US20020091248A1 - Myosin IXa and cyclic nucleotide gated channel-15 (CNGC-15) polynucleotides, polypeptides, compositions, methods, and uses thereof - Google Patents

Myosin IXa and cyclic nucleotide gated channel-15 (CNGC-15) polynucleotides, polypeptides, compositions, methods, and uses thereof Download PDF

Info

Publication number
US20020091248A1
US20020091248A1 US09/851,682 US85168201A US2002091248A1 US 20020091248 A1 US20020091248 A1 US 20020091248A1 US 85168201 A US85168201 A US 85168201A US 2002091248 A1 US2002091248 A1 US 2002091248A1
Authority
US
United States
Prior art keywords
polypeptide
seq
cngc
nucleotide sequence
leu
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US09/851,682
Inventor
Arwen Adams
Choi Chin
David Duhl
Susan Gorman
Song Leng
Val Sheffield
Juliet Welch
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.)
Novartis Vaccines and Diagnostics Inc
Original Assignee
Chiron Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chiron Corp filed Critical Chiron Corp
Priority to US09/851,682 priority Critical patent/US20020091248A1/en
Publication of US20020091248A1 publication Critical patent/US20020091248A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4716Muscle proteins, e.g. myosin, actin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • This invention relates to the fields of molecular biology and pharmaceutical research. More specifically, this invention relates to the identification and recombinant expression of two new genes, cyclic nucleotide gated channel-15 (CNGC-15) and Myosin IXa. Accordingly, isolation of the human myosin IXa gene will prove useful in the study, diagnosis and treatment of Bardet-Biedl Syndrome, Usher Syndrome and related conditions.
  • CNGC-15 cyclic nucleotide gated channel-15
  • Myosin IXa Myosin IXa
  • Usher Sydrome typel (USH1) is characterized by a profound congenital sensoneural hearing loss, vestibular dysfunction and prepubescent onset retinitis pigmentosa. Family studies indicated that three genes with different chromosomal locations are responsible for USH1; a defect in any one of these genes causes the disease. Of these, the gene USH1B mapped to a region homologous to the murine region containing the mouse deafness mutant shaker-1 which results from mutations in myosin VIIa. Subsequent work confirmed that myosin VIIa is the cause of Usher Sydrome type 1B and localized the myosin VIIa protein to the receptor cells of the inner ear (Hasson et al. (1995) Genomics 36:431-439) and the connecting cilia of photoreceptor cells in the retina (Liu et al. (1997) Cell Motil. Cytoskel. 37:240-252).
  • Retinitis pigmentosa is also associated with Bardet-Biedl Syndrome.
  • Bardet-Biedl Syndrome is further characterized by obesity, retinal degeneration, syndactyly and/or polydactyly, hypogenitalism and mental retardation (Schachat et al. (1982) Arch. Ophthalmol. 100:285-288; Green et al. (1989) N. Engl. J. Med. 321:1002-1009) and can result from aberrations in one of at least four different genes.
  • Other publications relating to myosin IXa include Bruford et al. (1997) Genomics 41:93-99 and Bement et al. (1994) Proc. Natl. Acad. Sc. USA 91:6549-6553.
  • the present invention discloses the amino acid and nucleic acid sequences of a new Cyclic nucleotide gated channel (CNGC) and Myosin that map to the region of the human chromosome associated with Bardet-Biedl Syndrome.
  • CNGCs comprise a family of multimeric protein ion channels that open in response to the binding of a cyclic nucleotide to an intracellular domain.
  • the two new proteins, CNGC-15 and Myosin IXa are useful in the study, diagnosis and treatment of Bardet-Biedl Syndrome and Usher Syndrome.
  • CNGC-15 and/or Myosin IXa polypeptides, or agonists or antagonists include hearing loss, retinis pigmentosa, obesity, hypogonadism, sterility, polydactyly, brachydactyly, syndactyly, mental retardation, renal abnormalities, hypertension, diabetes and cardiovascular abnormalities.
  • compositions and methods for expressing cyclic nucleotide gated channel-15 (CNGC-15) and Myosin IXa are provided.
  • the compositions comprise CNGC-15 and Myosin IXa polypeptides and derivatives thereof, nucleotide sequences, expression cassettes, transformed cells and antibodies to these polypeptides.
  • Methods for the expression and detection of CNGC-15 and Myosin IXa nucleotides and polypeptides and compositions for the treatment of these conditions are provided.
  • FIG. 1 sets forth the amino acid sequence of the human Myosin IXa polypeptide (SEQ ID NO: 1).
  • FIG. 2 sets forth the nucleotide sequence of the human Myosin IXa cDNA (SEQ ID NO: 2).
  • the ATG translational intiation codon at position 243 and the TGA translational stop codon at position 7887 are underlined.
  • FIG. 3 sets forth the nucleotide sequence of a human CNGC-15 partial cDNA (SEQ ID NO: 3).
  • the present invention is drawn to compositions and methods useful in the treatment of Bardet-Biedl Syndrome, Usher Syndrome, hearing loss, retinis pigmentosa, obesity, hypogonadism, sterility, polydactyly, brachydactyly, syndactyly, mental retardation, renal abnormalities, hypertension, diabetes and cardiovascular abnormalities. More particularly, two new polypeptides and the genes encoding them, CNGC-15 and Myosin IXa, have been identified that are useful in the treatment of these and a variety of other conditions.
  • the human Myosin IXa polypeptide and cDNA are shown in FIGS. 1 and 2, as SEQ ID NO: 1 and SEQ ID NO: 2, respectively.
  • the human CNGC-15 partial cDNA is shown in FIG. 3 as SEQ ID NO: 3.
  • Another object of the invention is to provide polypeptides that are mutants, fragments and derivatives of CNGC-15 or Myosin Ixa.
  • Regions of particular importance of Myosin IXa include: the open reading frame (nucleotides 243-7886 of SEQ ID NO: 2) encoding a 2548 amino acid protein (SEQ ID NO: 1); the N-terminal extension (residues 1-146 of SEQ ID NO: 1) encoded by nucleotides 243-680 of SEQ ID NO: 2; the head domain (residues 147-719 of SEQ ID NO: 1) encoded by nucleotides 683-2399 of SEQ ID NO: 2; the ATP binding loop (residues 239-246 of SEQ ID NO: 1) encoded by nucleotides 959-977 of SEQ ID NO: 2; the minimal length 2nd insert (residues 720-835 of SEQ ID NO: 1) encoded by nucleotides
  • the invention provides a Myosin IXa polypeptide comprising an amino acid sequence selected from the group consisting of:
  • the Myosin IXa polypeptides of the invention will have at least about 61% homology to SEQ ID NO:1, preferably at least about 70% homology, more preferably at least about 80% homology and most preferably at least about 90% homology.
  • the polypeptides of b), c), d), e), f) or above will have at least about 95% homology to SEQ ID NO:1, more preferably at least about 96% homology, even more preferably at least about 97% homology and most preferably at least about 98% homology.
  • the invention provides a CNGC-15 polypeptide comprising an amino acid sequence selected from the group consisting of:
  • the CNGC-15 polypeptides of the invention will have at least about 70% homology to a), b), c) or d) above, preferably at least about 80% homology, more preferably at least about 85% homology and most preferably at least about 90% homology.
  • a chimeric polypeptide comprising a CNGC-15 polypeptide, or fragment thereof and a polypeptide of interest.
  • the invention provides a chimeric polypeptide comprising a Myosin IXa polypeptide, or fragment thereof, fused to a polypeptide of interest. Nucleotide sequences encoding chimeric CNGC-15 and Myosin IXa polypeptides are also provided.
  • Yet another object of the invention is to provide polynucleotides that encode the mutants, fragments, and derivatives, as well as the native CNGC-15 and Myosin IXa. Accordingly, the invention provides an isolated nucleotide sequence selected from the group consisting of:
  • nucleotide sequence comprising at least about 390 contiguous bases of SEQ ID NO: 2;
  • nucleotide sequence comprising nucleotides 243-7085 of SEQ ID NO: 2;
  • nucleotide sequence comprising nucleotides 243-680 of SEQ ID NO: 2;
  • nucleotide sequence comprising nucleotides 683-2399 of SEQ ID NO: 2;
  • nucleotide sequence comprising nucleotides 959-977 of SEQ ID NO: 2;
  • nucleotide sequence comprising nucleotides 2402-2747 of SEQ ID NO: 2;
  • nucleotide sequence comprising nucleotides 6473-6899 of SEQ ID NO: 2.
  • the invention similarly provides an isolated nucleotide sequence selected from the group consisting of:
  • nucleotide sequence comprising at least 45 contiguous nucleotides of the sequence of SEQ ID NO:3;
  • polynucleotides can be operably linked to heterologous promoters to form expression cassettes.
  • the expression cassettes can be introduced into suitable host cells for expression of CNGC-15 and/or Myosin IXa polypeptides and derivatives thereof.
  • Another object of the invention is to provide a transformed cell transiently expressing or having stably incorporated into its genome an expression vector comprising a promoter operably linked to a nucleotide sequence encoding a CNGC-15 or Myosin IXa polypeptide, or a fragment, derivative mutant or fusion thereof.
  • a further object of the invention is to provide a method for producing a CNGC-15 or Myosin IXa polypeptide or a derivative thereof, comprising:
  • Another object of the invention is to provide a method for detecting CNGC-15 polynucleotides.
  • the method comprises:
  • nucleic acid probe which hybridizes to a nucleotide encoding a CNGC-15 polypeptide or a mutant, fragment or derivative thereof;
  • a further object of the invention is to provide a method for detecting unconventional Myosin polynucleotides.
  • the method comprises:
  • nucleic acid probe which hybridizes to a nucleotide encoding a Myosin IXa or a mutant, fragment or derivative thereof;
  • Yet another object of the invention is to provide antibodies to the CNGC-15 and Myosin IXa polypeptides.
  • Another object of the invention is to provide a method for detecting CNGC-15 polypeptides.
  • the method comprises:
  • Still another object of the invention is to provide a method for detecting Myosin IXa polypeptides.
  • the method comprises:
  • the invention further provides a method for treating Bardet-Biedl Syndrome, Usher Syndrome, hearing loss, retinis pigmentosa, obesity, hypogonadism, sterility, polydactyly, brachydactyly, syndactyly, mental retardation, renal abnormalities, hypertension, diabetes and cardiovascular abnormalities, comprising administering a therapeutically effective amount of a CNGC-15 or Myosin IXa polypeptide, or derivative thereof to a subject in need of such treatment.
  • the invention provides a composition comprising CNGC-15 or Myosin IXa or an active derivative thereof, and a pharmaceutically acceptable carrier.
  • native CNGC-15 and native Myosin IXa refer to the polypeptides found in nature.
  • native CNGC-15 and native Myosin IXa are polypeptides with substantial amino acid sequence identity to SEQ ID NO: 1 and SEQ ID NO: 3, respectively. This definition includes allelic variants and other naturally occurring modifications of SEQ ID NO: 1 and SEQ ID NO: 3.
  • CNGC-15 polypeptides and “Myosin IXa polypeptides” include mutants, fragments, derivatives and fusions as well as the native CNGC-15 or Myosin IXa.
  • “Mutants” of the native CNGC-15 or Myosin IXa are polypeptides having an amino acid sequence which retain at least about 50% amino acid sequence identity with SEQ ID NO: 1 or SEQ ID NO: 3, respectively; more typically, at least about 60%; even more typically, at least about 80%. Preferably mutants will retain at least about 85% amino acid sequence identity with SEQ ID NO: 1 or SEQ ID NO: 3; more preferably, at least about 90%; even more preferably, at least about 95%. These differences may be conservative amino acid substitutions, insertions or deletions in the amino acid sequence.
  • “Fragments” possess the same amino acid sequence of the native or mutant CNGC-15 or Myosin IXa polypeptides except the fragments lack the amino and/or carboxyl terminal sequences of the native or mutant polypeptide.
  • “Derivatives” possess the same amino acid sequence of the native, mutant or fragment CNGC-15 and Myosin IXa, but may contain amino acid substitutes, glycosylated residues or other chemical modifications.
  • Fusions or “chimeric polypeptides” are mutants, fragments, or the native CNGC-15 or Myosin IXa that also include amino and/or carboxyl terminal amino acid extensions.
  • Myosin Ixa fragments may be fused with fragments of other unconventional myosins.
  • Unconventional myosins are members of the myosin superfamily that display the general head, neck and tail domain structure of conventional myosins (Mooseker et al. (1995) Annu. Rev. Cell. Dev. Biol. 11:633-75), but differ from conventional myosins by changes in these and/or the inclusion of additional functional domains.
  • the head region shared among all myosin molecules consists of conserved amino acid sequences that span approximately 80 kDa while the neck region contains conserved motifs that bind one to six light chains of protein from the calmodulin/EF-hand family (Bahler et al. (1996) Curr. Op. Cell. Biol. 8:18-22). It is the unconventional myosin tail domains that show the most variation from conventional myosin and from each other. For example, although myosin Va has a coiled coil tail domain, as does conventional myosin, it also contains a PEST site (required for calpain cleavage) and a globular C-terminal domain which may be used for interactions with cargo or docking proteins.
  • Myosin VIIa also has a coiled coil tail domain, however it contains additional domains different from those found in myosin Va, such as the talin domain which is thought to be active in binding plasma membranes.
  • the class IX myosins (composed of rat myr5 and human myosin IXb) have even less similarity to conventional myosin and to the other nine classes of unconventional myosins. These molecules lack a coiled coil domain in the tail region, but do contain a GAP (GTP activating protein) domain which increases the GTPase activity of rhoA and Cdc42 (Reinhard et al. (1995) EMBO J. 14:697-704).
  • GAP GTP activating protein
  • Class IX myosins also have two large insertions in the conserved head domain that are absent in conventional and other unconventional myosins, and may act as regulatory domains affecting the mechanochemical properties of the protein.
  • all classes bind actin and have either been shown to be or are postulated to act as molecular motors (Wang et al. (1996) Science 273:660-663), thereby interacting with the actin cytoskeleton.
  • the characterization and chromosomal mapping of myosin Va and myosin VIIa suggested that these molecules may be responsible for specific human diseases. Indeed, defects in myosin Va are associated with Griscelli disease (Pastural et al. (1997) Nature Genet. 16:289-292), while mutations in myosin VIIa are responsible for Usher Sydrome type1B (Weil et al. (1995) Nature 374:60-61.
  • the number or type of the amino acid substitutions is not critical, nor is the length or number of the amino acid deletions, or amino acid extensions that are incorporated in the CNGC-15 or Myosin IXa polypeptides. However, all of these polypeptides will exhibit at least about 20% of one of the activities of the native CNGC-15 or Myosin IXa. More typically, the polypeptides exhibit at least about 40%, even more typically the polypeptides exhibit at least about 60% of one of the native CNGC-15 or Myosin IXa activities. All these polypeptides will retain at least about 50% amino acid identity with SEQ ID NO: 1 or SEQ ID NO: 3; more typically, at least about 60%; even more typically, at least about 80%. Preferably, these polypeptides will retain at least about 85% amino acid sequence identity with SEQ ID NO: 1 or the amino acid sequence encoded by SEQ ID NO: 3; more preferably, at least about 90%; even more preferably, at least about 95%.
  • Myosin IXa activities include immunological activities, ATP binding, ATPase activity (Shyamata et al. (1990) J Cell Biol 110:1137-1147), zinc binding (Reinhard et al. (1995) EMBO J 14:697-704; Quest et. (1994) J Biol Chem 269:2961-2970) calmodulin/EF binding (Glenney et al.
  • Hybridization refers to the association of two nucleic acid sequences to one another by hydrogen bonding. Factors that affect this bonding are discussed in Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual, Second Edition, Volume 2, chapter 9, pages 9.47 to 9.57.
  • a nucleic acid probe is said to “hybridize” with SEQ ID NO: 2 or SEQ ID NO: 3 if the probe can form a duplex or double stranded complex, which is stable enough to be detected.
  • Hybridization of the probe to a polynucleotide of SEQ ID NO: 2 or SEQ ID NO: 3 depends on (1) the sequence of the nucleic acid probe and (2) the hybridization conditions.
  • sequence of the probe need not be exactly complementary.
  • a non-complementary nucleotide sequence may be attached to the 5′ end of the probe, with the remainder of the probe sequence being complementary to SEQ ID NO: 2 or SEQ ID NO: 3.
  • non-complementary bases or longer sequences can be interspersed into the probe, provided that the probe sequence has sufficient complementarity with SEQ ID NO: 2 or SEQ ID NO: 3 to hybridize therewith and thereby form a duplex which can be detected.
  • the exact length and sequence of the probe will depend on the hybridization conditions, such as temperature, salt condition and the like.
  • the nucleic acid probe typically contains 15-25 or more nucleotides, although it may contain fewer nucleotides.
  • Short primers generally require cooler temperatures to form sufficiently stable hybrid complexes with the template.
  • Two sequences can be aligned using the methods and computer programs described above, and include BLAST, available over the world wide web at http://ww.ncbi.nlm.nih.gov/BLAST/.
  • Another alignment algorithm is Fasta, available in the Genetics Computing Group (GCG) package, Madison, Wis., USA, a wholly owned subsidiary of Oxford Molecular Group, Inc.
  • GCG Genetics Computing Group
  • Other techniques for alignment are described in Doolittle, Methods in Enzymology, Supra.
  • an alignment program that permits gaps in the sequence is utilized to align the sequences.
  • the Smith-Waterman is one type of algorithm that permits gaps in sequence alignments. See Meth. Mol. Biol. 70: 173-187 (1997).
  • the GAP program using the Needleman and Wunsch alignment method can be utilized to align sequences.
  • An alternative search strategy uses MPSRCH software, which runs on a MASPAR computer.
  • MPSRCH uses a Smith-Waterman algorithm to score sequences on a massively parallel computer. This approach improves ability to pick up distantly related matches, and is especially tolerant of small gaps and nucleotide sequence errors.
  • EST-encoded amino acid sequences can be used to search both protein and DNA databases.
  • the gap generation penalty will generally range from about 1 to about 5, usually, about 2 to about 4 and in many embodiments will be about 3.
  • the gap extension penalty will generally range from about 0.01 to 0.20 and in many instances will be 0.10.
  • the program has default parameters determined by the sequences inputted to be compared.
  • the sequence identity is determine used the parameters determined by the program. This program is available from Genetics Computing Group (GCG), Madison, Wis., USA.
  • sequence identity is: percentage of the alignment region length where the strongest alignment is found, percent sequence identity, and p value.
  • the percentage of the alignment region length is calculated by counting the number of residues of the individual sequence found in the region of strongest alignment. This number is divided by the total residue length of the query sequence to find a percentage.
  • Sequence A ASNPERTMIPVTRVGLIRYM
  • Sequence B YMMTEYLAIPV.RVGLPRYM 1 5 10 15
  • the region of alignment begins at amino acid 9 and ends at amino acid 19.
  • the total length of the query sequence is 20 amino acids.
  • the percent of the alignment region length is 11 divided by 20 or 55%.
  • Percent sequence identity is calculated by counting the number of amino acid matches between the two sequences and dividing total number of matches by the number of residues of the individual sequence found in the region of strongest alignment. For the example above, the percent identity would be 10 matches divided by 11 amino acids, or approximately, 90.9%
  • the percent of the alignment region length typically, is at least about 55% of total length one of the sequences; more typically, at least about 58%; even more typically; at least about 60% of the total residue length of one of the sequences.
  • percent length of the alignment region can be as much as about 62%; more usually, as much as about 64%; even more usually, as much as about 66%.
  • the percent length of alignment region is at least about 75% of the total length of one of the sequences; more typically, at least about 80%; even more typically, about 85%, about 90%, about 95%, about 98%, or 99%.
  • P value is the probability that the alignment was produced by chance.
  • the p value can be calculated according to Karlin et al., Proc. Natl. Acad. Sci. 87: 2264 (1990) and Karlin et al., Proc. Natl. Acad. Sci. 90: (1993).
  • the p value of multiple alignments using the same query sequence can be calculated using an heuristic approach described in Altschul et al., Nat. Genet. 6: 119 (1994). Alignment programs such as BLAST program can calculate the p value.
  • the two sequences typically, exhibit a p value is less than or equal to about 10 ⁇ 2 ; more usually; less than or equal to about 10 ⁇ 3 ; even more usually; less than or equal to about 10 ⁇ 4 . More typically, the p value is no more than about 10 ⁇ 5 ; more typically; no more than or equal to about 10 ⁇ 10 ; even more typically; no more than or equal to about 10 ⁇ 15 . Preferably, the p value is no more than about 10 ⁇ 20 ; more preferably, no more than about 10 ⁇ 30 ; even more preferably, no more than about 10 ⁇ 40 , about 10 ⁇ 50 , or 10 ⁇ 60 .
  • the boundaries of the region where the sequences align can be determined according to Doolittle, Methods in Enzymology, Supra; BLAST or FAST programs; or by determining the area where the sequence identity is highest.
  • Another factor to consider for determining identity or similarity is the location of the similarity or identity. Strong local alignment can indicate similarity even if the length of alignment is short. Sequence identity scattered throughout the length of the one sequence also can indicate a similarity between two sequences.
  • Stringency refers to conditions in a hybridization reaction that favor association of very similar sequences over sequences that differ. Factors affecting the stringency of hybridization are well known to those skilled in the art and are discussed in Sambrook et al. above at page 9.50.
  • stringent conditions will be those in which the salt concentration is less than about 1.5 M Na ion, typically about 0.01 to 1.0 M Na ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30° C. for short probes (e.g., 10 to 50 nucleotides) and at least about 60° C. for long probes (e.g., greater than 50 nucleotides).
  • Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide.
  • Exemplary moderate stringency conditions include hybridization in 40 to 45% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 0.5 ⁇ to 1 ⁇ SSC at 55 to 60° C.
  • Exemplary high stringency conditions include hybridization in 50% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 1 ⁇ SSC at 60 to 65° C.
  • a composition containing A is “substantially free of” B when at least about 85% by weight of the total A+B in the composition is A.
  • A comprises at least about 90% by weight of the total of A+B in the composition, more preferably at least about 95% or even 99% by weight.
  • heterologous refers to two biological components that are not found together in nature.
  • the components may be host cells, genes, or regulatory regions, such as promoters.
  • heterologous components are not found together in nature, they can function together, as when a promoter heterologous to a gene is operably linked to the gene.
  • Heterologous components may be from the same or different organisms. Another example is where a human CNGC-15 or Myosin IXa coding sequence is heterologous to a mouse host cell.
  • a “promoter” is a DNA sequence that initiates and regulates the transcription of a coding sequence when the promoter is operably linked to the coding sequence.
  • a promoter is “heterologous” to the coding sequence when the promoter is not operably linked to the coding sequence in nature.
  • a human CNGC-15 Myosin IXa promoter would comprise an expression cassette wherein a nucleotide sequence encoding CNGC-15 is operably linked to a heterologous promoter.
  • a “native” promoter is operably linked to the coding sequence in nature.
  • An “origin of replication” is a polynucleotide sequence that initiates and regulates replication of polynucleotides, such as an expression vector.
  • the origin of replication behaves as an autonomous unit of polynucleotide replication within a cell, capable of replication under its own control. With certain origins of replication, an expression vector can be reproduced at a high copy number in the presence of the appropriate proteins within the cell. Examples of origins are the 2 ⁇ and autonomously replicating sequences, which are effective in yeast; and the viral T-antigen, effective in COS-7 cells.
  • Host cells capable of producing CNGC-15 and/or Myosin IXa polypeptides are cultured “under conditions allowing expression.” Such conditions allow transcription and translation of the DNA molecule encoding the CNGC-15 and/or Myosin IXa polypeptide. These conditions include cultivation temperature, oxygen concentration, media composition, pH, etc. For example, if the trp promoter is utilized in the expression vector, the media will lack tryptophan to trigger the promoter and induce expression. The exact conditions will vary from host cell to host cell and from expression vector to expression vector.
  • antibody refers to a polypeptide or group of polypeptides composed of at least one antibody combining site.
  • An “antibody combining site” is the three-dimensional binding space with an internal surface shape and charge distribution complementary to the features of an epitope of an antigen, which allows a binding of the antibody with the antigen.
  • Antibody includes, for example, vertebrate antibodies, hybrid antibodies, chimeric antibodies, altered antibodies, univalent antibodies, the Fab proteins, and single domain antibodies.
  • Myosin IXa-modulated disorders and “CNGC-15 modulated-disorders” include, but are not limited to Bardet-Biedl Syndrome, Usher Syndrome, hearing loss, retinis pigmentosa, obesity, hypogonadism, sterility, polydactyly, brachydactyly, syndactyly, mental retardation, renal abnormalities, hypertension, diabetes and cardiovascular abnormalities
  • This invention provides the amino acid and nucleotide sequences of CNGC-15 and Myosin IXa. With these disclosed sequences, nucleic acid probe assays and expression cassettes and vectors for CNGC-15 and Myosin IXa polypeptides can be produced.
  • the expression vectors can be transformed into host cells to produce CNGC-15 and/or Myosin IXa polypeptides.
  • the purified polypeptides can be used to produce antibodies to detect CNGC-15 or Myosin IXa.
  • the host cells or extracts can be utilized for biological assays to isolate agonists or antagonists.
  • nucleic acid probe assays For example, PCR, branched DNA probe assays, or blotting techniques utilizing nucleic acid probes substantially identical or complementary to SEQ ID NO: 2 or SEQ ID NO: 3 can determine the presence of CNGC-15 or Myosin IXa cDNA or mRNA.
  • the nucleic acid probes will hybridize to a nucleotide sequence encoding the amino acid sequence shown in SEQ ID NO: 1 or encoded by SEQ ID NO: 3, or the complement of a sequence encoding SEQ ID NO: 1 or the polypeptide encoded by SEQ ID NO: 3.
  • SEQ ID NO: 2 and SEQ ID NO: 3 are preferred because they are the actual sequences present in human cells.
  • the nucleic acid probe will hybridize to the complement of a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 1 or encoding the polypeptide encoded by nucleotides 369-1307 of SEQ ID NO: 3 or to a complement of SEQ ID NO: 2 or SEQ ID NO: 3.
  • the nucleic acid probe will hybridize to SEQ ID NO: 2 or SEQ ID NO: 3 or to a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 1 or encoding the polypeptide encoded by SEQ ID NO: 3.
  • the nucleic acid probe sequences need not be identical to SEQ ID NO: 2, SEQ ID NO: 3 or their complement.
  • Probes are typically at least about 20 nucleotides, more preferably at least about 30 nucleotides.
  • the probes may be produced by synthetic procedures, such as the triester method of Matteucci et al. (1981) J. Am. Chem. Soc. 103:3185, or according to Urdea et al. (1983) Proc. Natl. Acad. Sci. USA 80:7461, or using commercially available automated oligonucleotide synthesizers.
  • One example of a nucleotide hybridization assay is described in Urdea et al. PCT WO92/02526 and Urdea et al. U.S. Pat. No. 5,124,246, herein incorporated by reference. Other methods of hybridization and detection are known to those skilled in the art.
  • PCR Polymerase Chain Reaction
  • mRNA, cDNA and genomic DNA can be detected by traditional blotting techniques described in Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual (New York, Cold Spring Harbor Laboratory).
  • CNGC-15 and Myosin IXa polypeptides are produced by recombinantly engineered host cells. These host cells are constructed by the introduction of a expression vector comprising a promoter operably linked to a CNGC-15 or Myosin IXa polypeptide coding sequence.
  • Such coding sequences can be constructed by synthesizing the entire gene or by altering existing CNGC-15 or Myosin IXa polypeptide coding sequences.
  • CNGC-15 and Myosin IXa polypeptides can be divided into four general categories: mutants, fragments, fusions, and the native CNGC-15 or Myosin IXa polypeptides.
  • the CNGC-15 polypeptides are those that occur in nature.
  • the amino acid sequence of such polypeptides may vary slightly from SEQ ID NO: 1.
  • the native CNGC-15 and Myosin IXa polypeptide coding sequence can be selected based on the amino acid sequence shown in SEQ ID NO: 1 or the amino acid sequence predicted by the open reading frame of SEQ ID NO: 3.
  • synthetic genes can be made using codons preferred by the host cell to encode the desired polypeptide.
  • codons preferred by the host cell See Urdea et al. (1983) Proc. Natl. Acad. Sci. USA 80:7461).
  • the desired native CNGC-15 and Myosin IXa polypeptide coding sequences can be cloned from nucleic acid libraries. Techniques for producing and probing nucleic acid sequence libraries are described, for example, in Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual (New York, Cold Spring Harbor Laboratory). Other recombinant techniques, such as site specific mutagenesis, PCR, enzymatic digestion and ligation, can also be used to construct the desired CNGC-15 polypeptide coding sequence.
  • the native CNGC-15 and Myosin IXa polypeptide coding sequences can be modified to create the other classes of CNGC-15 and Myosin IXa polypeptides.
  • mutants can be created by making conservative amino acid substitutions that maintain or enhance native CNGC-15 or Myosin IXa activity. The following are examples of conservative substitutions: Gly Ala; Val Ile Leu; Asp Glu; Lys Arg; Asn Gln; and Phe Trp Tyr.
  • Mutants can also contain amino acid deletions or insertions compared to the native CNGC-15 or Myosin IXa polypeptides. Mutants may include substitutions, insertions, and deletions of the native polypeptides.
  • Mutants will retain at least about 20% of the one of the activities of the native CNGC-15 or Myosin IXa.
  • the coding sequence of mutants can be constructed by in vitro mutagenesis of the native coding sequences.
  • mutants are a group of polypeptides with the non-disulfide bond participating cysteines substituted with a neutral amino acid, generally, with serines. These mutants may be stable over a broader temperature range than native CNGC-15 or Myosin IXa polypeptides. See, for example, Mark et al., U.S. Pat. No. 4,959,314.
  • Fragments differ from mutant or native CNGC-15 or Myosin IXa polypeptides by amino and/or carboxyl terminal amino acid deletions.
  • the number of amino acids that are truncated is not critical as long as the CNGC-15 or Myosin IXa fragment retains at least about 20% of the one of the activities of the native CNGC-15 or Myosin IXa polypeptide.
  • the coding sequence of such fragments can be easily constructed by cleaving the unwanted nucleotides from the mutant or native CNGC-15 or Myosin IXa polypeptide coding sequences.
  • Fusions are fragment, mutant, or native CNGC-15 or Myosin IXa polypeptides with additional amino acids at either or both of the termini.
  • the additional amino acid sequence is not necessarily homologous to sequence found in native polypeptides.
  • the fusions, just as all CNGC-15 and Myosin IXa polypeptides, retain at least about 20% of one of the activities of the native CNGC-15 or Myosin IXa polypeptides. Coding sequence of the fusions can be constructed by ligating synthetic polynucleotides encoding the additional amino acids to fragment, mutant, or native coding sequences. Activities of the CNGC-15 and Myosin IXa polypeptides can be determined by the methods described infra.
  • an expression vector will contain a promoter which is operable in the host cell and operably linked to a CNGC-15 or Myosin IXa coding sequence. Sequences that modulate gene expression, such as enhancers and binding sites for inducers or repressors may be present. Expression vectors may also include signal sequences, terminators, selectable markers, origins of replication, and sequences homologous to host cell sequences. These additional elements are optional but can be included to optimize expression.
  • Functional non-natural promoters may also be used, for example, synthetic promoters based on a consensus sequence of different promoters.
  • effective promoters can contain a regulatory region linked with a heterologous expression initiation region.
  • hybrid promoters are the E. coli lac operator linked to the E. coli tac transcription activation region; the yeast alcohol dehydrogenase (ADH) regulatory sequence linked to the yeast glyceraldehyde-3-phosphate-dehydrogenase (GAPDH) transcription activation region (U.S. Pat. Nos. 4,876,197 and 4,880,734, incorporated herein by reference); and the cytomegalovirus (CMV) enhancer linked to the SV40 (simian virus) promoter.
  • ADH yeast alcohol dehydrogenase
  • GPDH yeast glyceraldehyde-3-phosphate-dehydrogenase
  • a CNGC-15 or Myosin IXa polypeptide coding sequence may also be linked in reading frame to a signal sequence.
  • the signal sequence fragment typically encodes a peptide comprised of hydrophobic amino acids which directs the CNGC-15 or Myosin IXa polypeptide to the cell membrane or other subcellular compartment.
  • processing sites encoded between the leader fragment and the gene or fragment thereof that can be cleaved either in vivo or in vitro.
  • DNA encoding suitable signal sequences can be derived from genes for secreted endogenous host cell proteins, such as the yeast invertase gene (EP 12 873; JP 62,096,086), the A-factor gene (U.S. Pat. No. 4,588,684), interferon signal sequence (EP 60 057).
  • a preferred class of secretion leaders for yeast expression, are those that employ a fragment of the yeast alpha-factor gene, which contains both a “pre” signal sequence, and a “pro” region.
  • the types of alpha-factor fragments that can be employed include the full-length pre-pro alpha factor leader (about 83 amino acid residues) as well as truncated alpha-factor leaders (typically about 25 to about 50 amino acid residues) (U.S. Pat. Nos. 4,546,083 and 4,870,008, incorporated herein by reference; EP 324 274).
  • Additional leaders employing an alpha-factor leader fragment that provides for secretion include hybrid alpha-factor leaders made with a presequence of a first yeast signal sequence, but a pro-region from a second yeast alpha-factor. (See e.g., PCT WO 89/02463).
  • terminators are regulatory sequences, such as polyadenylation and transcription termination sequences, located 3′ or downstream of the stop codon of the coding sequences.
  • the terminator of native host cell proteins are operable when attached 3′ of a CNGC-15 or Myosin IXa polypeptide coding sequences. Examples are the Saccharomyces cerevisiae alpha-factor terminator and the baculovirus terminator.
  • viral terminators are also operable in certain host cells; for instance, the SV40 terminator is functional in CHO cells.
  • selectable markers an origin of replication, and homologous host cells sequences may optionally be included in an expression vector.
  • a selectable marker can be used to screen for host cells that potentially contain the expression vector. Such markers may render the host cell immune to drugs such as ampicillin, chloramphenicol, erythromycin, neomycin, and tetracycline.
  • markers may be biosynthetic genes, such as those in the histidine, tryptophan, and leucine pathways. Thus, when leucine is absent from the media, for example, only the cells with a biosynthetic gene in the leucine pathway will survive.
  • An origin of replication may be needed for the expression vector to replicate in the host cell. Certain origins of replication enable an expression vector to be reproduced at a high copy number in the presence of the appropriate proteins within the cell. Examples of origins are the 2 ⁇ and autonomously replicating sequences, which are effective in yeast; and the viral T-antigen, effective in COS-7 cells.
  • Expression vectors may be integrated into the host cell genome or remain autonomous within the cell. Polynucleotide sequences homologous to sequences within the host cell genome may be needed to integrate the expression cassette. The homologous sequences do not always need to be linked to the expression vector to be effective. For example, expression vectors can integrate into the CHO genome via an unattached dihydrofolate reductase gene. In yeast, it is more advantageous if the homologous sequences flank the expression cassette. Particularly useful homologous yeast genome sequences are those disclosed in PCT WO 90/01800, and the HIS4 gene sequences, described in Genbank, Accession No. J01331.
  • promoter, terminator, and other optional elements of an expression vector will also depend on the host cell chosen.
  • the invention is not dependent on the host cell selected. Convenience and the level of protein expression will dictate the optimal host cell.
  • a variety of hosts for expression are known in the art and available from the American Type Culture Collection (ATCC).
  • Bacterial hosts suitable for expressing a CNGC-15 and/or Myosin IXa polypeptide include, without limitation: Campylobacter, Bacillus, Escherichia, Lactobacillus, Pseudomonas, Staphylococcus, and Streptococcus.
  • Yeast hosts from the following genera may be utilized: Candida, Hansenula, Kluyveromyces, Pichia, Saccharomyces, Schizosaccharomyces, and Yarrowia.
  • Immortalized mammalian host cells include but are not limited to CHO cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), and other cell lines.
  • a number of insect cell hosts are also available for expression of heterologous proteins: Aedes aegypti, Bombyx mori, Drosophila melanogaster, and Spodoptera frugiperda (PCT WO 89/046699; Carbonell et al.
  • the desired CNGC-15 and/or Myosin IXa polypeptide expression vector is inserted into the host cell.
  • Methods of introducing exogenous DNA into bacterial hosts are well-known in the art, and typically protocol includes either treating the bacteria with CaCl 2 or other agents, such as divalent cations and DMSO.
  • DNA can also be introduced into bacterial cells by electroporation or viral infection. Transformation procedures usually vary with the bacterial species to be transformed. See e.g. (Masson et al. (1989) FEMS Microbiol. Lett. 60:273; Palva et al. (1982) Proc. Natl. Acad. Sci. USA 79:5582; EP Publ. Nos. 036 259 and 063 953; PCT WO 84/04541, Bacillus), (Miller et al. (1988) Proc. Natl.
  • Transformation methods for yeast hosts are well-known in the art, and typically include either the transformation of spheroplasts or of intact yeast cells treated with alkali cations. Electroporation is another means for transforming yeast hosts. See for example, Methods in Enzymology, Volume 194, 1991, “Guide to Yeast Genetics and Molecular Biology.” Transformation procedures usually vary with the yeast species to be transformed. See e.g. (Kurtz et al. (1986) Mol. Cell. Biol. 6:142; Kunze et al. (1985) J. Basic Microbiol. 25:141, Candida); (Gleeson et al. (1986) J. Gen. Microbiol. 132:3459; Roggenkamp et al.
  • Methods for introducing heterologous polynucleotides into mammalian cells include viral infection, dextran-mediated transfection, calcium phosphate precipitation, microparticle bombardment, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei.
  • a baculovirus vector is constructed in accordance with techniques that are known in the art, for example, as described in Kitts et al. (1993) Biotechniques 14:810-817; Smith et al. (1983) Mol. Cell. Biol. 3:2156; and Luckow and Summer (1989) Virol. 17:31.
  • a baculovirus expression vector is constructed substantially in accordance to Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987). Moreover, materials and methods for baculovirus/insect cell expression systems are commercially available in kit form, for example, the MaxBac kit from Invitrogen (San Diego, Calif.).
  • an insect cell can be infected with a virus containing a CNGC-15 or Myosin IXa polypeptide coding sequence.
  • the virus When the virus is replicating in the infected cell, the CNGC-15 or Myosin IXa polypeptide will be expressed if operably linked to a suitable promoter.
  • suitable insect cells and viruses include following without limitation.
  • Insect cells from any order of the Class Insecta can be used to express the polypeptide of this invention.
  • the orders Diptera and Lepidoptera are preferred.
  • Example of insect species are listed in Weiss et al. “Cell Culture Methods for Large-Scale Propagation of Baculoviruses,” in Granados et al. (eds., The Biology of Baculoviruses: Vol. II Practical Application for Insect Control, pp. 63-87 at p. 64 (1987).
  • Insect cell lines derived from the following insects are exemplary: Carpocapsa pomeonella (preferably, cell line CP-128); Trichoplusia ni (preferably, cell line TN-368); Autograph californica; Spodoptera frugiperda (preferably, cell line Sf9); Lymantria dispar; Mamestra brassicae; Aedes albopictus; Orgyia pseudotsugata; Neodiprio sertifer; Aedes aegypti; Antherae eucalypti; Gnorimoschema operceullela; Galleria mellonella; Spodoptera littolaris; Blatella germanic; Drosophila melanogaster; Heliothis zea; Spodoptera exigua; Rachiplusia ou; Plodia interpunctella, Amsaeta moorei; Agrotis c - nigrum, Adoxophyes orana; Agrotis se
  • Preferred insect cell lines are from Spodoptera frugiperda, and especially preferred is cell line Sf9.
  • the Sf9 cell line used in the examples herein was obtained from Max D. Summers (Texas A & M University, College Station, Tex., 77843, USA).
  • Other S. frugiperda cell lines, such as IPL-Sf-21AE III, are described in Vaughn et al. (1977) In Vitro 13:213-217.
  • the insect cell lines of this invention are suitable for the reproduction of numerous insect-pathogenic viruses such as parvoviruses, pox viruses, baculoviruses and rhabdoviruses, of which nucleopolyhedrosis viruses (NPV) and granulosis viruses (GV) from the group of baculoviruses are preferred. Further preferred are NPV viruses such as those from Autographa spp., Spodoptera spp., Trichoplusia spp., Rachiplusia spp., Gallerai spp., and Lymantria spp.
  • NPV nucleopolyhedrosis viruses
  • GV granulosis viruses
  • baculovirus strain Autographa californica NPV AcNPV
  • Rachiplusia ou NPV Galleria mellonella NPV
  • any plaque purified strains of AcNPV such as E2, R9, S1, M3, characterized and described by Smith et al. (1978) Virol. 89:517-527.
  • insect cells Spodoptera frugiperda type 9 are infected with baculovirus strain Autographa californica NPV (AcNPV) containing a CNGC-15 or Myosin IXa polypeptide coding sequence.
  • baculovirus strain Autographa californica NPV AcNPV
  • a baculovirus is produced by homologous recombination between a transfer vector containing the coding sequence and baculovirus sequences and a genomic baculovirus DNA.
  • the genomic baculovirus DNA is linearized and contains a dysfunctional essential gene.
  • the transfer vector preferably, contains the nucleotide sequences needed to restore the dysfunctional gene and a baculovirus polyhedrin promoter and terminator operably linked to the CNGC-15 or Myosin IXa coding sequence. (See Kitts et al. (1993) Biotechniques 14(5):810-817.
  • the transfer vector and linearized baculovirus genome are transfected into SF9 insect cells, and the resulting viruses probably containing the desired coding sequence. Without a functional essential gene the baculovirus genome cannot produce a viable virus. Thus, the viable viruses from the transfection most likely contain the CNGC-15 or Myosin IXa polypeptide coding sequence and the needed essential gene sequences from the transfer vector. Further, lack of occlusion bodies in the infected cells are another verification that the CNGC-15 or Myosin IXa polypeptide coding sequence was incorporated into the baculovirus genome.
  • the essential gene and the polyhedrin gene flank each other in the baculovirus genome.
  • the coding sequence in the transfer vector is flanked at its 5′ with the essential gene sequences and the polyhedrin promoter and at its 3′ with the polyhedrin terminator.
  • the CNGC-15 or Myosin IXa polypeptide coding sequence displaces the baculovirus polyhedrin gene.
  • Such baculoviruses without a polyhedrin gene will not produce occlusion bodies in the infected cells.
  • another means for determining if coding sequence was incorporated into the baculovirus genome is to sequence the recombinant baculovirus genomic DNA.
  • expression of the desired CNGC-15 or Myosin IXa polypeptide by cells infected with the recombinant baculovirus is another verification means.
  • Immunoassays and ligand binding assays can be utilized to confirm that the transformed host cell is expressing the desired CNGC-15 and/or Myosin IXa polypeptide.
  • an immunofluorescence assay can be performed on transformed host cells without separating the CNGC-15 or Myosin IXa polypeptides from the cell.
  • the host cells are first fixed onto a solid support, such as a microscope slide or microtiter well. This fixing step permeabilizes the cell membrane.
  • the fixed host cells are exposed to an anti-CNGC-15 or anti-Myosin IXa polypeptide antibody.
  • the fixed cells are exposed to a second antibody, which is labeled and binds to the anti-CNGC-15 or anti-Myosin IXa polypeptide antibody.
  • the secondary antibody is labeled with an fluorescent marker.
  • the host cells which express the CNGC-15 or Myosin IXa polypeptides will be fluorescently labeled and easily visualized under the microscope. See, for example, Hashido et al. (1992) Biochem & Biophys. Res. Comm. 187(3):1241-1248.
  • the CNGC-15 and Myosin IXa polypeptides do not need to be separated from the cell membrane for in vitro assays.
  • the host cells may be fixed to a solid support, such as a microtiter plate.
  • a crude membrane fraction can be separated from lysed host cells by centrifugation (See Adachi et al. (1992) FEBS Lett 311(2):179-183.
  • the fixed host cells or the crude membrane fraction is exposed to labeled ligand or ion.
  • the ligand is labeled with radioactive atoms.
  • the host cells which express the desired CNGC-15 or Myosin IXa polypeptide will bind with the labeled ligand which can be easily detected.
  • the purified CNGC-15 and Myosin IXa polypeptides are useful as compositions, for assays, and to produce antibodies.
  • CNGC-15 and Myosin IXa polypeptides can be isolated by a variety of steps including, for example, anion exchange chromatography, size exclusion chromatography, hydroxyapatite chromatography, hydrophobic interaction chromatography, metal chelation chromatography, reverse phase HPLC, affinity chromatography, and further ammonium sulfate precipitations. These techniques are well known to those of skill in the art.
  • the crude cell membrane fractions can be utilized. These membrane extracts can be isolated from cells which expressed CNGC-15 or Myosin IXa polypeptides by lysing the cells. Alternatively, whole cells, expressing CNGC-15 and/or Myosin IXa polypeptides, can be cultured in a microtiter plate.
  • Antibodies against CNGC-15 and Myosin IXa polypeptides are useful for affinity chromatography, immunofluorescent assays, and distinguishing CNGC-15 and Myosin IXa polypeptides.
  • Antibodies to the proteins of the invention may be prepared by conventional methods known to those skilled in the art.
  • monoclonal antibodies are prepared using the method of Kohler et al. (1975) Nature 256:495-496, or a modification thereof.
  • the antibodies may be labeled using conventional techniques. Suitable labels include fluorophores, chromophores, radioactive atoms (particularly 32 P and 125 I), electron-dense reagents, enzymes, and ligands having specific binding partners. Enzymes are typically detected by their activity. For example, horseradish peroxidase is usually detected by its ability to convert 3,3′,5,5′-tetra-methylbenzidine (TMB) to a blue pigment, quantifiable with a spectrophotometer.
  • TMB 3,3′,5,5′-tetra-methylbenzidine
  • Specific binding partner refers to a protein capable of binding a ligand molecule with high specificity, as for example in the case of an antigen and a monoclonal antibody specific therefor.
  • Other specific binding partners include biotin and avidin or streptavidin, IgG and protein A, and the numerous receptor-ligand couples known in the art. It should be understood that the above description is not meant to categorize the various labels into distinct classes, as the same label may serve in several different modes. For example, 125 I may serve as a radioactive label or as an electron-dense reagent. HRP may serve as enzyme or as antigen for a MAb. Further, one may combine various labels for desired effect.
  • MAbs and avidin also require labels in the practice of this invention: thus, one might label a MAb with biotin, and detect its presence with avidin labeled with 125 I, or with an anti-biotin MAb labeled with HRP.
  • CNGC-15 and Myosin IXa polypeptides can also be used to screen combinatorial libraries to identify agonist or antagonists.
  • a “library” of peptides may be synthesized following the methods disclosed in U.S. Pat. No. 5,010,175, and in PCT WO 91/17823, both incorporated herein by reference in full.
  • the peptide library is first screened for binding to the selected CNGC-15 or Myosin IXa polypeptide.
  • the peptides are then tested for their ability to inhibit or enhance CNGC-15 or Myosin IXa activity. Peptides exhibiting the desired activity are then isolated and sequenced.
  • CNGC-15 and Myosin IXa agonists or antagonists may be screened using any available method.
  • the assay conditions ideally should resemble the conditions under which the CNGC-15 or Myosin IXa activity is exhibited in vivo, i.e., under physiologic pH, temperature, ionic strength, etc.
  • Suitable agonists or antagonists will exhibit strong inhibition or enhancement of the CNGC-15 or Myosin IXa activity at concentrations which do not raise toxic side effects in the subject.
  • Agonists or antagonists which compete for binding to the CNGC-15 or Myosin IXa polypeptide may require concentrations equal to or greater than the native CNGC-15 or Myosin IXa concentration, while inhibitors capable of binding irreversibly to the polypeptide may be added in concentrations on the order of the native CNGC-15 or Myosin IXa concentration.
  • the non-lipophilic form of fura-2 will fluoresce when it binds to the free Ca 2+ ions, which are released after binding of a ligand to the receptor.
  • the fluorescence can be measured without lysing the cells at an excitation spectrum of 340 nm and at fluorescence spectrum of 500 nm. See Sakurai et al. EP 480 381 and Adachi et al. (1992) FEBS Lett 311 (2):179-183 for examples of assays measuring free intracellular Ca 2+ concentrations.
  • 3 H-inositol is added to the media of host cells expressing CNGC-15 or Myosin IXa polypeptides.
  • the 3 H-inositol taken up by the cells and after stimulation of the cells with endothelin or endothelin agonist, the resulting inositol triphosphate is separated from the mono and di-phosphate forms and measured. See Sakurai et al., EP 480 381.
  • Amersham provides an inosital 1,4,5-trisphosphate assay system.
  • Amersham provides tritylated inosital 1,4,5-trisphosphate and a receptor capable of distinguishing the radioactive inositol from other inositol phosphates. With these reagents an effective and accurate competition assay can be performed to determine the inositol triphosphate levels.
  • compositions can comprise either polypeptides, antibodies, or polynucleotides of the claimed invention.
  • the pharmaceutical compositions will comprise a therapeutically effective amount of either polypeptides, antibodies, or polynucleotides of the claimed invention.
  • terapéuticaally effective amount refers to an amount of a therapeutic agent to treat, ameliorate, or prevent a desired disease or condition, or to exhibit a detectable therapeutic or preventative effect.
  • the effect can be detected by, for example, chemical markers or antigen levels.
  • Therapeutic effects also include reduction in physical symptoms, such as decreased body temperature.
  • the precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition, and the therapeutics or combination of therapeutics selected for administration. Thus, it is not useful to specify an exact effective amount in advance. However, the effective amount for a given situation can be determined by routine experimentation and is within the judgment of the clinician.
  • an effective dose will be from about 0.01 mg/kg to 50 mg/kg or 0.05 mg/kg to about 10 mg/kg of the polypeptide or DNA construct in the individual to which it is administered
  • a pharmaceutical composition can also contain a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier refers to a carrier for administration of a therapeutic agent, such as antibodies or a polypeptide, genes, and other therapeutic agents.
  • the term refers to any pharmaceutical carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity.
  • Suitable carriers may be large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles. Such carriers are well known to those of ordinary skill in the art.
  • salts can be used therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like
  • organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • compositions may contain liquids such as water, saline, glycerol and ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
  • the therapeutic compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared. Liposomes are included within the definition of a pharmaceutically acceptable carrier.
  • compositions of the invention can be administered directly to the subject.
  • the subjects to be treated can be mammals or birds. In particular, human subjects can be treated.
  • compositions will generally be accomplished by injection, either subcutaneously, intraperitoneally, intravenously or intramuscularly or delivered to the interstitial space of a tissue.
  • the compositions can also be administered into a tumor or lesion.
  • Other modes of administration include oral and pulmonary administration, suppositories, and transdermal applications, needles, and gene guns or hyposprays.
  • Dosage treatment may be a single dose schedule or a multiple dose schedule.
  • the Myosin IXa or CNGC-15 polypeptides could be stably expressed in an organ of a mammal, and then the organ could be xenografted into a human in need of such treatment.
  • Gene therapy vehicles for delivery of constructs including a coding sequence of a therapeutic of the invention, to be delivered to the mammal for expression in the mammal can be administered either locally or systemically.
  • constructs can utilize viral or non-viral vector approaches in in vivo or ex vivo modality. Expression of such coding sequence can be induced using endogenous mammalian or heterologous promoters. Expression of the coding sequence in vivo can be either constitutive or regulated.
  • the invention includes gene delivery vehicles capable of expressing the contemplated nucleic acid sequences.
  • the gene delivery vehicle is preferably a viral vector and, more preferably, a retroviral, adenoviral, adeno-associated viral (AAV), herpes viral, or alphavirus vector.
  • the viral vector can also be an astrovirus, coronavirus, orthomyxovirus, papovavirus, paramyxovirus, parvovirus, picomavirus, poxvirus, or togavirus viral vector. See generally, Jolly et al. (1994) Cancer Gene Therapy 1:51-64; Kimura et al. (1994) Human Gene Therapy 5:845-852; Connelly et al. (1995) Human Gene Therapy 6:185-193; and Kaplitt et al. (1994) Nature Genetics 6:148-153.
  • Retroviral vectors are well known in the art and we contemplate that any retroviral gene therapy vector is employable in the invention, including B, C and D type retroviruses, xenotropic retroviruses (for example, NZB-X1, NZB-X2 and NZB9-1 (see O'Neill (1985) J. Vir. 53:160) polytropic retroviruses (for example, MCF and MCF-MLV (see Kelly et al. (1983) J. Vir. 45:291), spumaviruses and lentiviruses. See RNA Tumor Viruses, Second Edition, Cold Spring Harbor Laboratory, 1985.
  • xenotropic retroviruses for example, NZB-X1, NZB-X2 and NZB9-1 (see O'Neill (1985) J. Vir. 53:160
  • polytropic retroviruses for example, MCF and MCF-MLV (see Kelly et al. (1983) J. Vir. 45:291)
  • retroviral gene therapy vector may be derived from different retroviruses.
  • retrovector LTRs may be derived from a Murine Sarcoma Virus, a tRNA binding site from a Rous Sarcoma Virus, a packaging signal from a Murine Leukemia Virus, and an origin of second strand synthesis from an Avian Leukosis Virus.
  • Retroviral vectors may be used to generate transduction competent retroviral vector particles by introducing them into appropriate packaging cell lines (see U.S. Ser. No. 07/800,921, filed Nov. 29, 1991). Retrovirus vectors can be constructed for site-specific integration into host cell DNA by incorporation of a chimeric integrase enzyme into the retroviral particle. See, U.S. Ser. No. 08/445,466 filed May 22, 1995. It is preferable but not required that the recombinant viral vector is a replication defective recombinant virus.
  • Packaging cell lines suitable for use with the above-described retrovirus vectors are well known in the art, are readily prepared (see U.S. Ser. No. 08/240,030, filed May 9, 1994; see also WO 92/05266), and can be used to create producer cell lines (also termed vector cell lines or “V CLs”) for the production of recombinant vector particles.
  • the packaging cell lines are made from human parent cells (e.g., HT1080cells) or mink parent cell lines, which eliminates inactivation in human serum.
  • Preferred retroviruses for the construction of retroviral gene therapy vectors include Avian Leukosis Virus, Bovine Leukemia, Virus, Murine Leukemia Virus, Mink-Cell Focus-Inducing Virus, Murine Sarcoma Virus, Reticuloendotheliosis Virus and Rous Sarcoma Virus.
  • Particularly preferred Murine Leukemia Viruses include 4070A and 1504A (Hartley et al. (1976) J. Virol. 19:19-25), Abelson (ATCC No. VR-999), Friend (ATCC No. VR-245), Graffi, Gross (ATCC Nol VR-590), Kirsten, Harvey Sarcoma Virus and Rauscher (ATCC No.
  • Retroviruses may be obtained from depositories or collections such as the American Type Culture Collection (“ATCC”) in Rockville, Md. or isolated from known sources using commonly available techniques.
  • ATCC American Type Culture Collection
  • Exemplary known retroviral gene therapy vectors employable in this invention include those described in GB 2200651, EP 0415731, EP 0345242, WO 89/02468, WO 89/05349, WO 89/09271, WO 90/02806, WO 90/07936, WO 94/03622, WO 93/25698, WO 93/25234, WO 93/11230, WO 93/10218, WO 91/02805, in U.S. Pat. Nos. 5,219,740, 4,405,712, 4,861,719, 4,980,289 and 4,777,127, in U.S. Ser. No. 07/800,921 and in Vile et al.
  • Human adenoviral gene therapy vectors are also known in the art and employable in this invention. See, for example, Berkner et al. (1988) Biotechniques 6:616; Rosenfeld et al. (199 1) Science 252:431; and WO 93/07283, WO 93/06223, and WO 93/07282.
  • Exemplary known adenoviral gene therapy vectors employable in this invention include those described in the above referenced documents and in WO 94/12649, WO 93/03769, WO 93/19191, WO 94/28938, WO 95/11984, WO 95/00655, WO 95/27071, WO 95/29993, WO 95/34671, WO 96/05320, WO 94/08026, WO 94/11506, WO 93/06223, WO 94/24299, WO 95/14102, WO 95/24297, WO 95/02697, WO 94/28152, WO 94/24299, WO 95/09241, WO 95/25807, WO 95/05835, WO 94/18922, WO 95/09654.
  • administration of DNA linked to killed adenovirus as described in Curiel et al. (1992) Hum. Gene Ther. 3:147-154 may be employed.
  • the gene delivery vehicles of the invention also include adenovirus associated virus (AAV) vectors.
  • AAV adenovirus associated virus
  • Leading and preferred examples of such vectors for use in this invention are the AAV-2 based vectors disclosed in Srivastava, WO 93/09239.
  • Most preferred AAV vectors comprise the two AAV inverted terminal repeats in which the native D-sequences are modified by substitution of nucleotides, such that at least about 5 native nucleotides and up to 18 native nucleotides, preferably at least about 10 native nucleotides up to 18 native nucleotides, most preferably 10 native nucleotides are retained and the remaining nucleotides of the D-sequence are deleted or replaced with non-native nucleotides.
  • the native D-sequences of the AAV inverted terminal repeats are sequences of 20 consecutive nucleotides in each AAV inverted terminal repeat (i.e., there is one sequence at each end) which are not involved in HP formation.
  • the non-native replacement nucleotide may be any nucleotide other than the nucleotide found in the native D-sequence in the same position.
  • Other employable exemplary AAV vectors are pWP-19, pWN-1, both of which are disclosed in Nahreini et al. (1993) Gene 124:257-262.
  • Another example of such an AAV vector is psub201. See Samulski et al. (1987) J. Virol. 61:3096.
  • Double-D ITR vector is the Double-D ITR vector. Construction of the Double D ITR vector is disclosed in U.S. Pat. No. 5,478,745. Still other vectors are those disclosed in Carter, U.S. Pat. No. 4,797,368 and Muzyczka, U.S. Pat. No. 5,139,941, Chartejee, U.S. Pat. No. 5,474,935, and Kotin, PCT Patent Publication WO 94/288157. Yet a further example of an AAV vector employable in this invention is SSV9AFABTKneo, which contains the AFP enhancer and albumin promoter and directs expression predominantly in the liver. Its structure and construction are disclosed in SU, (1996) Human Gene Therapy 7:463-470. Additional AAV gene therapy vectors are described in U.S. Pat. Nos. 5,354,678, 5,173,414, 5,139,941, and 5,252,479.
  • the gene therapy vectors of the invention also include herpes vectors.
  • Leading and preferred examples are herpes simplex virus vectors containing a sequence encoding a thymidine kinase polypeptide such as those disclosed in U.S. Pat. No. 5,288,641 and EP 0176170 (Roizman).
  • Additional exemplary herpes simplex virus vectors include HFEM/ICP6-LacZ disclosed in WO 95/04139 (Wistar Institute), pHSVlac described in Geller et al.
  • alpha virus gene therapy vectors that can be employed in this invention.
  • Preferred alpha virus vectors are Sindbis viruses vectors. Togaviruses, Semliki Forest virus (ATCC VR-67; ATCC VR-1247), Middleberg virus (ATCC VR-370), Ross River virus (ATCC VR-373; ATCC VR-1246), Venezuelan equine encephalitis virus (ATCC VR923; ATCC VR-1250; ATCC VR-1249; ATCC VR-532), and those described in U.S. Pat. Nos. 5,091,309, 5,217,879, and WO 92/10578, WO 95/07994, U.S. Pat. Nos.
  • alpha viruses may be obtained from depositories or collections such as the ATCC in Rockville, Md. or isolated from known sources using commonly available techniques. Preferably, alphavirus vectors with reduced cytotoxicity are used (see co-owned U.S. Ser. No. 08/679640).
  • DNA vector systems such as eukaryotic layered expression systems are also useful for expressing the nucleic acids of the invention. See WO 95/07994 for a detailed description of eukaryotic layered expression systems.
  • the eukaryotic layered expression systems of the invention are derived from alphavirus vectors and most preferably from Sindbis viral vectors.
  • viral vectors suitable for use in the present invention include those derived from poliovirus, for example ATCC VR-58 and those described in Evans et al. (1989) Nature 339:385 and Sabin et al. (1973) J. Biol. Standardization 1:115; rhinovirus, for example ATCC VR-1110 and those described in Arnold et al. (1990) J. Cell Biochem L401; pox viruses such as canary pox virus or vaccinia virus, for example ATCC VR-111 and ATCC VR-2010 and those described in Fisher-Hoch et al. (1989) Proc. Natl. Acad. Sci. 86:317; Flexner et al. (1989) Ann. NY Acad. Sci.
  • measles virus for example ATCC VR-67 and VR-1247 and those described in EP 0440219; Aura virus, for example ATCC VR-368; Bebaru virus, for example ATCC VR-600 and ATCC VR-1240; Cabassou virus, for example ATCC VR-922; Chikungunya virus, for example ATCC VR-64 and ATCC VR-1241; Fort Morgan Virus, for example ATCC VR-924; Getah virus, for example ATCC VR-369 and ATCC VR-1243; Kyzylagach virus, for example ATCC VR-927; Mayaro virus, for example ATCC VR-66; Mucambo virus, for example ATCC VR-580 and ATCC VR-1244; Ndumu virus, for example ATCC VR-371; Pixuna virus, for example ATCC VR-372 and ATCC VR-1245; Tonate virus, for example ATCC VR-925; Triniti virus, for example ATCC VR-469; Una virus, for example ATCC VR-374; Whataroa
  • compositions of this invention into cells is not limited to the above mentioned viral vectors.
  • Other delivery methods and media may be employed such as, for example, nucleic acid expression vectors, polycationic condensed DNA linked or unlinked to killed adenovirus alone, for example see U.S. Ser. No. 08/366,787, filed Dec. 30, 1994 and Curiel et al. (1989) Hum. Gene Ther. 3:147-154; ligand linked DNA, for example see Wu et al. (1989) J. Biol. Chem. 264:16985-16987, eucaryotic cell delivery vehicles cells, for example see U.S. Ser. No. 08/240,030, filed May 9, 1994, and U.S. Ser. No.
  • Particle mediated gene transfer may be employed, for example see U.S. Serial No. 60/023,867. Briefly, the sequence can be inserted into conventional vectors that contain conventional control sequences for high level expression, and then incubated with synthetic gene transfer molecules such as polymeric DNA-binding cations like polylysine, protamine, and albumin, linked to cell targeting ligands such as asialoorosomucoid, as described in Wu et al. (1987) J. Biol. Chem. 262:4429-4432, insulin as described in Hucked, (1990) Biochem. Pharmacol. 40:253-263, galactose as described in Plank et al. (1992) Bioconjugate Chem. 3:533-539, lactose or transferrin.
  • synthetic gene transfer molecules such as polymeric DNA-binding cations like polylysine, protamine, and albumin, linked to cell targeting ligands such as asialoorosomucoid, as described in Wu
  • Naked DNA may also be employed.
  • Exemplary naked DNA introduction methods are described in WO 90/11092 and U.S. Pat. No. 5,580,859. Uptake efficiency may be improved using biodegradable latex beads. DNA coated latex beads are efficiently transported into cells after endocytosis initiation by the beads. The method may be improved further by treatment of the beads to increase hydrophobicity and thereby facilitate disruption of the endosome and release of the DNA into the cytoplasm.
  • Liposomes that can act as gene delivery vehicles are described in U.S. Pat. No. 5,422,120, WO 95/13796, WO 94/23697, WO 91/144445 and EP 524,968. As described in co-owned U.S. Serial No.
  • the nucleic acid sequences encoding a polypeptide can be inserted into conventional vectors that contain conventional control sequences for high level expression, and then be incubated with synthetic gene transfer molecules such as polymeric DNA-binding cations like polylysine, protamine, and albumin, linked to cell targeting ligands such as asialoorosomucoid, insulin, galactose, lactose, or transferrin.
  • synthetic gene transfer molecules such as polymeric DNA-binding cations like polylysine, protamine, and albumin, linked to cell targeting ligands such as asialoorosomucoid, insulin, galactose, lactose, or transferrin.
  • Other delivery systems include the use of liposomes to encapsulate DNA comprising the gene under the control of a variety of tissue-specific or ubiquitously-active promoters.
  • Further non-viral delivery suitable for use includes mechanical delivery systems such as the approach described in Woffendin et al.
  • the coding sequence and the product of expression of such can be delivered through deposition of photopolymerized hydrogel materials.
  • Other conventional methods for gene delivery that can be used for delivery of the coding sequence include, for example, use of hand-held gene transfer particle gun, as described in U.S. Pat. No. 5,149,655; use of ionizing radiation for activating transferred gene, as described in U.S. Pat. No. 5,206,152 and WO 92/11033.
  • Exemplary liposome and polycationic gene delivery vehicles are those described in U.S. Pat. Nos. 5,422,120 and 4,762,915; in WO 95/13796; WO 94/23697; and WO 91/14445; in EP 0524968; and in Stryer et al. (1975) Biochemistry, pp. 236-240, W. H. Freeman, San Francisco; Szoka et al. (1980) Biochim. Biophys. Acta. 600:1; Bayer, (1979) Biochim. Biophys. Acta. 550:464; Rivnay et al. (1987) Meth. Enzymol. 149:119; Wang et al. (1987) Proc. Natl. Acad. Sci. 84:7851; Plant (1989) Anal. Biochem. 176:420.
  • polypeptides which include, without limitation: asioloorosomucoid (ASOR); transferrin, asialoglycoproteins; antibodies; antibody fragments; ferritin; interleukins; interferons, granulocyte, macrophage colony stimulating factor (M-CSF), stem cell factor and erythropoietin.
  • ASOR asioloorosomucoid
  • transferrin asialoglycoproteins
  • antibodies antibody fragments
  • ferritin ferritin
  • interleukins interleukins
  • interferons granulocyte, macrophage colony stimulating factor (M-CSF), stem cell factor and erythropoietin.
  • M-CSF macrophage colony stimulating factor
  • stem cell factor erythropoietin.
  • RII proteins from other invasive organisms, such as the 17 amino acid peptide from the circumsporozoite protein of plasmodium falciparum known as RII.
  • Other groups that can be included are, for example: hormones, steroids, androgens, estrogens, thyroid hormone, or vitamins, folic acid.
  • polyalkylene glycol can be included with the desired polynucleotides.
  • the polyalkylene glycol is polyethylene glycol.
  • mono-, di-, or polysaccarides can be included.
  • the polysaccharide is dextran or DEAE-dextran.
  • the desired polynucleotide can also be encapsulated in lipids or packaged in liposomes prior to delivery to the subject or to cells derived therefrom.
  • Lipid encapsulation is generally accomplished using liposomes which are able to stably bind or entrap and retain nucleic acid.
  • the ratio of condensed polynucleotide to lipid preparation can vary but will generally be around 1:1 (mg DNA:micromoles lipid), or more of lipid.
  • liposomes as carriers for delivery of nucleic acids, see, Hug et al. (1991) Biochim. Biophys. Acta. 1097:1-17; Straubinger et al. (1983) Methods of Enzymology 101:512-527.
  • Liposomal preparations for use in the instant invention include cationic (positively charged), anionic (negatively charged) and neutral preparations.
  • Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Felgner et al. (1987) Proc. Natl. Acad. Sci. U.S.A. 84:7413-7416); mRNA (Malone et al. (1989) Proc. Natl. Acad. Sci. U.S.A. 86:6077-6081); and purified transcription factors (Debs et al. 1990) J. Biol. Chem. 265:10189-10192), in functional form.
  • Cationic liposomes are readily available.
  • N[1-2,3-dioleyloxy)propyl]-N,N,N-triethylammonium (DOTMA) liposomes are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, N.Y. (See, also, Felgner et al. (1987) Proc. Natl. Acad. Sci. U.S.A. 84:7413-7416).
  • Other commercially available liposomes include transfectace (DDAB/DOPE) and DOTAP/DOPE (Boerhinger).
  • Other cationic liposomes can be prepared from readily available materials using techniques well known in the art.
  • anionic and neutral liposomes are readily available, such as from Avanti Polar Lipids (Birmingham, Ala.), or can be easily prepared using readily available materials.
  • Such materials include phosphatidyl choline, cholesterol, phosphatidyl ethanolamine, dioleoyfphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), dioleoylphoshatidyl ethanolamine (DOPE), among others. These materials can also be mixed with the DOTMA and DOTAP starting materials in appropriate ratios. Methods for making liposomes using these materials are well known in the art.
  • the liposomes can comprise multilammelar vesicles (MLVs), small unilamellar vesicles (SUVs), or large unilamellar vesicles (LUVs).
  • MLVs multilammelar vesicles
  • SUVs small unilamellar vesicles
  • LUVs large unilamellar vesicles
  • the various liposome-nucleic acid complexes are prepared using methods known in the art. See, e.g., Straubinger et al. (1983) Methods of Immunology 101:512-527; Szoka et al. (1978) Proc. Natl. Acad Sci. U.S.A. 75:4194-4198; Papahadjopoulos et al. (1975) Biochim. Biophys. Acta. 394:483; Wilson et al.
  • lipoproteins can be included with the polynucleotide to be delivered.
  • lipoproteins to be utilized include: chylomicrons, HDL, IDL, LDL, and VLDL. Mutants, fragments, or fusions of these proteins can also be used. Also, modifications of naturally occurring lipoproteins can be used, such as acetylated LDL. These lipoproteins can target the delivery of polynucleotides to cells expressing lipoprotein receptors. Preferably, if lipoproteins are included with the polynucleotide to be delivered, no other targeting ligand is included in the composition.
  • Naturally occurring lipoproteins comprises of a lipid and a protein portion.
  • the protein portion is known as apoproteins.
  • apoproteins A, B, C, D, and E have been isolated and identified. At least two of these contain several proteins, designated by Roman numerals, AI, AII, AIV, CI, CII, and CIII.
  • a lipoprotein can comprise more than one apoprotein.
  • naturally occurring chylomicrons comprises of A, B, C, and E; over time these lipoproteins lose A and acquire C and E apoproteins.
  • VLDL comprises A, B, C, and E apoproteins;
  • LDL comprises apoprotein B;
  • HDL comprises apoproteins A, C, and E.
  • Lipoproteins contain a variety of lipids including triglycerides, cholesterol (free and esters), and phospholipids.
  • the composition of the lipids varies in naturally occurring lipoproteins.
  • chylomicrons comprise mainly triglycerides.
  • the composition of the lipids are chosen to aid in conformation of the apoprotein for receptor binding activity.
  • the composition of lipids can also be chosen to facilitate hydrophobic interaction and association with the polynucleotide binding molecule.
  • Naturally occurring lipoproteins can be isolated from serum by ultracentrifugation, for instance. Such methods are described in Meth. Enzy., Supra; Pitas et al. (1980) J. Biochem. 255:5454-5460; and Mahey et al. (1979) J. Clin. Invest. 64:743-750.
  • Lipoproteins can also be produced by in vitro or recombinant methods by expression of the apoprotein genes in a desired host cell. See, for example, Atkinson et al. (1986) Annu. Rev. Biophys. Chem. 15:403 and Radding et al. (1958) Biochim. Biophys. Acta. 30:443.
  • Lipoproteins can also be purchased from commercial suppliers, such as Biomedical Technologies, Inc., Stoughton, Mass., U.S.A.
  • Polycationic agents can be included, with or without lipoprotein, in a composition with the desired polynucleotide to be delivered.
  • Polycationic agents typically exhibit a net positive charge at physiological relevant pH and are capable of neutralizing the electrical charge of nucleic acids to facilitate delivery to a desired location. These agents have in vitro, ex vivo, and in vivo applications. Polycationic agents can be used to deliver nucleic acids to a living subject either intramuscularly, subcutaneously, etc.
  • polypeptides as polycationic agents: polylysine, polyarginine, polyomithine, and protamine.
  • Other examples include histones, protamines, human serum albumin, DNA binding proteins, non-histone chromosomal proteins, coat proteins from DNA viruses, such as ⁇ X174, transcriptional factors also contain domains that bind DNA and therefore may be useful as nucleic acid condensing agents.
  • transcriptional factors such as C/CEBP, c-jun, c-fos, AP-1, AP-2, AP-3, CPF, Prot-1, Sp-1, Oct-1, Oct-2, CREP, and TFIID contain basic domains that bind DNA sequences.
  • Organic polycationic agents include: spermine, spermidine, and putrescine.
  • polycationic agent can be extrapolated from the list above to construct other polypeptide polycationic agents or to produce synthetic polycationic agents.
  • Synthetic polycationic agents which are useful include, for example, DEAE-dextran, polybrene.
  • Lipofectin, and lipofectAMINE are monomers that form polycationic complexes when combined with polynucleotides. See for example, Zuckermann et al., PCT Appln. U.S. Ser. No. 97/14465.
  • a polynucleotide composition can comprises therapeutically effective amount of a gene therapy vehicle, as the term is defined above.
  • an effective dose will be from about 0.01 mg/kg to 50 mg/kg or 0.05 mg/kg to about 10 mg/kg of the DNA constructs in the individual to which it is administered.
  • the polynucleotide compositions of the invention can be administered (1) directly to the subject; (2) delivered ex vivo, to cells derived from the subject; or (3) in vitro for expression of recombinant proteins.
  • the subjects to be treated can be mammals or birds. Also, human subjects can be treated.
  • compositions will generally be accomplished by injection, either subcutaneously, intraperitoneally, intravenously or intramuscularly or delivered to the interstitial space of a tissue.
  • the compositions can also be administered into a tumor or lesion.
  • Other modes of administration include oral and pulmonary administration, suppositories, and transdermal applications, needles, and gene guns or hyposprays.
  • Dosage treatment may be a single dose schedule or a multiple dose schedule.
  • Methods for the ex vivo delivery and reimplantation of transformed cells into a subject are known in the art and described in e.g., International Publication No. WO 93/14778.
  • Examples of cells useful in ex vivo applications include, for example, stem cells, particularly hematopoietic, lymph cells, macrophages, dendritic cells, or tumor cells.
  • nucleic acids for both ex vivo and in vitro applications can be accomplished by the following procedures, for example, dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei, all well known in the art.
  • Fragments in the 1-2 kb range were isolated and cloned into the vector Perfectly BluntTM (Novagen). For each BAC, 192 clones were randomly selected and sequenced using the ABI TAQ dye chemistry/system.
  • PCR primers were designed for MarathonTM 5′ and 3′ RACETM reactions using MarathonTM prepared leukocyte, retina and testis cDNA (Clontech) as templates. Resulting fragments were cloned into pCRII (Invitrogen), sequenced and used for primer design for subsequent MarathonTM reactions. Nucleotide sequence for both strands of the partial cDNAs was obtained using the ABI TAQ dye terminator chemistry/system. Sequence comparison analysis was done using MASPAR and GeneWorksTM (Intelligenetics).
  • a sequence representing a transcript of 8473 bp was obtained.
  • Human myosin IXa was mapped on the Stanford G3 radiation hybrid panel using oligos having homology to myosin IXa cDNA. Duplicate reactions consistently linked this region of myosin IXa to marker SHGC-31014 located on chromosome 15 in bin #50 confirming that the myosin IXa gene is located on human chromosome 15q21.
  • An ATG is present at base 243 and is in good context having an A at the ⁇ 3 position and a G at +4 (Kozak et al. (1995)). This ATG initiates a single ORF that continues for 7643 bases to a stop signal (UGA) present at base 7886. Sequence analysis and exon mapping of the myo IXa transcript indicated that myosin IXa contains 42 exons and predicts a protein of 2548 amino acids with an estimated molecular mass of 283 kDa (FIG. 1), a size similar to the myosin IXb gene product (Wirth et al. (1996)).
  • the myosin IXa gene spans over 185 kb in the human genome (data not shown) and produces a transcript whose predicted protein contains the domains described for class IX unconventional myosins.
  • the predicted amino acid sequence of human myosin IXa, myosin IXb, and rat myr5 were compared.
  • the N-terminal extension of myosin IXa shares 40% identity with the other class IX myosins.
  • Myosin IXa had 82% identity with myosin IXb and myr5 within the head domain that is conserved among known myosin classes, although myosin IXa also contained an 18 residue insertion absent in the other class IX family members.
  • myosin IXa contained two separate insertions of 33 and 18 residues compared to the other class IX family members. These insertions conform to the “IQ motif” and suggest that myosin IXa may bind 6 light chains of the clamodulin/EF hand superfamily (Mercer et al. 1991) instead of the 4 predicted to bind to myosin IXb and myr5.
  • the tail region of myosin IXa has 58% identity with the GAP domain found in myr5 and myosin IXb. However, the tail region located between the neck region and the GAP domain has numerous insertions not found in the other class IX family members that add an additional 327 residues compared to myr5 or 286 residues compared to myosin IXb.
  • a transcript of ⁇ 12.5 kb was visible in all tissues tested except liver. In addition, a transcript of ⁇ 8.5 kb was present in testis and placenta.
  • MarathonTM cDNA (Clontech) from testis, whole brain and cerebellum was amplified using a primer specific to the last exon (to bases 7704-7727) and the MarathonTM adaptor primer AP1. This PCR reaction produced a single band in all cases which corresponded to the product predicted from the cloned sequence.
  • two brain cDNA libraries frontal cortex and cerebellum (Stratagene) were screened using the GAP domain probe. No evidence was found for either alternative splicing or polyadenylation at the 3′ end of the myosin IXa transcript. Alternative splicing within and/or between the remaining identified exons was ruled out by conducting PCR amplification of whole brain, cerebellum, retina and testis cDNA with primers that spanned intron/exon junctions. No differences in product sizes were seen between those tissues expressing only the 12.5 kb transcript and testis. This result was confirmed by conducting multiple hybridizations to a frontal cortex cDNA library using probes representing each portion of the myosin IXA transcript.
  • the 4 kb difference between the two transcript sizes present in testis was not due to alternative splicing between the identified exons nor due to alternative polyadenylation and most likely resides in the 5′ untranslated region of the myosin IXa transcript. Therefore, the additional 4 kb present in the larger transcript is likely located in the 5′ UTR.
  • At least one alternative splicing event does occur within the myosin IXa coding region.
  • two distinct bands were generated from PCR reactions conducted on leukocyte cDNA with primers specific to the head region. Sequence analysis of these bands indicated that a cryptic splice site exists at bases 375-380 and acts as a splice donor site, creating a truncated exon #2. Identical PCR reactions conducted on cDNA from testis and retina produced only the larger product.
  • a fragment specific to the mouse N-terminal extension of myosin IXa was obtained by conducting a PCR reaction on mouse genomic DNA using primers corresponding to bases 252.271 and bases 608-590 of the human myosin IXa sequence. This probe was prepared as described above and hybridized to mouse poly A + Northern blots prepared using standard methods from freshly dissected tissues. A single transcript of ⁇ 9 kb was detected.
  • Myosin IXa was first visible in RNA from 11.5 d embryos, absent from 13.5 d embryos and reappeared in 15.5 d embryos. Myosin IXa was also present in RNA obtained from 13.5 d old limb buds and brain.
  • Myo IXa Functions as a GTPase Activating Protein
  • Myr5 acts as a GTP activating protein on members of the rho subfamily of GTP binding proteins (Reinhard et al. (1995) EMBO J. 14:697-704). We determined that myosin IXa is also a functional GAP by making an expression construct that contained the predicted GAP domain (bases 6141 to 8035 of SEQ ID NO: 2) and conducting GAP assays with the small GTP binding proteins rhoA, ras, rac and Cdc42.
  • a partial CNGC-15 cDNA, 1281 nucleotides in length was isolated from a human BAC contigs as described in Example 1.
  • the cDNA sequence is shown in FIG. 3.
  • the ATG codon at nucleotide 36 g initiates an open reading frame that contiues through the rest of the cDNA.
  • Comparison of the nucleotide to Genbank, EST and patent databases showed that this partial cDNA has 76.2% homology to a mouse ion channel BCGN-3 mRNA (Genbank accession number AF064874) and lesser homology to mouse and human ion channels. The longest stretch of 100% homology with sequences in the databases was 44 nucleotides.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention discloses the amino acid and nucleic acid sequences of a new CNGC and Myosin that map to the region of the human chromosome associated with Bardet-Biedl Syndrome. Cyclic nucleotide gated channels (CNGCs) comprise a family of multimeric protein ion channels that open in response to the binding of a cyclic nucleotide to an intracellular domain. The two new proteins, CNGC-15 and Myosin IXa, are useful in the study, diagnosis and treatment of Bardet-Biedl Syndrome and Usher Syndrome. Other indications that can be treated by CNGC-15 and/or Myosin IXa polypeptides, or agonists or antagonists include hearing loss, retinis pigmentosa, obesity, hypogonadism, sterility, polydactyly, brachydactyly, syndactyly, mental retardation, renal abnormalities, hypertension, diabetes and cardiovascular abnormalities.
Compositions and methods for expressing cyclic nucleotide gated channel-15 (CNGC-15) and Myosin IXa are provided. The compositions comprise CNGC-15 and Myosin IXa polypeptides and derivatives thereof, nucleotide sequences, expression cassettes, transformed cells and antibodies to these polypeptides. Methods for the expression and detection of CNGC-15 and Myosin IXa nucleotides and polypeptides and compositions for the treatment of these conditions are provided.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application Serial No. 60/062,858, filed Oct. 15, 1997, U.S. Provisional Application No. 60/062,241, filed Oct. 17, 1997, and U.S. Provisional Application No. 60/068,953, filed Dec. 30, 1997, the contents of which are herein incorporated by reference.[0001]
  • FIELD OF THE INVENTION
  • This invention relates to the fields of molecular biology and pharmaceutical research. More specifically, this invention relates to the identification and recombinant expression of two new genes, cyclic nucleotide gated channel-15 (CNGC-15) and Myosin IXa. Accordingly, isolation of the human myosin IXa gene will prove useful in the study, diagnosis and treatment of Bardet-Biedl Syndrome, Usher Syndrome and related conditions. [0002]
  • BACKGROUND OF THE INVENTION
  • Usher Sydrome typel (USH1) is characterized by a profound congenital sensoneural hearing loss, vestibular dysfunction and prepubescent onset retinitis pigmentosa. Family studies indicated that three genes with different chromosomal locations are responsible for USH1; a defect in any one of these genes causes the disease. Of these, the gene USH1B mapped to a region homologous to the murine region containing the mouse deafness mutant shaker-1 which results from mutations in myosin VIIa. Subsequent work confirmed that myosin VIIa is the cause of Usher Sydrome type 1B and localized the myosin VIIa protein to the receptor cells of the inner ear (Hasson et al. (1995) [0003] Genomics 36:431-439) and the connecting cilia of photoreceptor cells in the retina (Liu et al. (1997) Cell Motil. Cytoskel. 37:240-252).
  • Retinitis pigmentosa is also associated with Bardet-Biedl Syndrome. Bardet-Biedl Syndrome is further characterized by obesity, retinal degeneration, syndactyly and/or polydactyly, hypogenitalism and mental retardation (Schachat et al. (1982) [0004] Arch. Ophthalmol. 100:285-288; Green et al. (1989) N. Engl. J. Med. 321:1002-1009) and can result from aberrations in one of at least four different genes. Other publications relating to myosin IXa include Bruford et al. (1997) Genomics 41:93-99 and Bement et al. (1994) Proc. Natl. Acad. Sc. USA 91:6549-6553.
  • All references cited herein are incorporated by reference. [0005]
  • SUMMARY OF THE INVENTION
  • The present invention discloses the amino acid and nucleic acid sequences of a new Cyclic nucleotide gated channel (CNGC) and Myosin that map to the region of the human chromosome associated with Bardet-Biedl Syndrome. CNGCs comprise a family of multimeric protein ion channels that open in response to the binding of a cyclic nucleotide to an intracellular domain. The two new proteins, CNGC-15 and Myosin IXa, are useful in the study, diagnosis and treatment of Bardet-Biedl Syndrome and Usher Syndrome. Other indications that can be treated by CNGC-15 and/or Myosin IXa polypeptides, or agonists or antagonists include hearing loss, retinis pigmentosa, obesity, hypogonadism, sterility, polydactyly, brachydactyly, syndactyly, mental retardation, renal abnormalities, hypertension, diabetes and cardiovascular abnormalities. [0006]
  • Compositions and methods for expressing cyclic nucleotide gated channel-15 (CNGC-15) and Myosin IXa are provided. The compositions comprise CNGC-15 and Myosin IXa polypeptides and derivatives thereof, nucleotide sequences, expression cassettes, transformed cells and antibodies to these polypeptides. Methods for the expression and detection of CNGC-15 and Myosin IXa nucleotides and polypeptides and compositions for the treatment of these conditions are provided.[0007]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 sets forth the amino acid sequence of the human Myosin IXa polypeptide (SEQ ID NO: 1). [0008]
  • FIG. 2 sets forth the nucleotide sequence of the human Myosin IXa cDNA (SEQ ID NO: 2). The ATG translational intiation codon at position 243 and the TGA translational stop codon at position 7887 are underlined. [0009]
  • FIG. 3 sets forth the nucleotide sequence of a human CNGC-15 partial cDNA (SEQ ID NO: 3).[0010]
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is drawn to compositions and methods useful in the treatment of Bardet-Biedl Syndrome, Usher Syndrome, hearing loss, retinis pigmentosa, obesity, hypogonadism, sterility, polydactyly, brachydactyly, syndactyly, mental retardation, renal abnormalities, hypertension, diabetes and cardiovascular abnormalities. More particularly, two new polypeptides and the genes encoding them, CNGC-15 and Myosin IXa, have been identified that are useful in the treatment of these and a variety of other conditions. The human Myosin IXa polypeptide and cDNA are shown in FIGS. 1 and 2, as SEQ ID NO: 1 and SEQ ID NO: 2, respectively. The human CNGC-15 partial cDNA is shown in FIG. 3 as SEQ ID NO: 3. [0011]
  • Thus, it is an object of the invention to provide native CNGC-15 and Myosin IXa substantially free of other human proteins. [0012]
  • Another object of the invention is to provide polypeptides that are mutants, fragments and derivatives of CNGC-15 or Myosin Ixa. Regions of particular importance of Myosin IXa include: the open reading frame (nucleotides 243-7886 of SEQ ID NO: 2) encoding a 2548 amino acid protein (SEQ ID NO: 1); the N-terminal extension (residues 1-146 of SEQ ID NO: 1) encoded by nucleotides 243-680 of SEQ ID NO: 2; the head domain (residues 147-719 of SEQ ID NO: 1) encoded by nucleotides 683-2399 of SEQ ID NO: 2; the ATP binding loop (residues 239-246 of SEQ ID NO: 1) encoded by nucleotides 959-977 of SEQ ID NO: 2; the minimal length 2nd insert (residues 720-835 of SEQ ID NO: 1) encoded by nucleotides 2402-2747 of SEQ ID NO: 2; the light chain binding domain (residues 972-1166 of SEQ ID NO: 1) encoded by nucleotides 3158-3740 of SEQ ID NO: 2; and the GAP domain (residues 2074-2219) of SEQ ID NO: 1 encoded by nucleotides 6473-6899 of SEQ ID NO: 2. [0013]
  • Accordingly, the invention provides a Myosin IXa polypeptide comprising an amino acid sequence selected from the group consisting of: [0014]
  • a) SEQ ID NO: 1; [0015]
  • b) residues 1-146 of SEQ ID NO: 1; [0016]
  • c) residues 147-719 of SEQ ID NO: 1; [0017]
  • d) residues 239-246 of SEQ ID NO: 1; [0018]
  • e) residues 720-835 of SEQ ID NO: 1; [0019]
  • f) residues 972-1166 of SEQ ID NO: 1; [0020]
  • g) residues 2074-2219 of SEQ ID NO: 1; [0021]
  • h) a polypeptide having at least about 61% homology to SEQ ID NO: 1; [0022]
  • i) a polypeptide having at least about 95% homology to an amino acid sequence of b), c), d), e), f) or g); and [0023]
  • j) a polypeptide of at least about 90 contiguous residues of SEQ ID NO: 1. [0024]
  • The Myosin IXa polypeptides of the invention will have at least about 61% homology to SEQ ID NO:1, preferably at least about 70% homology, more preferably at least about 80% homology and most preferably at least about 90% homology. The polypeptides of b), c), d), e), f) or above, will have at least about 95% homology to SEQ ID NO:1, more preferably at least about 96% homology, even more preferably at least about 97% homology and most preferably at least about 98% homology. [0025]
  • Similarly, the invention provides a CNGC-15 polypeptide comprising an amino acid sequence selected from the group consisting of: [0026]
  • a) a polypeptide encoded by SEQ ID NO: 3; [0027]
  • b) a polypeptide encoded by nucleotides 369-1307 of SEQ ID NO: 3; [0028]
  • c) a polypeptide having at least about 80% homology to the polypeptide encoded by nucleotides 369-1307 of SEQ ID NO: 3; and [0029]
  • d) a polypeptide of at least about 20 contiguous residues of the polypeptide encoded by nucleotides 369-1307 of SEQ ID NO: 3. [0030]
  • The CNGC-15 polypeptides of the invention will have at least about 70% homology to a), b), c) or d) above, preferably at least about 80% homology, more preferably at least about 85% homology and most preferably at least about 90% homology. [0031]
  • In another aspect of the invention provides a chimeric polypeptide comprising a CNGC-15 polypeptide, or fragment thereof and a polypeptide of interest. Similarly, the invention provides a chimeric polypeptide comprising a Myosin IXa polypeptide, or fragment thereof, fused to a polypeptide of interest. Nucleotide sequences encoding chimeric CNGC-15 and Myosin IXa polypeptides are also provided. [0032]
  • Yet another object of the invention is to provide polynucleotides that encode the mutants, fragments, and derivatives, as well as the native CNGC-15 and Myosin IXa. Accordingly, the invention provides an isolated nucleotide sequence selected from the group consisting of: [0033]
  • a) a nucleotide sequence encoding a native Myosin IXa polypeptide; [0034]
  • b) a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 1; [0035]
  • c) the nucleotide sequence of SEQ ID NO: 2; [0036]
  • d) a nucleotide sequence comprising at least about 390 contiguous bases of SEQ ID NO: 2; [0037]
  • e) a nucleotide sequence comprising nucleotides 243-7085 of SEQ ID NO: 2; [0038]
  • f) a nucleotide sequence comprising nucleotides 243-680 of SEQ ID NO: 2; [0039]
  • g) a nucleotide sequence comprising nucleotides 683-2399 of SEQ ID NO: 2; [0040]
  • h) a nucleotide sequence comprising nucleotides 959-977 of SEQ ID NO: 2; [0041]
  • i) a nucleotide sequence comprising nucleotides 2402-2747 of SEQ ID NO: 2; [0042]
  • j) a nucleotide sequence comprising nucleotides 3158-3740 of SEQ ID NO: 2; and [0043]
  • k) a nucleotide sequence comprising nucleotides 6473-6899 of SEQ ID NO: 2. [0044]
  • The invention similarly provides an isolated nucleotide sequence selected from the group consisting of: [0045]
  • a) a nucleotide sequence encoding a native CNGC-15 polypeptide; [0046]
  • b) a nucleotide sequence encoding the polypeptide encoded by nucleotides 369-1307 of SEQ ID NO: 3; [0047]
  • c) the nucleotide sequence of SEQ ID NO: 3; [0048]
  • d) a nucleotide sequence comprising at least 45 contiguous nucleotides of the sequence of SEQ ID NO:3; and [0049]
  • e) a nucleotide sequence that hybridizes under stringent conditions to a denatured DNA having a nucleotide sequence of a), b), c) or d). [0050]
  • These polynucleotides can be operably linked to heterologous promoters to form expression cassettes. The expression cassettes can be introduced into suitable host cells for expression of CNGC-15 and/or Myosin IXa polypeptides and derivatives thereof. [0051]
  • Another object of the invention is to provide a transformed cell transiently expressing or having stably incorporated into its genome an expression vector comprising a promoter operably linked to a nucleotide sequence encoding a CNGC-15 or Myosin IXa polypeptide, or a fragment, derivative mutant or fusion thereof. [0052]
  • A further object of the invention is to provide a method for producing a CNGC-15 or Myosin IXa polypeptide or a derivative thereof, comprising: [0053]
  • a) providing a host cell transiently or stably transformed with an expression vector comprising a promoter operably linked to a nucleotide sequence encoding said CNGC-15 or Myosin IXa polypeptide or derivative thereof; and [0054]
  • b) culturing the host cell under conditions which allow expression of said polypeptide. [0055]
  • Another object of the invention is to provide a method for detecting CNGC-15 polynucleotides. The method comprises: [0056]
  • a) providing a nucleic acid probe which hybridizes to a nucleotide encoding a CNGC-15 polypeptide or a mutant, fragment or derivative thereof; [0057]
  • b) contacting the probe with a sample of polynucleotides under hybridizing conditions to form a duplex; and [0058]
  • c) detecting said duplexes. [0059]
  • A further object of the invention is to provide a method for detecting unconventional Myosin polynucleotides. The method comprises: [0060]
  • a) providing a nucleic acid probe which hybridizes to a nucleotide encoding a Myosin IXa or a mutant, fragment or derivative thereof; [0061]
  • b) contacting the probe with a sample of polynucleotides under hybridizing conditions to form a duplex; and [0062]
  • c) detecting said duplexes. [0063]
  • Yet another object of the invention is to provide antibodies to the CNGC-15 and Myosin IXa polypeptides. [0064]
  • Another object of the invention is to provide a method for detecting CNGC-15 polypeptides. The method comprises: [0065]
  • a) providing an antibody that binds to a CNGC-15 polypeptide; [0066]
  • b) contacting the antibody to a sample under binding conditions to form a duplex; and [0067]
  • c) detecting said duplexes. [0068]
  • Still another object of the invention is to provide a method for detecting Myosin IXa polypeptides. The method comprises: [0069]
  • a) providing an antibody that binds to a Myosin IXa polypeptide; [0070]
  • b) contacting the antibody to a sample under binding conditions to form a duplex; and [0071]
  • c) detecting said duplexes. [0072]
  • The invention further provides a method for treating Bardet-Biedl Syndrome, Usher Syndrome, hearing loss, retinis pigmentosa, obesity, hypogonadism, sterility, polydactyly, brachydactyly, syndactyly, mental retardation, renal abnormalities, hypertension, diabetes and cardiovascular abnormalities, comprising administering a therapeutically effective amount of a CNGC-15 or Myosin IXa polypeptide, or derivative thereof to a subject in need of such treatment. In still another aspect, the invention provides a composition comprising CNGC-15 or Myosin IXa or an active derivative thereof, and a pharmaceutically acceptable carrier. [0073]
  • Modes for Carrying Out the Invention [0074]
  • A. Definitions [0075]
  • As used herein, the terms “native CNGC-15” and “native Myosin IXa” refer to the polypeptides found in nature. for example, native CNGC-15 and native Myosin IXa are polypeptides with substantial amino acid sequence identity to SEQ ID NO: 1 and SEQ ID NO: 3, respectively. This definition includes allelic variants and other naturally occurring modifications of SEQ ID NO: 1 and SEQ ID NO: 3. [0076]
  • “CNGC-15 polypeptides” and “Myosin IXa polypeptides” include mutants, fragments, derivatives and fusions as well as the native CNGC-15 or Myosin IXa. [0077]
  • “Mutants” of the native CNGC-15 or Myosin IXa are polypeptides having an amino acid sequence which retain at least about 50% amino acid sequence identity with SEQ ID NO: 1 or SEQ ID NO: 3, respectively; more typically, at least about 60%; even more typically, at least about 80%. Preferably mutants will retain at least about 85% amino acid sequence identity with SEQ ID NO: 1 or SEQ ID NO: 3; more preferably, at least about 90%; even more preferably, at least about 95%. These differences may be conservative amino acid substitutions, insertions or deletions in the amino acid sequence. [0078]
  • “Fragments” possess the same amino acid sequence of the native or mutant CNGC-15 or Myosin IXa polypeptides except the fragments lack the amino and/or carboxyl terminal sequences of the native or mutant polypeptide. [0079]
  • “Derivatives” possess the same amino acid sequence of the native, mutant or fragment CNGC-15 and Myosin IXa, but may contain amino acid substitutes, glycosylated residues or other chemical modifications. [0080]
  • “Fusions” or “chimeric polypeptides” are mutants, fragments, or the native CNGC-15 or Myosin IXa that also include amino and/or carboxyl terminal amino acid extensions. For example, Myosin Ixa fragments may be fused with fragments of other unconventional myosins. Unconventional myosins are members of the myosin superfamily that display the general head, neck and tail domain structure of conventional myosins (Mooseker et al. (1995) [0081] Annu. Rev. Cell. Dev. Biol. 11:633-75), but differ from conventional myosins by changes in these and/or the inclusion of additional functional domains. The head region shared among all myosin molecules consists of conserved amino acid sequences that span approximately 80 kDa while the neck region contains conserved motifs that bind one to six light chains of protein from the calmodulin/EF-hand family (Bahler et al. (1996) Curr. Op. Cell. Biol. 8:18-22). It is the unconventional myosin tail domains that show the most variation from conventional myosin and from each other. For example, although myosin Va has a coiled coil tail domain, as does conventional myosin, it also contains a PEST site (required for calpain cleavage) and a globular C-terminal domain which may be used for interactions with cargo or docking proteins. Myosin VIIa also has a coiled coil tail domain, however it contains additional domains different from those found in myosin Va, such as the talin domain which is thought to be active in binding plasma membranes. As a third example, the class IX myosins (composed of rat myr5 and human myosin IXb) have even less similarity to conventional myosin and to the other nine classes of unconventional myosins. These molecules lack a coiled coil domain in the tail region, but do contain a GAP (GTP activating protein) domain which increases the GTPase activity of rhoA and Cdc42 (Reinhard et al. (1995) EMBO J. 14:697-704). Class IX myosins also have two large insertions in the conserved head domain that are absent in conventional and other unconventional myosins, and may act as regulatory domains affecting the mechanochemical properties of the protein. Despite the variation among these three examples of unconventional myosin classes, all classes bind actin and have either been shown to be or are postulated to act as molecular motors (Wang et al. (1996) Science 273:660-663), thereby interacting with the actin cytoskeleton. The characterization and chromosomal mapping of myosin Va and myosin VIIa suggested that these molecules may be responsible for specific human diseases. Indeed, defects in myosin Va are associated with Griscelli disease (Pastural et al. (1997) Nature Genet. 16:289-292), while mutations in myosin VIIa are responsible for Usher Sydrome type1B (Weil et al. (1995) Nature 374:60-61.
  • The number or type of the amino acid substitutions is not critical, nor is the length or number of the amino acid deletions, or amino acid extensions that are incorporated in the CNGC-15 or Myosin IXa polypeptides. However, all of these polypeptides will exhibit at least about 20% of one of the activities of the native CNGC-15 or Myosin IXa. More typically, the polypeptides exhibit at least about 40%, even more typically the polypeptides exhibit at least about 60% of one of the native CNGC-15 or Myosin IXa activities. All these polypeptides will retain at least about 50% amino acid identity with SEQ ID NO: 1 or SEQ ID NO: 3; more typically, at least about 60%; even more typically, at least about 80%. Preferably, these polypeptides will retain at least about 85% amino acid sequence identity with SEQ ID NO: 1 or the amino acid sequence encoded by SEQ ID NO: 3; more preferably, at least about 90%; even more preferably, at least about 95%. [0082]
  • Myosin IXa activities include immunological activities, ATP binding, ATPase activity (Shyamata et al. (1990) J Cell Biol 110:1137-1147), zinc binding (Reinhard et al. (1995) EMBO J 14:697-704; Quest et. (1994) J Biol Chem 269:2961-2970) calmodulin/EF binding (Glenney et al. (1980) J Biol Chem 255:10551-10554; Cheney, (1992) Current Opin Cell Biol 4:27-35), binding to G-coupled receptors, GTPase activity (Reinhard, Supra; Ridley (1992) Cell 70:401-410), membrane binding (Reizes (1994) PNAS USA 91:6439-6443) phorbolester binding (Reinhard, Supra), modulation of cell to cell interactions, calcium release and cytoskeletal rearrangements, actin binding (Ruppert et al. (1993) J Cell Biol 120:1393-1403; Ridley et al. (1992) Cell 70:389-399; Ridley et al. (1992) Cell 70:401-410), formation of focal complexes (Ridley, Supra), synthesis of phosphatidylinositol-4,5-bis-phosphate, activation of transcription and DNA synthesis and transduction of chemical energy into mechanical force along actin filaments (Collins et al. (1990) J Cell Biol 110:1137-1147). [0083]
  • “Hybridization” refers to the association of two nucleic acid sequences to one another by hydrogen bonding. Factors that affect this bonding are discussed in Sambrook et al. (1989) [0084] Molecular Cloning: A Laboratory Manual, Second Edition, Volume 2, chapter 9, pages 9.47 to 9.57. A nucleic acid probe is said to “hybridize” with SEQ ID NO: 2 or SEQ ID NO: 3 if the probe can form a duplex or double stranded complex, which is stable enough to be detected. Hybridization of the probe to a polynucleotide of SEQ ID NO: 2 or SEQ ID NO: 3 depends on (1) the sequence of the nucleic acid probe and (2) the hybridization conditions. The sequence of the probe need not be exactly complementary. For example, a non-complementary nucleotide sequence may be attached to the 5′ end of the probe, with the remainder of the probe sequence being complementary to SEQ ID NO: 2 or SEQ ID NO: 3. Alternatively, non-complementary bases or longer sequences can be interspersed into the probe, provided that the probe sequence has sufficient complementarity with SEQ ID NO: 2 or SEQ ID NO: 3 to hybridize therewith and thereby form a duplex which can be detected. The exact length and sequence of the probe will depend on the hybridization conditions, such as temperature, salt condition and the like. For example, for diagnostic applications, depending on the complexity of the analyte sequence, the nucleic acid probe typically contains 15-25 or more nucleotides, although it may contain fewer nucleotides. Short primers generally require cooler temperatures to form sufficiently stable hybrid complexes with the template.
  • Two sequences can be aligned using the methods and computer programs described above, and include BLAST, available over the world wide web at http://ww.ncbi.nlm.nih.gov/BLAST/. Another alignment algorithm is Fasta, available in the Genetics Computing Group (GCG) package, Madison, Wis., USA, a wholly owned subsidiary of Oxford Molecular Group, Inc. Other techniques for alignment are described in Doolittle, Methods in Enzymology, Supra. Preferably, an alignment program that permits gaps in the sequence is utilized to align the sequences. The Smith-Waterman is one type of algorithm that permits gaps in sequence alignments. See [0085] Meth. Mol. Biol. 70: 173-187 (1997). Also, the GAP program using the Needleman and Wunsch alignment method can be utilized to align sequences. An alternative search strategy uses MPSRCH software, which runs on a MASPAR computer. MPSRCH uses a Smith-Waterman algorithm to score sequences on a massively parallel computer. This approach improves ability to pick up distantly related matches, and is especially tolerant of small gaps and nucleotide sequence errors. EST-encoded amino acid sequences can be used to search both protein and DNA databases.
  • Of interest is the BestFit program using the local homology algorithm of Smith Waterman (Advances in Applied Mathematics 2: 482-289 (1981)) to determine sequence identity. The gap generation penalty will generally range from about 1 to about 5, usually, about 2 to about 4 and in many embodiments will be about 3. The gap extension penalty will generally range from about 0.01 to 0.20 and in many instances will be 0.10. The program has default parameters determined by the sequences inputted to be compared. Preferably, the sequence identity is determine used the parameters determined by the program. This program is available from Genetics Computing Group (GCG), Madison, Wis., USA. [0086]
  • Additional factors used to determine sequence identity are: percentage of the alignment region length where the strongest alignment is found, percent sequence identity, and p value. [0087]
  • The percentage of the alignment region length is calculated by counting the number of residues of the individual sequence found in the region of strongest alignment. This number is divided by the total residue length of the query sequence to find a percentage. An example is shown below: [0088]
    Sequence A: ASNPERTMIPVTRVGLIRYM
         | ||| |||| |||
    Sequence B: YMMTEYLAIPV.RVGLPRYM
       1  5  10 15
  • The region of alignment begins at amino acid 9 and ends at amino acid 19. The total length of the query sequence is 20 amino acids. The percent of the alignment region length is 11 divided by 20 or 55%. [0089]
  • Percent sequence identity is calculated by counting the number of amino acid matches between the two sequences and dividing total number of matches by the number of residues of the individual sequence found in the region of strongest alignment. For the example above, the percent identity would be 10 matches divided by 11 amino acids, or approximately, 90.9% [0090]
  • For the alignment results to be considered high similarity, the percent of the alignment region length, typically, is at least about 55% of total length one of the sequences; more typically, at least about 58%; even more typically; at least about 60% of the total residue length of one of the sequences. Usually, percent length of the alignment region can be as much as about 62%; more usually, as much as about 64%; even more usually, as much as about 66%. Typically, the percent length of alignment region is at least about 75% of the total length of one of the sequences; more typically, at least about 80%; even more typically, about 85%, about 90%, about 95%, about 98%, or 99%. [0091]
  • P value is the probability that the alignment was produced by chance. For a single alignment, the p value can be calculated according to Karlin et al., [0092] Proc. Natl. Acad. Sci. 87: 2264 (1990) and Karlin et al., Proc. Natl. Acad. Sci. 90: (1993). The p value of multiple alignments using the same query sequence can be calculated using an heuristic approach described in Altschul et al., Nat. Genet. 6: 119 (1994). Alignment programs such as BLAST program can calculate the p value.
  • The two sequences, typically, exhibit a p value is less than or equal to about 10[0093] −2; more usually; less than or equal to about 10−3; even more usually; less than or equal to about 10−4. More typically, the p value is no more than about 10−5; more typically; no more than or equal to about 10−10; even more typically; no more than or equal to about 10−15. Preferably, the p value is no more than about 10−20; more preferably, no more than about 10−30; even more preferably, no more than about 10−40, about 10−50, or 10−60.
  • The boundaries of the region where the sequences align can be determined according to Doolittle, Methods in Enzymology, Supra; BLAST or FAST programs; or by determining the area where the sequence identity is highest. [0094]
  • Another factor to consider for determining identity or similarity is the location of the similarity or identity. Strong local alignment can indicate similarity even if the length of alignment is short. Sequence identity scattered throughout the length of the one sequence also can indicate a similarity between two sequences. [0095]
  • “Stringency” refers to conditions in a hybridization reaction that favor association of very similar sequences over sequences that differ. Factors affecting the stringency of hybridization are well known to those skilled in the art and are discussed in Sambrook et al. above at page 9.50. [0096]
  • In general, convenient hybridization temperatures in the presence of 50% formamide are 42° C. for a probe with is 95% to 100% homologous to the target fragment, 37° C. for 90% to 95% homology, and 32° C. for 85% to 90% homology. For lower homologies, formamide content should be lowered and temperature adjusted accordingly, using the equation above. If the homology between the probe and the target fragment are not known, the simplest approach is to start with both hybridization and wash conditions which are nonstringent. If nonspecific bands or high background are observed after autoradiography, the filter can be washed at high stringency and reexposed. If the time required for exposure makes this approach impractical, several hybridization and/or washing stringencies should be tested in parallel. [0097]
  • Typically, stringent conditions will be those in which the salt concentration is less than about 1.5 M Na ion, typically about 0.01 to 1.0 M Na ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30° C. for short probes (e.g., 10 to 50 nucleotides) and at least about 60° C. for long probes (e.g., greater than 50 nucleotides). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide. Exemplary low stringency conditions include hybridization with a buffer solution of 30 to 35% formamide, 1 M NaCl, 1% SDS (sodium dodecyl sulphate) at 37° C., and a wash in 1× to 2×SSC (20×SSC=3.0 M NaCl/0.3 M trisodium citrate) at 50 to 55° C. Exemplary moderate stringency conditions include hybridization in 40 to 45% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 0.5× to 1×SSC at 55 to 60° C. Exemplary high stringency conditions include hybridization in 50% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 1×SSC at 60 to 65° C. [0098]
  • A composition containing A is “substantially free of” B when at least about 85% by weight of the total A+B in the composition is A. Preferably, A comprises at least about 90% by weight of the total of A+B in the composition, more preferably at least about 95% or even 99% by weight. [0099]
  • The term “heterologous” refers to two biological components that are not found together in nature. The components may be host cells, genes, or regulatory regions, such as promoters. Although the heterologous components are not found together in nature, they can function together, as when a promoter heterologous to a gene is operably linked to the gene. Heterologous components may be from the same or different organisms. Another example is where a human CNGC-15 or Myosin IXa coding sequence is heterologous to a mouse host cell. [0100]
  • A “promoter” is a DNA sequence that initiates and regulates the transcription of a coding sequence when the promoter is operably linked to the coding sequence. A promoter is “heterologous” to the coding sequence when the promoter is not operably linked to the coding sequence in nature. For example, a human CNGC-15 Myosin IXa promoter would comprise an expression cassette wherein a nucleotide sequence encoding CNGC-15 is operably linked to a heterologous promoter. A “native” promoter is operably linked to the coding sequence in nature. [0101]
  • An “origin of replication” is a polynucleotide sequence that initiates and regulates replication of polynucleotides, such as an expression vector. The origin of replication behaves as an autonomous unit of polynucleotide replication within a cell, capable of replication under its own control. With certain origins of replication, an expression vector can be reproduced at a high copy number in the presence of the appropriate proteins within the cell. Examples of origins are the 2μ and autonomously replicating sequences, which are effective in yeast; and the viral T-antigen, effective in COS-7 cells. [0102]
  • Host cells capable of producing CNGC-15 and/or Myosin IXa polypeptides are cultured “under conditions allowing expression.” Such conditions allow transcription and translation of the DNA molecule encoding the CNGC-15 and/or Myosin IXa polypeptide. These conditions include cultivation temperature, oxygen concentration, media composition, pH, etc. For example, if the trp promoter is utilized in the expression vector, the media will lack tryptophan to trigger the promoter and induce expression. The exact conditions will vary from host cell to host cell and from expression vector to expression vector. [0103]
  • As used herein, the term “antibody” refers to a polypeptide or group of polypeptides composed of at least one antibody combining site. An “antibody combining site” is the three-dimensional binding space with an internal surface shape and charge distribution complementary to the features of an epitope of an antigen, which allows a binding of the antibody with the antigen. “Antibody” includes, for example, vertebrate antibodies, hybrid antibodies, chimeric antibodies, altered antibodies, univalent antibodies, the Fab proteins, and single domain antibodies. [0104]
  • “Myosin IXa-modulated disorders” and “CNGC-15 modulated-disorders” include, but are not limited to Bardet-Biedl Syndrome, Usher Syndrome, hearing loss, retinis pigmentosa, obesity, hypogonadism, sterility, polydactyly, brachydactyly, syndactyly, mental retardation, renal abnormalities, hypertension, diabetes and cardiovascular abnormalities [0105]
  • B. General Method [0106]
  • This invention provides the amino acid and nucleotide sequences of CNGC-15 and Myosin IXa. With these disclosed sequences, nucleic acid probe assays and expression cassettes and vectors for CNGC-15 and Myosin IXa polypeptides can be produced. The expression vectors can be transformed into host cells to produce CNGC-15 and/or Myosin IXa polypeptides. The purified polypeptides can be used to produce antibodies to detect CNGC-15 or Myosin IXa. Also, the host cells or extracts can be utilized for biological assays to isolate agonists or antagonists. [0107]
  • Nucleic Acid CNGC-15 Probe Assays [0108]
  • mRNA levels in different cell types can be detected with nucleic acid probe assays. For example, PCR, branched DNA probe assays, or blotting techniques utilizing nucleic acid probes substantially identical or complementary to SEQ ID NO: 2 or SEQ ID NO: 3 can determine the presence of CNGC-15 or Myosin IXa cDNA or mRNA. [0109]
  • For genomic analysis or detection of denatured DNA, the nucleic acid probes will hybridize to a nucleotide sequence encoding the amino acid sequence shown in SEQ ID NO: 1 or encoded by SEQ ID NO: 3, or the complement of a sequence encoding SEQ ID NO: 1 or the polypeptide encoded by SEQ ID NO: 3. Though many different nucleotide sequences will encode the amino acid sequences, SEQ ID NO: 2 and SEQ ID NO: 3 are preferred because they are the actual sequences present in human cells. For single-stranded cDNA detection, the nucleic acid probe will hybridize to the complement of a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 1 or encoding the polypeptide encoded by nucleotides 369-1307 of SEQ ID NO: 3 or to a complement of SEQ ID NO: 2 or SEQ ID NO: 3. For mRNA detection, the nucleic acid probe will hybridize to SEQ ID NO: 2 or SEQ ID NO: 3 or to a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 1 or encoding the polypeptide encoded by SEQ ID NO: 3. The nucleic acid probe sequences need not be identical to SEQ ID NO: 2, SEQ ID NO: 3 or their complement. [0110]
  • Probes are typically at least about 20 nucleotides, more preferably at least about 30 nucleotides. The probes may be produced by synthetic procedures, such as the triester method of Matteucci et al. (1981) [0111] J. Am. Chem. Soc. 103:3185, or according to Urdea et al. (1983) Proc. Natl. Acad. Sci. USA 80:7461, or using commercially available automated oligonucleotide synthesizers. One example of a nucleotide hybridization assay is described in Urdea et al. PCT WO92/02526 and Urdea et al. U.S. Pat. No. 5,124,246, herein incorporated by reference. Other methods of hybridization and detection are known to those skilled in the art.
  • Alternatively, the Polymerase Chain Reaction (PCR) is another well-known means for detecting small amounts of target nucleic acids. The assay is described in Mullis et al. (1987) [0112] Meth. Enzymol. 155:335-350; U.S. Pat. Nos. 4,683,195; and 4,683,202, incorporated herein by reference. Also, mRNA, cDNA and genomic DNA can be detected by traditional blotting techniques described in Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual (New York, Cold Spring Harbor Laboratory).
  • Expression of CNGC-15 and Myosin IXa Polypeptides [0113]
  • Preferably, CNGC-15 and Myosin IXa polypeptides are produced by recombinantly engineered host cells. These host cells are constructed by the introduction of a expression vector comprising a promoter operably linked to a CNGC-15 or Myosin IXa polypeptide coding sequence. [0114]
  • Such coding sequences can be constructed by synthesizing the entire gene or by altering existing CNGC-15 or Myosin IXa polypeptide coding sequences. CNGC-15 and Myosin IXa polypeptides can be divided into four general categories: mutants, fragments, fusions, and the native CNGC-15 or Myosin IXa polypeptides. The CNGC-15 polypeptides are those that occur in nature. The amino acid sequence of such polypeptides may vary slightly from SEQ ID NO: 1. The native CNGC-15 and Myosin IXa polypeptide coding sequence can be selected based on the amino acid sequence shown in SEQ ID NO: 1 or the amino acid sequence predicted by the open reading frame of SEQ ID NO: 3. For example, synthetic genes can be made using codons preferred by the host cell to encode the desired polypeptide. (See Urdea et al. (1983) [0115] Proc. Natl. Acad. Sci. USA 80:7461). Alternatively, the desired native CNGC-15 and Myosin IXa polypeptide coding sequences can be cloned from nucleic acid libraries. Techniques for producing and probing nucleic acid sequence libraries are described, for example, in Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual (New York, Cold Spring Harbor Laboratory). Other recombinant techniques, such as site specific mutagenesis, PCR, enzymatic digestion and ligation, can also be used to construct the desired CNGC-15 polypeptide coding sequence.
  • The native CNGC-15 and Myosin IXa polypeptide coding sequences can be modified to create the other classes of CNGC-15 and Myosin IXa polypeptides. For example, mutants can be created by making conservative amino acid substitutions that maintain or enhance native CNGC-15 or Myosin IXa activity. The following are examples of conservative substitutions: Gly [0116]
    Figure US20020091248A1-20020711-P00900
    Ala; Val
    Figure US20020091248A1-20020711-P00900
    Ile
    Figure US20020091248A1-20020711-P00900
    Leu; Asp
    Figure US20020091248A1-20020711-P00900
    Glu; Lys
    Figure US20020091248A1-20020711-P00900
    Arg; Asn
    Figure US20020091248A1-20020711-P00900
    Gln; and Phe
    Figure US20020091248A1-20020711-P00900
    Trp
    Figure US20020091248A1-20020711-P00900
    Tyr. Mutants can also contain amino acid deletions or insertions compared to the native CNGC-15 or Myosin IXa polypeptides. Mutants may include substitutions, insertions, and deletions of the native polypeptides.
  • Mutants will retain at least about 20% of the one of the activities of the native CNGC-15 or Myosin IXa. The coding sequence of mutants can be constructed by in vitro mutagenesis of the native coding sequences. [0117]
  • A subset of mutants is a group of polypeptides with the non-disulfide bond participating cysteines substituted with a neutral amino acid, generally, with serines. These mutants may be stable over a broader temperature range than native CNGC-15 or Myosin IXa polypeptides. See, for example, Mark et al., U.S. Pat. No. 4,959,314. [0118]
  • Fragments differ from mutant or native CNGC-15 or Myosin IXa polypeptides by amino and/or carboxyl terminal amino acid deletions. The number of amino acids that are truncated is not critical as long as the CNGC-15 or Myosin IXa fragment retains at least about 20% of the one of the activities of the native CNGC-15 or Myosin IXa polypeptide. The coding sequence of such fragments can be easily constructed by cleaving the unwanted nucleotides from the mutant or native CNGC-15 or Myosin IXa polypeptide coding sequences. [0119]
  • Fusions are fragment, mutant, or native CNGC-15 or Myosin IXa polypeptides with additional amino acids at either or both of the termini. The additional amino acid sequence is not necessarily homologous to sequence found in native polypeptides. The fusions, just as all CNGC-15 and Myosin IXa polypeptides, retain at least about 20% of one of the activities of the native CNGC-15 or Myosin IXa polypeptides. Coding sequence of the fusions can be constructed by ligating synthetic polynucleotides encoding the additional amino acids to fragment, mutant, or native coding sequences. Activities of the CNGC-15 and Myosin IXa polypeptides can be determined by the methods described infra. [0120]
  • Expression Vectors [0121]
  • At the minimum, an expression vector will contain a promoter which is operable in the host cell and operably linked to a CNGC-15 or Myosin IXa coding sequence. Sequences that modulate gene expression, such as enhancers and binding sites for inducers or repressors may be present. Expression vectors may also include signal sequences, terminators, selectable markers, origins of replication, and sequences homologous to host cell sequences. These additional elements are optional but can be included to optimize expression. [0122]
  • Functional non-natural promoters may also be used, for example, synthetic promoters based on a consensus sequence of different promoters. Also, effective promoters can contain a regulatory region linked with a heterologous expression initiation region. Examples of hybrid promoters are the [0123] E. coli lac operator linked to the E. coli tac transcription activation region; the yeast alcohol dehydrogenase (ADH) regulatory sequence linked to the yeast glyceraldehyde-3-phosphate-dehydrogenase (GAPDH) transcription activation region (U.S. Pat. Nos. 4,876,197 and 4,880,734, incorporated herein by reference); and the cytomegalovirus (CMV) enhancer linked to the SV40 (simian virus) promoter.
  • A CNGC-15 or Myosin IXa polypeptide coding sequence may also be linked in reading frame to a signal sequence. The signal sequence fragment typically encodes a peptide comprised of hydrophobic amino acids which directs the CNGC-15 or Myosin IXa polypeptide to the cell membrane or other subcellular compartment. Preferably, there are processing sites encoded between the leader fragment and the gene or fragment thereof that can be cleaved either in vivo or in vitro. DNA encoding suitable signal sequences can be derived from genes for secreted endogenous host cell proteins, such as the yeast invertase gene (EP 12 873; JP 62,096,086), the A-factor gene (U.S. Pat. No. 4,588,684), interferon signal sequence (EP 60 057). [0124]
  • A preferred class of secretion leaders, for yeast expression, are those that employ a fragment of the yeast alpha-factor gene, which contains both a “pre” signal sequence, and a “pro” region. The types of alpha-factor fragments that can be employed include the full-length pre-pro alpha factor leader (about 83 amino acid residues) as well as truncated alpha-factor leaders (typically about 25 to about 50 amino acid residues) (U.S. Pat. Nos. 4,546,083 and 4,870,008, incorporated herein by reference; EP 324 274). Additional leaders employing an alpha-factor leader fragment that provides for secretion include hybrid alpha-factor leaders made with a presequence of a first yeast signal sequence, but a pro-region from a second yeast alpha-factor. (See e.g., PCT WO 89/02463). [0125]
  • Typically, terminators are regulatory sequences, such as polyadenylation and transcription termination sequences, located 3′ or downstream of the stop codon of the coding sequences. Usually, the terminator of native host cell proteins are operable when attached 3′ of a CNGC-15 or Myosin IXa polypeptide coding sequences. Examples are the [0126] Saccharomyces cerevisiae alpha-factor terminator and the baculovirus terminator. Further, viral terminators are also operable in certain host cells; for instance, the SV40 terminator is functional in CHO cells.
  • For convenience, selectable markers, an origin of replication, and homologous host cells sequences may optionally be included in an expression vector. A selectable marker can be used to screen for host cells that potentially contain the expression vector. Such markers may render the host cell immune to drugs such as ampicillin, chloramphenicol, erythromycin, neomycin, and tetracycline. Also, markers may be biosynthetic genes, such as those in the histidine, tryptophan, and leucine pathways. Thus, when leucine is absent from the media, for example, only the cells with a biosynthetic gene in the leucine pathway will survive. [0127]
  • An origin of replication may be needed for the expression vector to replicate in the host cell. Certain origins of replication enable an expression vector to be reproduced at a high copy number in the presence of the appropriate proteins within the cell. Examples of origins are the 2μ and autonomously replicating sequences, which are effective in yeast; and the viral T-antigen, effective in COS-7 cells. [0128]
  • Expression vectors may be integrated into the host cell genome or remain autonomous within the cell. Polynucleotide sequences homologous to sequences within the host cell genome may be needed to integrate the expression cassette. The homologous sequences do not always need to be linked to the expression vector to be effective. For example, expression vectors can integrate into the CHO genome via an unattached dihydrofolate reductase gene. In yeast, it is more advantageous if the homologous sequences flank the expression cassette. Particularly useful homologous yeast genome sequences are those disclosed in PCT WO 90/01800, and the HIS4 gene sequences, described in Genbank, Accession No. J01331. [0129]
  • The choice of promoter, terminator, and other optional elements of an expression vector will also depend on the host cell chosen. The invention is not dependent on the host cell selected. Convenience and the level of protein expression will dictate the optimal host cell. A variety of hosts for expression are known in the art and available from the American Type Culture Collection (ATCC). Bacterial hosts suitable for expressing a CNGC-15 and/or Myosin IXa polypeptide include, without limitation: Campylobacter, Bacillus, Escherichia, Lactobacillus, Pseudomonas, Staphylococcus, and Streptococcus. Yeast hosts from the following genera may be utilized: Candida, Hansenula, Kluyveromyces, Pichia, Saccharomyces, Schizosaccharomyces, and Yarrowia. Immortalized mammalian host cells include but are not limited to CHO cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), and other cell lines. A number of insect cell hosts are also available for expression of heterologous proteins: [0130] Aedes aegypti, Bombyx mori, Drosophila melanogaster, and Spodoptera frugiperda (PCT WO 89/046699; Carbonell et al. (1985) J. Virol. 56:153; Wright et al. (1986) Nature 321:718; Smith et al. (1983) Mol. Cell. Biol. 3:2156; and see generally, Fraser et al. (1989) In Vitro Cell. Dev. Biol. 25:225).
  • Transformation [0131]
  • After vector construction, the desired CNGC-15 and/or Myosin IXa polypeptide expression vector is inserted into the host cell. Many transformation techniques exist for inserting expression vectors into bacterial, yeast, insect, and mammalian cells. The transformation procedure to introduce the expression vector depends upon the host to be transformed. [0132]
  • Methods of introducing exogenous DNA into bacterial hosts are well-known in the art, and typically protocol includes either treating the bacteria with CaCl[0133] 2 or other agents, such as divalent cations and DMSO. DNA can also be introduced into bacterial cells by electroporation or viral infection. Transformation procedures usually vary with the bacterial species to be transformed. See e.g. (Masson et al. (1989) FEMS Microbiol. Lett. 60:273; Palva et al. (1982) Proc. Natl. Acad. Sci. USA 79:5582; EP Publ. Nos. 036 259 and 063 953; PCT WO 84/04541, Bacillus), (Miller et al. (1988) Proc. Natl. Acad. Sci. 85:856; Wang et al. (1990) J. Bacteriol. 172:949, Campylobacter), (Cohen et al. (1973) Proc. Natl. Acad. Sci. 69:2110; Dower et al. (1988) Nucleic Acids Res. 16:6127; Kushner et al. (1978) “An Improved Method for Transformation of Escherichia coli with ColE1-derived plasmids in Genetic Engineering: Proceedings of the International Symposium on Genetic Engineering (eds. H. W. Boyer and S. Nicosia); Mandel et al. (1970) J. Mol. Biol. 53:159; Taketo et al. (1988) Biochim. Biophys. Acta 949:318; Escherichia), (Chassy et al. (1987) FEMS Microbiol. Lett. 44:173, Lactobacillus); (Fiedler et al. (1988) Anal. Biochem. 1 70:38, Pseudomonas); (Augustin et al. (1990) FEMS Microbiol. Lett. 66:203, Staphylococcus), Barany et al. (1980) J. Bacteriol. 144:698; Harlander et al. (1987) “Transformation of Streptococcus lactis by electroporation,” in Streptococcal Genetics (ed. J. Ferretti and R. Curtiss, III); Perry et al. (1981) Infec. Immun. 32:1295; Powell et al. (1988) Appl. Environ. Microbiol. 54:655; Somkuti et al. (1987) Proc. 4th Evr. Cong. Biotechnology 1:412, Streptococcus).
  • Transformation methods for yeast hosts are well-known in the art, and typically include either the transformation of spheroplasts or of intact yeast cells treated with alkali cations. Electroporation is another means for transforming yeast hosts. See for example, [0134] Methods in Enzymology, Volume 194, 1991, “Guide to Yeast Genetics and Molecular Biology.” Transformation procedures usually vary with the yeast species to be transformed. See e.g. (Kurtz et al. (1986) Mol. Cell. Biol. 6:142; Kunze et al. (1985) J. Basic Microbiol. 25:141, Candida); (Gleeson et al. (1986) J. Gen. Microbiol. 132:3459; Roggenkamp et al. (1986) Mol. Gen. Genet. 202:302, Hansenula); (Das et al. (1984) J. Bacteriol. 158:1165; De Louvencourt et al. (1983) J. Bacteriol. 154:1165; Van den Berg et al. (1990) Biotechnology 8:135, Kluyveromyces); (Cregg et al. (1985) Mol. Cell. Biol. 5:3376; Kunze et al. (1985) J. Basic Microbiol. 25:141; U.S. Pat. Nos. 4,837,148 and 4,929,955, Pichia); (Hinnen et al. (1978) Proc. Natl. Acad. Sci. USA 75:1929; Ito et al. (1983) J. Bacteriol. 153:163, Saccharomyces); (Beach and Nurse (1981) Nature 300:706, Schizosaccharomyces); (Davidow et al. (1985) Curr. Genet. 10:39; Gaillardin et al. (1985) Curr. Genet. 10:49, Yarrowia).
  • Methods for introducing heterologous polynucleotides into mammalian cells are known in the art and include viral infection, dextran-mediated transfection, calcium phosphate precipitation, microparticle bombardment, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei. [0135]
  • The method for construction of an expression vector for transformation of insect cells for expression of recombinant herein is slightly different than that generally applicable to the construction of a bacterial expression vector, a yeast expression vector, or a mammalian expression vector. In an embodiment of the present invention, a baculovirus vector is constructed in accordance with techniques that are known in the art, for example, as described in Kitts et al. (1993) [0136] Biotechniques 14:810-817; Smith et al. (1983) Mol. Cell. Biol. 3:2156; and Luckow and Summer (1989) Virol. 17:31. In one embodiment of the present invention, a baculovirus expression vector is constructed substantially in accordance to Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987). Moreover, materials and methods for baculovirus/insect cell expression systems are commercially available in kit form, for example, the MaxBac kit from Invitrogen (San Diego, Calif.).
  • Also, methods for introducing heterologous DNA into an insect host cell are known in the art. For example, an insect cell can be infected with a virus containing a CNGC-15 or Myosin IXa polypeptide coding sequence. When the virus is replicating in the infected cell, the CNGC-15 or Myosin IXa polypeptide will be expressed if operably linked to a suitable promoter. A variety of suitable insect cells and viruses are known and include following without limitation. [0137]
  • Insect cells from any order of the Class Insecta can be used to express the polypeptide of this invention. The orders Diptera and Lepidoptera are preferred. Example of insect species are listed in Weiss et al. “Cell Culture Methods for Large-Scale Propagation of Baculoviruses,” in Granados et al. (eds., [0138] The Biology of Baculoviruses: Vol. II Practical Application for Insect Control, pp. 63-87 at p. 64 (1987). Insect cell lines derived from the following insects are exemplary: Carpocapsa pomeonella (preferably, cell line CP-128); Trichoplusia ni (preferably, cell line TN-368); Autograph californica; Spodoptera frugiperda (preferably, cell line Sf9); Lymantria dispar; Mamestra brassicae; Aedes albopictus; Orgyia pseudotsugata; Neodiprio sertifer; Aedes aegypti; Antherae eucalypti; Gnorimoschema operceullela; Galleria mellonella; Spodoptera littolaris; Blatella germanic; Drosophila melanogaster; Heliothis zea; Spodoptera exigua; Rachiplusia ou; Plodia interpunctella, Amsaeta moorei; Agrotis c-nigrum, Adoxophyes orana; Agrotis segetum; Bombyx mori; Hyponomeuta malinellu; Colias eurytheme; Anticarsia germmetalia, Apanteles melanoscelu; Arctia caja; and Porthetria dispar. Preferred insect cell lines are from Spodoptera frugiperda, and especially preferred is cell line Sf9. The Sf9 cell line used in the examples herein was obtained from Max D. Summers (Texas A & M University, College Station, Tex., 77843, USA). Other S. frugiperda cell lines, such as IPL-Sf-21AE III, are described in Vaughn et al. (1977) In Vitro 13:213-217.
  • The insect cell lines of this invention are suitable for the reproduction of numerous insect-pathogenic viruses such as parvoviruses, pox viruses, baculoviruses and rhabdoviruses, of which nucleopolyhedrosis viruses (NPV) and granulosis viruses (GV) from the group of baculoviruses are preferred. Further preferred are NPV viruses such as those from Autographa spp., Spodoptera spp., Trichoplusia spp., Rachiplusia spp., Gallerai spp., and Lymantria spp. More preferred are baculovirus strain [0139] Autographa californica NPV (AcNPV), Rachiplusia ou NPV, Galleria mellonella NPV, and any plaque purified strains of AcNPV, such as E2, R9, S1, M3, characterized and described by Smith et al. (1978) Virol. 89:517-527.
  • Typically, insect cells [0140] Spodoptera frugiperda type 9 (SF9) are infected with baculovirus strain Autographa californica NPV (AcNPV) containing a CNGC-15 or Myosin IXa polypeptide coding sequence. Such a baculovirus is produced by homologous recombination between a transfer vector containing the coding sequence and baculovirus sequences and a genomic baculovirus DNA. Preferably, the genomic baculovirus DNA is linearized and contains a dysfunctional essential gene. The transfer vector, preferably, contains the nucleotide sequences needed to restore the dysfunctional gene and a baculovirus polyhedrin promoter and terminator operably linked to the CNGC-15 or Myosin IXa coding sequence. (See Kitts et al. (1993) Biotechniques 14(5):810-817.
  • The transfer vector and linearized baculovirus genome are transfected into SF9 insect cells, and the resulting viruses probably containing the desired coding sequence. Without a functional essential gene the baculovirus genome cannot produce a viable virus. Thus, the viable viruses from the transfection most likely contain the CNGC-15 or Myosin IXa polypeptide coding sequence and the needed essential gene sequences from the transfer vector. Further, lack of occlusion bodies in the infected cells are another verification that the CNGC-15 or Myosin IXa polypeptide coding sequence was incorporated into the baculovirus genome. [0141]
  • The essential gene and the polyhedrin gene flank each other in the baculovirus genome. The coding sequence in the transfer vector is flanked at its 5′ with the essential gene sequences and the polyhedrin promoter and at its 3′ with the polyhedrin terminator. Thus, when the desired recombination event occurs the CNGC-15 or Myosin IXa polypeptide coding sequence displaces the baculovirus polyhedrin gene. Such baculoviruses without a polyhedrin gene will not produce occlusion bodies in the infected cells. Of course, another means for determining if coding sequence was incorporated into the baculovirus genome is to sequence the recombinant baculovirus genomic DNA. Alternatively, expression of the desired CNGC-15 or Myosin IXa polypeptide by cells infected with the recombinant baculovirus is another verification means. [0142]
  • Monitoring CNGC-15 Polypeptide Expression Levels [0143]
  • Immunoassays and ligand binding assays can be utilized to confirm that the transformed host cell is expressing the desired CNGC-15 and/or Myosin IXa polypeptide. [0144]
  • For example, an immunofluorescence assay can be performed on transformed host cells without separating the CNGC-15 or Myosin IXa polypeptides from the cell. The host cells are first fixed onto a solid support, such as a microscope slide or microtiter well. This fixing step permeabilizes the cell membrane. Next, the fixed host cells are exposed to an anti-CNGC-15 or anti-Myosin IXa polypeptide antibody. Preferably, to increase the sensitivity of the assay, the fixed cells are exposed to a second antibody, which is labeled and binds to the anti-CNGC-15 or anti-Myosin IXa polypeptide antibody. Typically, the secondary antibody is labeled with an fluorescent marker. The host cells which express the CNGC-15 or Myosin IXa polypeptides will be fluorescently labeled and easily visualized under the microscope. See, for example, Hashido et al. (1992) [0145] Biochem & Biophys. Res. Comm. 187(3):1241-1248.
  • Also, the CNGC-15 and Myosin IXa polypeptides do not need to be separated from the cell membrane for in vitro assays. The host cells may be fixed to a solid support, such as a microtiter plate. Alternatively, a crude membrane fraction can be separated from lysed host cells by centrifugation (See Adachi et al. (1992) [0146] FEBS Lett 311(2):179-183. The fixed host cells or the crude membrane fraction is exposed to labeled ligand or ion. Typically, the ligand is labeled with radioactive atoms. The host cells which express the desired CNGC-15 or Myosin IXa polypeptide will bind with the labeled ligand which can be easily detected.
  • Purification [0147]
  • The purified CNGC-15 and Myosin IXa polypeptides are useful as compositions, for assays, and to produce antibodies. [0148]
  • CNGC-15 and Myosin IXa polypeptides can be isolated by a variety of steps including, for example, anion exchange chromatography, size exclusion chromatography, hydroxyapatite chromatography, hydrophobic interaction chromatography, metal chelation chromatography, reverse phase HPLC, affinity chromatography, and further ammonium sulfate precipitations. These techniques are well known to those of skill in the art. [0149]
  • For ligand binding studies, patch clamp analysis or other in vitro assays, the crude cell membrane fractions can be utilized. These membrane extracts can be isolated from cells which expressed CNGC-15 or Myosin IXa polypeptides by lysing the cells. Alternatively, whole cells, expressing CNGC-15 and/or Myosin IXa polypeptides, can be cultured in a microtiter plate. [0150]
  • Antibodies [0151]
  • Antibodies against CNGC-15 and Myosin IXa polypeptides are useful for affinity chromatography, immunofluorescent assays, and distinguishing CNGC-15 and Myosin IXa polypeptides. [0152]
  • Antibodies to the proteins of the invention, both polyclonal and monoclonal, may be prepared by conventional methods known to those skilled in the art. For example, monoclonal antibodies are prepared using the method of Kohler et al. (1975) [0153] Nature 256:495-496, or a modification thereof.
  • If desired, the antibodies (whether polyclonal or monoclonal) may be labeled using conventional techniques. Suitable labels include fluorophores, chromophores, radioactive atoms (particularly [0154] 32P and 125I), electron-dense reagents, enzymes, and ligands having specific binding partners. Enzymes are typically detected by their activity. For example, horseradish peroxidase is usually detected by its ability to convert 3,3′,5,5′-tetra-methylbenzidine (TMB) to a blue pigment, quantifiable with a spectrophotometer. “Specific binding partner” refers to a protein capable of binding a ligand molecule with high specificity, as for example in the case of an antigen and a monoclonal antibody specific therefor. Other specific binding partners include biotin and avidin or streptavidin, IgG and protein A, and the numerous receptor-ligand couples known in the art. It should be understood that the above description is not meant to categorize the various labels into distinct classes, as the same label may serve in several different modes. For example, 125I may serve as a radioactive label or as an electron-dense reagent. HRP may serve as enzyme or as antigen for a MAb. Further, one may combine various labels for desired effect. For example, MAbs and avidin also require labels in the practice of this invention: thus, one might label a MAb with biotin, and detect its presence with avidin labeled with 125I, or with an anti-biotin MAb labeled with HRP. Other permutations and possibilities will be readily apparent to those of ordinary skill in the art, and are considered as equivalents within the scope of the instant invention.
  • Screening for Agonists and Antagonists [0155]
  • CNGC-15 and Myosin IXa polypeptides can also be used to screen combinatorial libraries to identify agonist or antagonists. For example, a “library” of peptides may be synthesized following the methods disclosed in U.S. Pat. No. 5,010,175, and in PCT WO 91/17823, both incorporated herein by reference in full. The peptide library is first screened for binding to the selected CNGC-15 or Myosin IXa polypeptide. The peptides are then tested for their ability to inhibit or enhance CNGC-15 or Myosin IXa activity. Peptides exhibiting the desired activity are then isolated and sequenced. [0156]
  • CNGC-15 and Myosin IXa agonists or antagonists may be screened using any available method. The assay conditions ideally should resemble the conditions under which the CNGC-15 or Myosin IXa activity is exhibited in vivo, i.e., under physiologic pH, temperature, ionic strength, etc. Suitable agonists or antagonists will exhibit strong inhibition or enhancement of the CNGC-15 or Myosin IXa activity at concentrations which do not raise toxic side effects in the subject. Agonists or antagonists which compete for binding to the CNGC-15 or Myosin IXa polypeptide may require concentrations equal to or greater than the native CNGC-15 or Myosin IXa concentration, while inhibitors capable of binding irreversibly to the polypeptide may be added in concentrations on the order of the native CNGC-15 or Myosin IXa concentration. [0157]
  • Signal Transduction Assays [0158]
  • Most cellular Ca[0159] 2+ions are sequestered in the mitochondria, endoplasmic reticulum, and other cytoplasmic vesicles, but binding of endothelin to receptor will trigger the increase of free Ca2+ ions in the cytoplasm. With fluorescent dyes, such as fura-2, the concentration of free Ca2+can be monitored. The ester of fura-2 is added to the media of the host cells expressing receptor polypeptides. The ester of fura-2 is lipophilic and diffuses across the membrane. Once inside the cell, the fura-2 ester is hydrolyzed by cytosolic esterases to its non-lipophilic form, and then the dye cannot diffuse back out of the cell. The non-lipophilic form of fura-2 will fluoresce when it binds to the free Ca2+ ions, which are released after binding of a ligand to the receptor. The fluorescence can be measured without lysing the cells at an excitation spectrum of 340 nm and at fluorescence spectrum of 500 nm. See Sakurai et al. EP 480 381 and Adachi et al. (1992) FEBS Lett 311 (2):179-183 for examples of assays measuring free intracellular Ca2+ concentrations.
  • The rise of free cytosolic Ca[0160] 2+concentrations is preceded by the hydrolysis of phosphatidylinositol 4,5-bisphosphate. Hydrolysis of this phospholipid by the plasma-membrane enzyme phospholipase C yields 1,2-diacylglycerol (DAG), which remains in the membrane, and the water-soluble inosital 1,4,5-trisphosphate (IP3). Binding of endothelin or endothelin agonists will increase the concentration of DAG and IP3. Thus, signal transduction activity can be measured by monitoring the concentration of these hydrolysis products.
  • To measure the IP[0161] 3 concentrations, radioactively labeled 3H-inositol is added to the media of host cells expressing CNGC-15 or Myosin IXa polypeptides. The 3H-inositol taken up by the cells and after stimulation of the cells with endothelin or endothelin agonist, the resulting inositol triphosphate is separated from the mono and di-phosphate forms and measured. See Sakurai et al., EP 480 381. Alternatively, Amersham provides an inosital 1,4,5-trisphosphate assay system. With this system Amersham provides tritylated inosital 1,4,5-trisphosphate and a receptor capable of distinguishing the radioactive inositol from other inositol phosphates. With these reagents an effective and accurate competition assay can be performed to determine the inositol triphosphate levels.
  • Pharmaceutical Compositions [0162]
  • Pharmaceutical compositions can comprise either polypeptides, antibodies, or polynucleotides of the claimed invention. The pharmaceutical compositions will comprise a therapeutically effective amount of either polypeptides, antibodies, or polynucleotides of the claimed invention. [0163]
  • The term “therapeutically effective amount” as used herein refers to an amount of a therapeutic agent to treat, ameliorate, or prevent a desired disease or condition, or to exhibit a detectable therapeutic or preventative effect. The effect can be detected by, for example, chemical markers or antigen levels. Therapeutic effects also include reduction in physical symptoms, such as decreased body temperature. The precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition, and the therapeutics or combination of therapeutics selected for administration. Thus, it is not useful to specify an exact effective amount in advance. However, the effective amount for a given situation can be determined by routine experimentation and is within the judgment of the clinician. [0164]
  • For purposes of the present invention, an effective dose will be from about 0.01 mg/kg to 50 mg/kg or 0.05 mg/kg to about 10 mg/kg of the polypeptide or DNA construct in the individual to which it is administered [0165]
  • A pharmaceutical composition can also contain a pharmaceutically acceptable carrier. The term “pharmaceutically acceptable carrier” refers to a carrier for administration of a therapeutic agent, such as antibodies or a polypeptide, genes, and other therapeutic agents. The term refers to any pharmaceutical carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity. Suitable carriers may be large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles. Such carriers are well known to those of ordinary skill in the art. [0166]
  • Pharmaceutically acceptable salts can be used therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. A thorough discussion of pharmaceutically acceptable excipients is available in [0167] Remington's Pharmaceutical Sciences (Mack Pub. Co., N.J. 1991).
  • Pharmaceutically acceptable carriers in therapeutic compositions may contain liquids such as water, saline, glycerol and ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles. Typically, the therapeutic compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared. Liposomes are included within the definition of a pharmaceutically acceptable carrier. [0168]
  • Delivery Methods [0169]
  • Once formulated, the compositions of the invention can be administered directly to the subject. The subjects to be treated can be mammals or birds. In particular, human subjects can be treated. [0170]
  • Direct delivery of the compositions will generally be accomplished by injection, either subcutaneously, intraperitoneally, intravenously or intramuscularly or delivered to the interstitial space of a tissue. The compositions can also be administered into a tumor or lesion. Other modes of administration include oral and pulmonary administration, suppositories, and transdermal applications, needles, and gene guns or hyposprays. Dosage treatment may be a single dose schedule or a multiple dose schedule. [0171]
  • Alternatively, the Myosin IXa or CNGC-15 polypeptides could be stably expressed in an organ of a mammal, and then the organ could be xenografted into a human in need of such treatment. [0172]
  • Gene Delivery Vehicles [0173]
  • Gene therapy vehicles for delivery of constructs including a coding sequence of a therapeutic of the invention, to be delivered to the mammal for expression in the mammal, can be administered either locally or systemically. These constructs can utilize viral or non-viral vector approaches in in vivo or ex vivo modality. Expression of such coding sequence can be induced using endogenous mammalian or heterologous promoters. Expression of the coding sequence in vivo can be either constitutive or regulated. [0174]
  • The invention includes gene delivery vehicles capable of expressing the contemplated nucleic acid sequences. The gene delivery vehicle is preferably a viral vector and, more preferably, a retroviral, adenoviral, adeno-associated viral (AAV), herpes viral, or alphavirus vector. The viral vector can also be an astrovirus, coronavirus, orthomyxovirus, papovavirus, paramyxovirus, parvovirus, picomavirus, poxvirus, or togavirus viral vector. See generally, Jolly et al. (1994) [0175] Cancer Gene Therapy 1:51-64; Kimura et al. (1994) Human Gene Therapy 5:845-852; Connelly et al. (1995) Human Gene Therapy 6:185-193; and Kaplitt et al. (1994) Nature Genetics 6:148-153.
  • Retroviral vectors are well known in the art and we contemplate that any retroviral gene therapy vector is employable in the invention, including B, C and D type retroviruses, xenotropic retroviruses (for example, NZB-X1, NZB-X2 and NZB9-1 (see O'Neill (1985) [0176] J. Vir. 53:160) polytropic retroviruses (for example, MCF and MCF-MLV (see Kelly et al. (1983) J. Vir. 45:291), spumaviruses and lentiviruses. See RNA Tumor Viruses, Second Edition, Cold Spring Harbor Laboratory, 1985.
  • Portions of the retroviral gene therapy vector may be derived from different retroviruses. For example, retrovector LTRs may be derived from a Murine Sarcoma Virus, a tRNA binding site from a Rous Sarcoma Virus, a packaging signal from a Murine Leukemia Virus, and an origin of second strand synthesis from an Avian Leukosis Virus. [0177]
  • These recombinant retroviral vectors may be used to generate transduction competent retroviral vector particles by introducing them into appropriate packaging cell lines (see U.S. Ser. No. 07/800,921, filed Nov. 29, 1991). Retrovirus vectors can be constructed for site-specific integration into host cell DNA by incorporation of a chimeric integrase enzyme into the retroviral particle. See, U.S. Ser. No. 08/445,466 filed May 22, 1995. It is preferable but not required that the recombinant viral vector is a replication defective recombinant virus. [0178]
  • Packaging cell lines suitable for use with the above-described retrovirus vectors are well known in the art, are readily prepared (see U.S. Ser. No. 08/240,030, filed May 9, 1994; see also WO 92/05266), and can be used to create producer cell lines (also termed vector cell lines or “V CLs”) for the production of recombinant vector particles. Preferably, the packaging cell lines are made from human parent cells (e.g., HT1080cells) or mink parent cell lines, which eliminates inactivation in human serum. [0179]
  • Preferred retroviruses for the construction of retroviral gene therapy vectors include Avian Leukosis Virus, Bovine Leukemia, Virus, Murine Leukemia Virus, Mink-Cell Focus-Inducing Virus, Murine Sarcoma Virus, Reticuloendotheliosis Virus and Rous Sarcoma Virus. Particularly preferred Murine Leukemia Viruses include 4070A and 1504A (Hartley et al. (1976) [0180] J. Virol. 19:19-25), Abelson (ATCC No. VR-999), Friend (ATCC No. VR-245), Graffi, Gross (ATCC Nol VR-590), Kirsten, Harvey Sarcoma Virus and Rauscher (ATCC No. VR-998) and Moloney Murine Leukemia Virus (ATCC No. VR-190). Such retroviruses may be obtained from depositories or collections such as the American Type Culture Collection (“ATCC”) in Rockville, Md. or isolated from known sources using commonly available techniques.
  • Exemplary known retroviral gene therapy vectors employable in this invention include those described in GB 2200651, EP 0415731, EP 0345242, WO 89/02468, WO 89/05349, WO 89/09271, WO 90/02806, WO 90/07936, WO 94/03622, WO 93/25698, WO 93/25234, WO 93/11230, WO 93/10218, WO 91/02805, in U.S. Pat. Nos. 5,219,740, 4,405,712, 4,861,719, 4,980,289 and 4,777,127, in U.S. Ser. No. 07/800,921 and in Vile et al. (1993) [0181] Cancer Res. 53:3860-3864; Vile et al. (1993) Cancer Res. 53:962-967; Ram et al. (1993) Cancer Res. 53.83-88; Takamiya et al. (1992) J. Neurosci. Res. 33:493-503; Baba et al. (1993) J. Neurosurg. 79:729-735; Mann et al. (1983) Cell 33:153; Cane et al. (1984) Proc. Natl. Acad. Sci. 81:6349; and Miller et al. (1990) Human Gene Therapy 1.
  • Human adenoviral gene therapy vectors are also known in the art and employable in this invention. See, for example, Berkner et al. (1988) [0182] Biotechniques 6:616; Rosenfeld et al. (199 1) Science 252:431; and WO 93/07283, WO 93/06223, and WO 93/07282. Exemplary known adenoviral gene therapy vectors employable in this invention include those described in the above referenced documents and in WO 94/12649, WO 93/03769, WO 93/19191, WO 94/28938, WO 95/11984, WO 95/00655, WO 95/27071, WO 95/29993, WO 95/34671, WO 96/05320, WO 94/08026, WO 94/11506, WO 93/06223, WO 94/24299, WO 95/14102, WO 95/24297, WO 95/02697, WO 94/28152, WO 94/24299, WO 95/09241, WO 95/25807, WO 95/05835, WO 94/18922, WO 95/09654. Alternatively, administration of DNA linked to killed adenovirus as described in Curiel et al. (1992) Hum. Gene Ther. 3:147-154 may be employed.
  • The gene delivery vehicles of the invention also include adenovirus associated virus (AAV) vectors. Leading and preferred examples of such vectors for use in this invention are the AAV-2 based vectors disclosed in Srivastava, WO 93/09239. Most preferred AAV vectors comprise the two AAV inverted terminal repeats in which the native D-sequences are modified by substitution of nucleotides, such that at least about 5 native nucleotides and up to 18 native nucleotides, preferably at least about 10 native nucleotides up to 18 native nucleotides, most preferably 10 native nucleotides are retained and the remaining nucleotides of the D-sequence are deleted or replaced with non-native nucleotides. The native D-sequences of the AAV inverted terminal repeats are sequences of 20 consecutive nucleotides in each AAV inverted terminal repeat (i.e., there is one sequence at each end) which are not involved in HP formation. The non-native replacement nucleotide may be any nucleotide other than the nucleotide found in the native D-sequence in the same position. Other employable exemplary AAV vectors are pWP-19, pWN-1, both of which are disclosed in Nahreini et al. (1993) [0183] Gene 124:257-262. Another example of such an AAV vector is psub201. See Samulski et al. (1987) J. Virol. 61:3096. Another exemplary AAV vector is the Double-D ITR vector. Construction of the Double D ITR vector is disclosed in U.S. Pat. No. 5,478,745. Still other vectors are those disclosed in Carter, U.S. Pat. No. 4,797,368 and Muzyczka, U.S. Pat. No. 5,139,941, Chartejee, U.S. Pat. No. 5,474,935, and Kotin, PCT Patent Publication WO 94/288157. Yet a further example of an AAV vector employable in this invention is SSV9AFABTKneo, which contains the AFP enhancer and albumin promoter and directs expression predominantly in the liver. Its structure and construction are disclosed in SU, (1996) Human Gene Therapy 7:463-470. Additional AAV gene therapy vectors are described in U.S. Pat. Nos. 5,354,678, 5,173,414, 5,139,941, and 5,252,479.
  • The gene therapy vectors of the invention also include herpes vectors. Leading and preferred examples are herpes simplex virus vectors containing a sequence encoding a thymidine kinase polypeptide such as those disclosed in U.S. Pat. No. 5,288,641 and EP 0176170 (Roizman). Additional exemplary herpes simplex virus vectors include HFEM/ICP6-LacZ disclosed in WO 95/04139 (Wistar Institute), pHSVlac described in Geller et al. (1988) [0184] Science 241:1667-1669 and in WO 90/09441 and WO 92/07945, HSV Us3::pgC-lacZ described in Fink, (1992) Human Gene Therapy 3:11-19 and HSV 7134, 2 RH 105 and GAL4 described in EP 0453242 (Breakefield), and those deposited with the ATCC as accession numbers ATCC VR-977 and ATCC VR-260.
  • Also contemplated are alpha virus gene therapy vectors that can be employed in this invention. Preferred alpha virus vectors are Sindbis viruses vectors. Togaviruses, Semliki Forest virus (ATCC VR-67; ATCC VR-1247), Middleberg virus (ATCC VR-370), Ross River virus (ATCC VR-373; ATCC VR-1246), Venezuelan equine encephalitis virus (ATCC VR923; ATCC VR-1250; ATCC VR-1249; ATCC VR-532), and those described in U.S. Pat. Nos. 5,091,309, 5,217,879, and WO 92/10578, WO 95/07994, U.S. Pat. Nos. 5,091,309 and 5,217,879 are employable. Such alpha viruses may be obtained from depositories or collections such as the ATCC in Rockville, Md. or isolated from known sources using commonly available techniques. Preferably, alphavirus vectors with reduced cytotoxicity are used (see co-owned U.S. Ser. No. 08/679640). [0185]
  • DNA vector systems such as eukaryotic layered expression systems are also useful for expressing the nucleic acids of the invention. See WO 95/07994 for a detailed description of eukaryotic layered expression systems. Preferably, the eukaryotic layered expression systems of the invention are derived from alphavirus vectors and most preferably from Sindbis viral vectors. [0186]
  • Other viral vectors suitable for use in the present invention include those derived from poliovirus, for example ATCC VR-58 and those described in Evans et al. (1989) [0187] Nature 339:385 and Sabin et al. (1973) J. Biol. Standardization 1:115; rhinovirus, for example ATCC VR-1110 and those described in Arnold et al. (1990) J. Cell Biochem L401; pox viruses such as canary pox virus or vaccinia virus, for example ATCC VR-111 and ATCC VR-2010 and those described in Fisher-Hoch et al. (1989) Proc. Natl. Acad. Sci. 86:317; Flexner et al. (1989) Ann. NY Acad. Sci. 569:86; Flexner et al. (1990) Vaccine 8:17; in U.S. Pat. Nos. 4,603,112 and 4,769,330 and in WO 89/01973; SV40 virus, for example ATCC VR-305 and those described in Mulligan et al. (1979) Nature 277:108 and Madzak et al. (1992) J. Gen. Vir. 73:1533; influenza virus, for example ATCC VR-797 and recombinant influenza viruses made employing reverse genetics techniques as described in U.S. Pat. No. 5,166,057 and in Enami et al. (1990) Proc. Natl Acad. Sci. 87:3802-3805, Enami et al. (1991) J. Virol. 65:2711-2713; Luytjes et al. (1989) Cell 59:110 (see also McMicheal, (1983) NE J. Med. 309:13 and Yap et al. (1978) Nature 273:238 and Yap et al. (1979) Nature 2 77:108); human immunodeficiency virus as described in EP 0386882 and in Buchschacher et al. (1992) J. Vir. 66:2731; measles virus, for example ATCC VR-67 and VR-1247 and those described in EP 0440219; Aura virus, for example ATCC VR-368; Bebaru virus, for example ATCC VR-600 and ATCC VR-1240; Cabassou virus, for example ATCC VR-922; Chikungunya virus, for example ATCC VR-64 and ATCC VR-1241; Fort Morgan Virus, for example ATCC VR-924; Getah virus, for example ATCC VR-369 and ATCC VR-1243; Kyzylagach virus, for example ATCC VR-927; Mayaro virus, for example ATCC VR-66; Mucambo virus, for example ATCC VR-580 and ATCC VR-1244; Ndumu virus, for example ATCC VR-371; Pixuna virus, for example ATCC VR-372 and ATCC VR-1245; Tonate virus, for example ATCC VR-925; Triniti virus, for example ATCC VR-469; Una virus, for example ATCC VR-374; Whataroa virus, for example ATCC VR-926; Y-62-33 virus, for example ATCC VR-375; O'Nyong virus, Eastern encephalitis virus, for example ATCC VR-65f and ATCC VR-1242; Western encephalitis virus, for example ATCC VR-70, ATCC VR-1251, ATCC VR-622 and ATCC VR-1252; and coronavirus, for example ATCC VR-740 and those described in Hamre et al. (1966) Proc. Soc. Exp. Biol. Med. 121:190.
  • Delivery of the compositions of this invention into cells is not limited to the above mentioned viral vectors. Other delivery methods and media may be employed such as, for example, nucleic acid expression vectors, polycationic condensed DNA linked or unlinked to killed adenovirus alone, for example see U.S. Ser. No. 08/366,787, filed Dec. 30, 1994 and Curiel et al. (1989) [0188] Hum. Gene Ther. 3:147-154; ligand linked DNA, for example see Wu et al. (1989) J. Biol. Chem. 264:16985-16987, eucaryotic cell delivery vehicles cells, for example see U.S. Ser. No. 08/240,030, filed May 9, 1994, and U.S. Ser. No. 08/404,796, deposition of photopolymerized hydrogel materials, hand-held gene transfer particle gun, as described in U.S. Pat. No. 5,149,655, ionizing radiation as described in U.S. Pat. No. 5,206,152 and in WO 92/11033, nucleic charge neutralization or fusion with cell membranes. Additional approaches are described in Philip et al. (1994) Mol. Cell Biol. 14:2411-2418 and in Woffendin et al. (1994) Proc. Natl. Acad Sci. 91:1581-1585.
  • Particle mediated gene transfer may be employed, for example see U.S. Serial No. 60/023,867. Briefly, the sequence can be inserted into conventional vectors that contain conventional control sequences for high level expression, and then incubated with synthetic gene transfer molecules such as polymeric DNA-binding cations like polylysine, protamine, and albumin, linked to cell targeting ligands such as asialoorosomucoid, as described in Wu et al. (1987) [0189] J. Biol. Chem. 262:4429-4432, insulin as described in Hucked, (1990) Biochem. Pharmacol. 40:253-263, galactose as described in Plank et al. (1992) Bioconjugate Chem. 3:533-539, lactose or transferrin.
  • Naked DNA may also be employed. Exemplary naked DNA introduction methods are described in WO 90/11092 and U.S. Pat. No. 5,580,859. Uptake efficiency may be improved using biodegradable latex beads. DNA coated latex beads are efficiently transported into cells after endocytosis initiation by the beads. The method may be improved further by treatment of the beads to increase hydrophobicity and thereby facilitate disruption of the endosome and release of the DNA into the cytoplasm. [0190]
  • Liposomes that can act as gene delivery vehicles are described in U.S. Pat. No. 5,422,120, WO 95/13796, WO 94/23697, WO 91/144445 and EP 524,968. As described in co-owned U.S. Serial No. 60/023,867, on non-viral delivery, the nucleic acid sequences encoding a polypeptide can be inserted into conventional vectors that contain conventional control sequences for high level expression, and then be incubated with synthetic gene transfer molecules such as polymeric DNA-binding cations like polylysine, protamine, and albumin, linked to cell targeting ligands such as asialoorosomucoid, insulin, galactose, lactose, or transferrin. Other delivery systems include the use of liposomes to encapsulate DNA comprising the gene under the control of a variety of tissue-specific or ubiquitously-active promoters. Further non-viral delivery suitable for use includes mechanical delivery systems such as the approach described in Woffendin et al. (1994) [0191] Proc. Natl. Acad. Sci. U.S.A. 91 (24):11581-11585. Moreover, the coding sequence and the product of expression of such can be delivered through deposition of photopolymerized hydrogel materials. Other conventional methods for gene delivery that can be used for delivery of the coding sequence include, for example, use of hand-held gene transfer particle gun, as described in U.S. Pat. No. 5,149,655; use of ionizing radiation for activating transferred gene, as described in U.S. Pat. No. 5,206,152 and WO 92/11033.
  • Exemplary liposome and polycationic gene delivery vehicles are those described in U.S. Pat. Nos. 5,422,120 and 4,762,915; in WO 95/13796; WO 94/23697; and WO 91/14445; in EP 0524968; and in Stryer et al. (1975) [0192] Biochemistry, pp. 236-240, W. H. Freeman, San Francisco; Szoka et al. (1980) Biochim. Biophys. Acta. 600:1; Bayer, (1979) Biochim. Biophys. Acta. 550:464; Rivnay et al. (1987) Meth. Enzymol. 149:119; Wang et al. (1987) Proc. Natl. Acad. Sci. 84:7851; Plant (1989) Anal. Biochem. 176:420.
  • Pharmaceutical Compositions [0193]
  • In addition to the pharmaceutically acceptable carriers and salts described above, the following additional agents can be used with polynucleotide compositions. [0194]
  • A. Polypeptides [0195]
  • One example is polypeptides which include, without limitation: asioloorosomucoid (ASOR); transferrin, asialoglycoproteins; antibodies; antibody fragments; ferritin; interleukins; interferons, granulocyte, macrophage colony stimulating factor (M-CSF), stem cell factor and erythropoietin. Viral antigens, such as envelope proteins, can also be used. Also, proteins from other invasive organisms, such as the 17 amino acid peptide from the circumsporozoite protein of plasmodium falciparum known as RII. [0196]
  • B. Hormones, Vitamins, Etc. [0197]
  • Other groups that can be included are, for example: hormones, steroids, androgens, estrogens, thyroid hormone, or vitamins, folic acid. [0198]
  • C. Polyalkylenes, Polysaccharides, Etc. [0199]
  • Also, polyalkylene glycol can be included with the desired polynucleotides. In a preferred embodiment, the polyalkylene glycol is polyethylene glycol. In addition, mono-, di-, or polysaccarides can be included. In a preferred embodiment of this aspect, the polysaccharide is dextran or DEAE-dextran. Also, chitosan and poly(lactide-co-glycolide) [0200]
  • D. Lipids, and Liposomes [0201]
  • The desired polynucleotide can also be encapsulated in lipids or packaged in liposomes prior to delivery to the subject or to cells derived therefrom. [0202]
  • Lipid encapsulation is generally accomplished using liposomes which are able to stably bind or entrap and retain nucleic acid. The ratio of condensed polynucleotide to lipid preparation can vary but will generally be around 1:1 (mg DNA:micromoles lipid), or more of lipid. For a review of the use of liposomes as carriers for delivery of nucleic acids, see, Hug et al. (1991) [0203] Biochim. Biophys. Acta. 1097:1-17; Straubinger et al. (1983) Methods of Enzymology 101:512-527.
  • Liposomal preparations for use in the instant invention include cationic (positively charged), anionic (negatively charged) and neutral preparations. Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Felgner et al. (1987) [0204] Proc. Natl. Acad. Sci. U.S.A. 84:7413-7416); mRNA (Malone et al. (1989) Proc. Natl. Acad. Sci. U.S.A. 86:6077-6081); and purified transcription factors (Debs et al. 1990) J. Biol. Chem. 265:10189-10192), in functional form.
  • Cationic liposomes are readily available. For example, N[1-2,3-dioleyloxy)propyl]-N,N,N-triethylammonium (DOTMA) liposomes are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, N.Y. (See, also, Felgner et al. (1987) [0205] Proc. Natl. Acad. Sci. U.S.A. 84:7413-7416). Other commercially available liposomes include transfectace (DDAB/DOPE) and DOTAP/DOPE (Boerhinger). Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g., Szoka et al. (1978) Proc. Natl. Acad. Sci. U.S.A. 75:4194-4198; PCT Publication No. WO 90/11092 for a description of the synthesis of DOTAP (1,2-bis(oleoyloxy)-3-(trimethylammonio)propane) liposomes.
  • Similarly, anionic and neutral liposomes are readily available, such as from Avanti Polar Lipids (Birmingham, Ala.), or can be easily prepared using readily available materials. Such materials include phosphatidyl choline, cholesterol, phosphatidyl ethanolamine, dioleoyfphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), dioleoylphoshatidyl ethanolamine (DOPE), among others. These materials can also be mixed with the DOTMA and DOTAP starting materials in appropriate ratios. Methods for making liposomes using these materials are well known in the art. [0206]
  • The liposomes can comprise multilammelar vesicles (MLVs), small unilamellar vesicles (SUVs), or large unilamellar vesicles (LUVs). The various liposome-nucleic acid complexes are prepared using methods known in the art. See, e.g., Straubinger et al. (1983) [0207] Methods of Immunology 101:512-527; Szoka et al. (1978) Proc. Natl. Acad Sci. U.S.A. 75:4194-4198; Papahadjopoulos et al. (1975) Biochim. Biophys. Acta. 394:483; Wilson et al. (1979) Cell 17:77; Deamer et al. (1976) Biochim. Biophys. Acta. 443:629; Ostro et al. (1977) Biochem. Biophys. Res. Commun. 76:836; Fraley et al. (1979) Proc. Natl. Acad. Sci. U.S.A. 76:3348; Enoch et al. (1979) Proc. Natl. Acad. Sci. U.S.A. 76:145; Fraley et al. (1980) J. Biol. Chem. 255:10431; Szoka et al. (1978) Proc. Natl. Acad. Sci. U.S.A. 75:145; and Schaefer-Ridder et al. (1982) Science 215:166.
  • E. Liproproteins [0208]
  • In addition, lipoproteins can be included with the polynucleotide to be delivered. Examples of lipoproteins to be utilized include: chylomicrons, HDL, IDL, LDL, and VLDL. Mutants, fragments, or fusions of these proteins can also be used. Also, modifications of naturally occurring lipoproteins can be used, such as acetylated LDL. These lipoproteins can target the delivery of polynucleotides to cells expressing lipoprotein receptors. Preferably, if lipoproteins are included with the polynucleotide to be delivered, no other targeting ligand is included in the composition. [0209]
  • Naturally occurring lipoproteins comprises of a lipid and a protein portion. The protein portion is known as apoproteins. At the present, apoproteins A, B, C, D, and E have been isolated and identified. At least two of these contain several proteins, designated by Roman numerals, AI, AII, AIV, CI, CII, and CIII. [0210]
  • A lipoprotein can comprise more than one apoprotein. For example, naturally occurring chylomicrons comprises of A, B, C, and E; over time these lipoproteins lose A and acquire C and E apoproteins. VLDL comprises A, B, C, and E apoproteins; LDL comprises apoprotein B; and HDL comprises apoproteins A, C, and E. [0211]
  • The amino acids of these apoproteins are known and are described in, for example, Breslow (1985) [0212] Annu. Rev. Biochem. 54:699; Law et al. (1986) Adv. Exp. Med. Biol. 151:162; Chen et al. 1986) J. Biol. Chem. 261:12918; Kane et al. (1980) Proc. Natl. Acad. Sci. U.S.A. 77:2465; and Utermann et al. (1984) Hum. Genet. 65:232.
  • Lipoproteins contain a variety of lipids including triglycerides, cholesterol (free and esters), and phospholipids. The composition of the lipids varies in naturally occurring lipoproteins. For example, chylomicrons comprise mainly triglycerides. A more detailed description of the lipid content of naturally occurring lipoproteins can be found, for example, in [0213] Meth. Enzym. 128 (1986). The composition of the lipids are chosen to aid in conformation of the apoprotein for receptor binding activity. The composition of lipids can also be chosen to facilitate hydrophobic interaction and association with the polynucleotide binding molecule.
  • Naturally occurring lipoproteins can be isolated from serum by ultracentrifugation, for instance. Such methods are described in [0214] Meth. Enzy., Supra; Pitas et al. (1980) J. Biochem. 255:5454-5460; and Mahey et al. (1979) J. Clin. Invest. 64:743-750.
  • Lipoproteins can also be produced by in vitro or recombinant methods by expression of the apoprotein genes in a desired host cell. See, for example, Atkinson et al. (1986) [0215] Annu. Rev. Biophys. Chem. 15:403 and Radding et al. (1958) Biochim. Biophys. Acta. 30:443.
  • Lipoproteins can also be purchased from commercial suppliers, such as Biomedical Technologies, Inc., Stoughton, Mass., U.S.A. [0216]
  • Further description of lipoproteins can be found in Zuckermann et al., PCT. Appln. No. U.S. Ser. No. 97/14465. [0217]
  • F. Polycationic Agents [0218]
  • Polycationic agents can be included, with or without lipoprotein, in a composition with the desired polynucleotide to be delivered. [0219]
  • Polycationic agents, typically exhibit a net positive charge at physiological relevant pH and are capable of neutralizing the electrical charge of nucleic acids to facilitate delivery to a desired location. These agents have in vitro, ex vivo, and in vivo applications. Polycationic agents can be used to deliver nucleic acids to a living subject either intramuscularly, subcutaneously, etc. [0220]
  • The following are examples of useful polypeptides as polycationic agents: polylysine, polyarginine, polyomithine, and protamine. Other examples include histones, protamines, human serum albumin, DNA binding proteins, non-histone chromosomal proteins, coat proteins from DNA viruses, such as φX174, transcriptional factors also contain domains that bind DNA and therefore may be useful as nucleic acid condensing agents. Briefly, transcriptional factors such as C/CEBP, c-jun, c-fos, AP-1, AP-2, AP-3, CPF, Prot-1, Sp-1, Oct-1, Oct-2, CREP, and TFIID contain basic domains that bind DNA sequences. [0221]
  • Organic polycationic agents include: spermine, spermidine, and putrescine. [0222]
  • The dimensions and the physical properties of a polycationic agent can be extrapolated from the list above to construct other polypeptide polycationic agents or to produce synthetic polycationic agents. [0223]
  • Synthetic Polycationic Agents [0224]
  • Synthetic polycationic agents which are useful include, for example, DEAE-dextran, polybrene. Lipofectin, and lipofectAMINE are monomers that form polycationic complexes when combined with polynucleotides. See for example, Zuckermann et al., PCT Appln. U.S. Ser. No. 97/14465. [0225]
  • A polynucleotide composition can comprises therapeutically effective amount of a gene therapy vehicle, as the term is defined above. For purposes of the present invention, an effective dose will be from about 0.01 mg/kg to 50 mg/kg or 0.05 mg/kg to about 10 mg/kg of the DNA constructs in the individual to which it is administered. [0226]
  • Delivery Methods [0227]
  • Once formulated, the polynucleotide compositions of the invention can be administered (1) directly to the subject; (2) delivered ex vivo, to cells derived from the subject; or (3) in vitro for expression of recombinant proteins. The subjects to be treated can be mammals or birds. Also, human subjects can be treated. [0228]
  • Direct delivery of the compositions will generally be accomplished by injection, either subcutaneously, intraperitoneally, intravenously or intramuscularly or delivered to the interstitial space of a tissue. The compositions can also be administered into a tumor or lesion. Other modes of administration include oral and pulmonary administration, suppositories, and transdermal applications, needles, and gene guns or hyposprays. Dosage treatment may be a single dose schedule or a multiple dose schedule. [0229]
  • Methods for the ex vivo delivery and reimplantation of transformed cells into a subject are known in the art and described in e.g., International Publication No. WO 93/14778. Examples of cells useful in ex vivo applications include, for example, stem cells, particularly hematopoietic, lymph cells, macrophages, dendritic cells, or tumor cells. [0230]
  • Generally, delivery of nucleic acids for both ex vivo and in vitro applications can be accomplished by the following procedures, for example, dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei, all well known in the art. [0231]
  • EXAMPLES
  • The examples presented below are provided as a further guide to the practitioner of ordinary skill in the art, and are not to be construed as limiting the invention in any way. [0232]
  • Example 1 Cloning and Sequence Analysis of Myosin IXa
  • In order to identify novel transcripts in the human chromosomal region 15p22-23 which could be candidates for the Bardet-Biedl Sydrome gene that maps to this locus (BBS4; Bruford et al. (1997) [0233] Genomics 41:83-99) a BAC contig spanning this locus was constructed. A random clone sub-library was generated for each BAC. BAC clones forming a contig across the chromosome 15q22-23 region were cleaved with CviJI under conditions that produced partial restriction fragments ranging in size from ˜500 bp to ˜4 kb.
  • Fragments in the 1-2 kb range were isolated and cloned into the vector Perfectly Blunt™ (Novagen). For each BAC, 192 clones were randomly selected and sequenced using the ABI TAQ dye chemistry/system. [0234]
  • Comparison of these high throughput sample sequences originating from [0235] human chromosome 15 showed homology with 5 human EST clones (accession numbers AA287851, AA79085, AA235795, AA368273, HUMM9AA) and with myosin IXa (accession number L29148). Several clones were identified that showed significant homology to myr5 and human myosin IXb, and 100% identity to the entire 250 bp sequence known for human myosin IXa. Myosin IXa had previously been mapped to human chromosome 15q21-q25. Based on the information obtained from the comparisons of the myosin IXb sequence to the sequences of the inserts in these BAC sub-libraries, PCR primers were designed for Marathon™ 5′ and 3′ RACE™ reactions using Marathon™ prepared leukocyte, retina and testis cDNA (Clontech) as templates. Resulting fragments were cloned into pCRII (Invitrogen), sequenced and used for primer design for subsequent Marathon™ reactions. Nucleotide sequence for both strands of the partial cDNAs was obtained using the ABI TAQ dye terminator chemistry/system. Sequence comparison analysis was done using MASPAR and GeneWorks™ (Intelligenetics).
  • A sequence representing a transcript of 8473 bp was obtained. Human myosin IXa was mapped on the Stanford G3 radiation hybrid panel using oligos having homology to myosin IXa cDNA. Duplicate reactions consistently linked this region of myosin IXa to marker SHGC-31014 located on [0236] chromosome 15 in bin #50 confirming that the myosin IXa gene is located on human chromosome 15q21.
  • An ATG is present at base 243 and is in good context having an A at the −3 position and a G at +4 (Kozak et al. (1995)). This ATG initiates a single ORF that continues for 7643 bases to a stop signal (UGA) present at base 7886. Sequence analysis and exon mapping of the myo IXa transcript indicated that myosin IXa contains 42 exons and predicts a protein of 2548 amino acids with an estimated molecular mass of 283 kDa (FIG. 1), a size similar to the myosin IXb gene product (Wirth et al. (1996)). [0237]
  • The myosin IXa gene spans over 185 kb in the human genome (data not shown) and produces a transcript whose predicted protein contains the domains described for class IX unconventional myosins. To determine the similarity between family members, the predicted amino acid sequence of human myosin IXa, myosin IXb, and rat myr5 were compared. The N-terminal extension of myosin IXa shares 40% identity with the other class IX myosins. Myosin IXa had 82% identity with myosin IXb and myr5 within the head domain that is conserved among known myosin classes, although myosin IXa also contained an 18 residue insertion absent in the other class IX family members. The position designated as the second flexible loop (20/50 kDa junction; which is proposed to contact actin (Rayment et al. (993); Schroder et al. (1993)) and is the site of the second insertion in the head region of the class IX myosins, contained an additional 32 residues in myosin IXa in addition to having only 31% homology within the 120 residues shared with myosin IXb and myr5. [0238]
  • Within the neck region, myosin IXa contained two separate insertions of 33 and 18 residues compared to the other class IX family members. These insertions conform to the “IQ motif” and suggest that myosin IXa may bind 6 light chains of the clamodulin/EF hand superfamily (Mercer et al. 1991) instead of the 4 predicted to bind to myosin IXb and myr5. [0239]
  • The tail region of myosin IXa has 58% identity with the GAP domain found in myr5 and myosin IXb. However, the tail region located between the neck region and the GAP domain has numerous insertions not found in the other class IX family members that add an additional 327 residues compared to myr5 or 286 residues compared to myosin IXb. [0240]
  • Example 2 Expression of Myo IXa
  • To determine the expression profile of myosin IXa, human tissue Northern blots were hybridized with three different probes that represented the myosin IXa N-terminal extension plus a small portion of the conserved head domain, the GAP domain and the 3′ untranslated region. Briefly, human northern analysis was conducted on MTN Human, Human II and Human Brain II blots (Clontech). Probes corresponding to bases 9-1081, 6141-6831, and 7854-8304 were prepared using Amersham's Rediprime™ kit with [0241] 32PαdCTP, hybridized in Express Hybridization solution (Clontech) as described by the manufacturer and exposed to BioMax™ film (Amersham).
  • A transcript of ˜12.5 kb was visible in all tissues tested except liver. In addition, a transcript of ˜8.5 kb was present in testis and placenta. To ascertain whether the 4 kb difference between the two testis messages involved alternative splicing and/or polyadenylation at the 3′ region of the message, Marathon™ cDNA (Clontech) from testis, whole brain and cerebellum was amplified using a primer specific to the last exon (to bases 7704-7727) and the Marathon™ adaptor primer AP1. This PCR reaction produced a single band in all cases which corresponded to the product predicted from the cloned sequence. In addition, two brain cDNA libraries (frontal cortex and cerebellum (Stratagene)) were screened using the GAP domain probe. No evidence was found for either alternative splicing or polyadenylation at the 3′ end of the myosin IXa transcript. Alternative splicing within and/or between the remaining identified exons was ruled out by conducting PCR amplification of whole brain, cerebellum, retina and testis cDNA with primers that spanned intron/exon junctions. No differences in product sizes were seen between those tissues expressing only the 12.5 kb transcript and testis. This result was confirmed by conducting multiple hybridizations to a frontal cortex cDNA library using probes representing each portion of the myosin IXA transcript. