal peptide and the promoter is inserted into suitable restriction enzyme sites in the selected plasmid to direct intracellular expression of vertebrate fused. For secretion, DNA encoding vertebratefused can be cloned into the selected P1272 plasmid, together with DNA encoding the ADH2/GAPDH promoter, the yeast alpha-factor secretory signal/leader sequence, and linker sequences (if needed) for expression of vertebrate fused.
Yeast cells, such as yeast strain ABI10, can then be transformed with the expression plasmids described above and cultured in selected fermentation media. The transformed yeast supernatants can be analyzed by precipitation with 10% trichloroacetic acid and separation by SDS-PAGE, followed by staining of the gels with Coomassie Blue stain.
Recombinant vertebrate fused can subsequently be isolated and purified by removing the yeast cells from the fermentation medium by centrifugation and then concentrating the medium using selected cartridge filters. The concentrate containing vertebrate fused may further be purified using selected column chromatography resins.
EXAMPLE 14 Expression of vertebrate fused in Baculovirus-Infected Insect Cells The following method describes recombinant expression of vertebrate fused in Baculovirus-infected insect cells.
The vertebrate fused is fused upstream of an epitope tag contained within a baculovirus expression vector. Such epitope tags include poly-his tags and immunoglobulin tags (like Fc regions of IgG). A variety of plasmids may be employed, including plasmids derived from commercially available plasmids such as pVL1393 (Novager). Briefly, the vertebrate fused or the desired portion of the vertebrate fused (such as the sequence encoding the extracellular domain of a transmembrane protein) is amplified by PCR with primers complementary to the 5' and 3' regions. The 5' primer may incorporate flanking (selected) restriction enzyme sites. The product is then digested with those selected restriction enzymes and subcloned into the expression vector.
Recombinant baculovirus is generated by co-transfecting the above plasmid and BaculoGoldTM virus DNA (Pharmingen) into Spodoptera frugiperda cells (ATCC CRL 1711) using lipofectin (commercially available from GIBCO-BRL). After 4 5 days of incubation at 28 0 C, the released viruses are harvested and used for further amplifications. Viral infection and protein expression is performed as described by O'Reilley et al., Baculovirus expression vectors: A Laboratory Manual, Oxford: Oxford University Press (1994).
Expressed poly-his tagged vertebrate fused can then be purified, for example, by Ni 2+-chelate affinity chromatography as follows. Extracts are prepared from recombinant virus-infected Sf9 cells as described by Rupert et al., Nature, 362:175-179 (1993). Briefly, Sf9 cells are washed, resuspended in sonication buffer (25 mL Hepes, pH 7.9; 12.5 mM MgCI 2 0.1 mM EDTA; 10% Glycerol; 0.1% NP-40; 0.4 M KCI), and sonicated twice for 20 seconds on ice. The sonicates are cleared by centrifugation, and the supernatant is diluted 50-fold in loading buffer (50 mM phosphate, 300 mM NaCI, 10% Glycerol, pH 7.8) and filtered through a 0.45 pim filter. A Ni 2+-NTA agarose column (commercially available from Qiagen) is prepared with a bed volume of 5 mL, washed with 25 mL of water and equilibrated with 25 mL of loading buffer. The filtered cell extract is loaded onto the column at 0.5 mL per minute. The column is washed to P1272 baseline A 2 80 with loading buffer, at which point fraction collection is started. Next, the column is washed with a secondary wash buffer (50 mM phosphate; 300 mM NaCI, 10% Glycerol, pH which elutes nonspecifically bound protein. After reaching A 280 baseline again, the column is developed with a 0 to 500 mM Imidazole gradient in the secondary wash buffer. One mL fractions are collected and analyzed by SDS- PAGE and silver staining or western blot with Ni 2+-NTA-conjugated to alkaline phosphatase (Qiagen).
Fractions containing the eluted His l0-tagged vertebrate fused are pooled and dialyzed against loading buffer.
Alternatively, purification of the IgG tagged (or Fc tagged) vertebrate fused can be performed using known chromatography techniques, including for instance, Protein A or protein G column chromatography EXAMPLE Preparation of Antibodies that Bind Vertebrate fused This example illustrates preparation of monoclonal antibodies, which can specifically bind vertebrate fused.
Techniques for producing the monoclonal antibodies are known in the art and are described, for instance, in Goding, supra. Immunogens that may be employed include purified vertebrate fused, fusion proteins containing vertebrate fused, and cells expressing recombinant vertebrate fused on the cell surface.
Selection of the immunogen can be made by the skilled artisan without undue experimentation.
Mice, such as Balb/c, are immunized with the vertebrate fused immunogen emulsified in complete Freund's adjuvant and injected subcutaneously or intraperitoneally in an amount from 1-100 micrograms. Alternatively, the immunogen is emulsified in MPL-TDM adjuvant (Ribi Immunochemical Research, Hamilton, MT) and injected into the animal's hind foot pads. The immunized mice are then boosted 10 to 12 days later with additional immunogen emulsified in the selected adjuvant. Thereafter, for several weeks, the mice may also be boosted with additional immunization injections. Serum samples m-y be periodically obtained from the mice by retro-orbital bleeding for testing in ELISA assays to detect vertebrate fused antibodies.
After a suitable antibody titer has been detected, the animals "positive" for antibodies can be injected with a final intravenous injection of vertebrate fused. Three to four days later, the mice are sacrificed and the spleen cells are harvested. The spleen cells are then fused (using 35% polyethylene glycol) to a selected murine myeloma cell line such as P3X63AgU.l, available from ATCC, No. CRL 1597. The fusions generate hybridoma cells which can then be plated in 96 well tissue culture plates containing HAT (hypoxanthine, aminopterin, and thymidine) medium to inhibit proliferation of non-fused cells, myeloma hybrids, and spleen cell hybrids.
The hybridoma cells will be screened in an ELISA for reactivity against vertebrate fused.
Determination of "positive" hybridoma cells secreting the desired monoclonal antibodies against vertebrate fused is within the skill in the art.
The positive hybridoma cells can be injected intraperitoneally into syngeneic Balb/c mice to produce ascites containing the anti- vertebrate fused monoclonal antibodies. Alternatively, the hybridoma cells can be grown in tissue culture flasks or roller bottles. Purification of the monoclonal antibodies P1272 produced in the ascites can be accomplished using ammonium sulfate precipitation, followed by gel exclusion chromatography. Alternatively, affinity chromatography based upon binding of antibody to protein A or protein G can be employed.
Deposit of Material The following materials have been deposited with the American Type Culture Collection, 12301 Parklawn Drive, Rockville, MD, USA (ATCC): Designation: ATCC Dep. No. Deposit Date 1272 209637 2/19/98 This deposit was made under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure and the Regulations thereunder (Budapest Treaty). This assures maintenance of a viable culture of the deposit for 30 years from the date of deposit. The deposit will be made available by ATCC under the terms of the Budapest Treaty, and subject to an agreement between Genentech, Inc. and ATCC, which assures permanent and unrestricted availability of the progeny of the culture of the deposit to the public upon issuance of the pertinent U.S.
patent or upon laying open to the public of any U.S. or foreign patent application, whichever comes first, and assures availability of the progeny to one determined by the U.S. Commissioner of Patents and Trademarks to be entitled thereto according to 35 USC §122 and the Commissioner's rules pursuant thereto (including 37 CFR 1.14 with particular reference to 886 OG 638).
The assignee of the present application has agreed that if a culture of the materials on deposit should die or be lost or destroyed when cultivated under suitable conditions, the materials will be promptly replaced on notification with another of the same. Availability of the deposited material is not to be construed as a license to practice the invention in contravention of the rights granted under the authority of any government in accordance with its patent laws.
The foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the invention. The present invention is not to be limited in scope by the construct deposited, since the deposited embodiment is intended as a single illustration of certain aspects of the invention and any constructs that are functionally equivalent are within the scope of this invention. The deposit of material herein does not constitute an admission that the written description herein contained is inadequate to enable the practice of any aspect of the invention, including the best mode thereof, nor is it to be construed as limiting the scope of the claims to the specific illustrations that it represents. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims.

Claims (44)

1. An isolated nucleic acid molecule, which is antisense to a nucleic acid molecule having at least an 80% nucleic acid sequence identity to a DNA molecule which encodes a vertebrate fused polypeptide comprising the sequence of amino acids 1 to about 260 of Figure 1 (SEQ ID NO:24) and which has fused biological activity.
2. A nucleic acid of claim 1, wherein the sequence identity is at least
3. A nucleic acid of claim 2, wherein the sequence identity is at least
4. A nucleic acid of claim 3, wherein the sequence identity is at least A nucleic acid of any one of claims 1 to 4, which is antisense to a nucleic acid molecule having at least sequence identity to a DNA molecule which encodes a human fused polypeptide comprising the sequence of amino acids 1 to 1315 of Figure 1 (SEQ ID NO:2).
6. A nucleic acid of claim 5, wherein the sequence identity is at least
7. A nucleic acid of claim 6, wherein the sequence identity is at least
8. A nucleic acid of claim 7, wherein the sequence identity is at least
9. A nucleic acid of any one of claims 1 to 8, which is antisense to a nucleic acid molecule encoding the vertebrate fused polypeptide fragment having amino acid residues 1 to 260 of Figure 1 (SEQ ID NO:24). H.\Simeona\Keep\Speci\PS2113 divisional.doc 11/02/04 50 A nucleic acid of any one of claims 1 to 9, which is antisense to a nucleic acid molecule encoding a vertebrate fused polypeptide having a lysine at amino acid position 33.
11. An isolated nucleic acid, which is antisense to a nucleic acid molecule having at least an 80% nucleic acid sequence identity to a DNA molecule encoding the same mature polypeptide as that encoded by the cDNA in ATCC Deposit No. 209637.
12. A nucleic acid of claim 11, wherein the sequence identity is at least
13. A nucleic acid of claim 12, wherein the sequence identity is at least
14. A nucleic acid of claim 13, wherein the sequence identity is at least A vector comprising a nucleic acid of any one of claims 1 to 14.
16. A vector of claim 15, operably linked to control sequences recognized by a host cell transformed with the vector.
17. A host cell transformed with a vector of claim or claim 16.
18. A host cell of claim 17, which is mammalian.
19. A host cell of claim 18, which is CHO cell. A host cell of claim 17, which is prokaryotic. H.\Simeona\Keep\Speci\P52113 divisional.doc 11/02/04 51
21. A host cell of claim 20, which is E. coli.
22. A host cell of claim 17, which is a yeast cell.
23. A host cell of claim 22, which is Saccharomyces cerevisiae.
24. An antibody which specifically binds to an isolated vertebrate fused polypeptide, the polypeptide having at least 80% amino' acid sequence identity to amino acid residues 1 to 1315 of Figure 1 (SEQ ID NO:2), and having fused biological activity. An antibody of claim 24, wherein the polypeptide has at least 85% amino acid sequence identity to amino acid residues 1 to 1315 of Figure 1 (SEQ ID NO:2) and has fused biological activity.
26. An antibody of claim 25, wherein the polypeptide has at least 90% amino acid sequence identity to amino acid residues 1 to 1315 of Figure 1 (SEQ ID NO:2) and has fused biological activity.
27. An antibody of claim 26, wherein the polypeptide has at least 95% amino acid sequence identity to amino acid residues 1 to 1315 of Figure 1 (SEQ ID NO:2) and has fused biological activity.
28. An antibody of any one of claims 24 to 27, wherein the polypeptide comprises amino acid residues 1 to 1315 of Figure 1 (SEQ ID NO:2). S29. An antibody which specifically binds to an isolated, native sequence vertebrate fused polypeptide, the polypeptide having at least 80% amino acid sequence identity to the human fused polypeptide encoded by the cDNA deposited under accession number ATCC 209637, and H.\Simeona\Keep\Speci\P52113 divisional.doc 11/02/04 52 having fused biological activity. An antibody of claim 29, wherein the sequence identity is at least
31. An antibody of claim 30, herein the sequence identity is at least
32. An antibody of claim 31, wherein the sequence identity is at least
33. An antibody of any one of claims 24 to 32, wherein the polypeptide is encoded by the cDNA deposited under accession number ATCC 209637.
34. An antibody of any one of claims 24 to 33, wherein the vertebrate fused polypeptide is fused to a heterologous amino acid sequence.
35. An antibody of claim 34, wherein the heterologous amino acid sequence is an epitope tag sequence.
36. An antibody of claim 34 or claim 35, wherein the heterologous amino acid sequence is a constant region of an immunoglobulin.
37. A method of treating a disease or disorder characterised by inactive or insufficient Hh signalling, the method comprising the step of administering to a subject in need thereof an effective amount of a nucleic acid molecule of any one of claims 1 to 14 or an antibody of any one of claims 24 to 36.
38. A method of claim 37, wherein the disease or disorder is a degenerative disorder of the nervous system, gut disease, bone disease, skin disease, diseases of the testis, ulcers, lung disease or diseases of the pancreas. H,\Simeona\Keep\Speci\P52113 divisional.doc 11/02/04 53
39. A method of claim 37 or claim 38, wherein the disease or disorder is selected from the group consisting of Parkinson's disease, memory deficits, Alzheimer's disease, Lou Gehrig's disease, Huntington's disease, schziophrenia, stroke, drug addiction, diabetes, and osteoporosis. A method of treating a disease or disorder characterised by Hh signalling, the method comprising the step of administering to a subject in need thereof an effective amount of an antisense nucleic acid molecule of any one of claims 1 to 14 or an antibody of any one of claims 24 to 36.
41. A method of claim 40, wherein the disease or disorder is Basal Cell Carcinoma.
42. A method of male contraception, the method comprising the step of administering to a subject in need thereof an effective amount of an antisense nucleic acid molecule of any one of claims 1 to 14 or an antibody of any one of claims 24 to 36.
43. Use of a nucleic acid molecule of any one of claims 1 to 14 or an antibody of any one of claims 24 to 36, in the treatment of a disease or disorder characterised by inactive or insufficient Hh signalling.
44. Use of a nucleic acid molecule of any one of claims 1 to 14 or an antibody of any one of claims 24 to 36, in the preparation of a medicament for the treatment of a disease or disorder characterised by inactive or insufficient Hh signalling. Use of claim 42 or claim 43, wherein the disease or disorder is a degenerative disorder of the nervous H:\Simeona\Keep\Speci\P52113 divisional.doc 11/02/04 54 system, gut disease, bone disease, skin disease, diseases of the testis, ulcers, lung disease or diseases of the pancreas.
46. Use of claim 42 or claim 43, wherein the disease or disorder is selected from the group consisting of Parkinson's disease, memory deficits, Alzheimer's disease, Lou Gehrig's disease, Huntington's disease, schziophrenia, stroke, drug addiction, diabetes, and osteoporosis.
47. Use of a nucleic acid molecule of any one of claims 1 to 14 or an antibody of any one of claims 24 to 36, in the treatment of a disease or disorder characterised by Hh signalling.
48. Use of a nucleic acid molecule of any one of claims 1 to 14 or an antibody of any one of claims 24 to 36, in the preparation of a medicament for the treatment of- a disease or disorder characterised by Hh signalling.
49. Use of claim 46 or claim 47, wherein the disease or disorder is Basal Cell Carcinoma. Use of a nucleic acid molecule of any one of claims 1 to 14 or an antibody of any one of claims 24 to 36, in male contraception.
51. Use of a nucleic acid molecule of any one of claims 1 to 14 or an antibody of any one of claims 24 to 36, in the preparation of a medicament for male contraception.
52. An antisense nucleic acid according to any one of claims 1 or 11, substantially as herein described with reference to any of the examples and figures.
53. An antibody according to claim 24 or claim 29, H.\Simeona\Keep\Speci\P52113 divisional.doc 11/02/04 55 substantially as herein described with reference to any of the examples and figures.
54. A method according to any one of claims 37, 40 or 42, substantially as herein described with reference to any of the examples or figures. Use according to any one of claims 43, 44, 47, 48, 50 or 51, substantially as herein described with reference to any of the examples or figures. Dated this 12th day of February 2004 GENENTECH, INC. By their Patent Attorneys GRIFFITH HACK Fellows Institute of Patent and Trade Mark Attorneys of Australia H,\Simeona\Keep\Speci\P52113 divisional.doc 11/02/04
AU2004200576A 1998-02-26 2004-02-12 Human homolog of the drosophila protein "fused" Expired AU2004200576B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/031563 1998-02-26
AU28779/99A AU767491B2 (en) 1998-02-26 1999-02-25 Human homolog of the drosophila protein "fused"
PCT/US1999/004112 WO1999043828A1 (en) 1998-02-26 1999-02-25 Human homolog of the drosophila protein 'fused'

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU28779/99A Division AU767491B2 (en) 1998-02-26 1999-02-25 Human homolog of the drosophila protein "fused"

Publications (2)

Publication Number Publication Date
AU2004200576A1 true AU2004200576A1 (en) 2004-03-11
AU2004200576B2 AU2004200576B2 (en) 2006-10-26

Family

ID=39365577

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2004200576A Expired AU2004200576B2 (en) 1998-02-26 2004-02-12 Human homolog of the drosophila protein "fused"

Country Status (1)

Country Link
AU (1) AU2004200576B2 (en)

Also Published As

Publication number Publication date
AU2004200576B2 (en) 2006-10-26

Similar Documents

Publication Publication Date Title
EP1056868B1 (en) Human homolog of the drosophila protein 'fused'
JP2010046077A (en) Vertebrate patched-2 protein
US8394939B2 (en) Nucleic acid encoding fused
AU2004200576B2 (en) Human homolog of the drosophila protein "fused"
US6531579B1 (en) Fused polypeptides
US7811777B2 (en) Methods of screening for agonists and antagonists of patched-2
US8648174B2 (en) Ucp4
US6727079B1 (en) cDNA encoding a gene BOG (B5T Over-expressed Gene) and its protein product
CA2349839C (en) Ucp5
US7342102B2 (en) Uncoupling protein 5 (UCP5)
ZA200106937B (en) Human suppressor of fused.
NZ513762A (en) Human suppressor of fused

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired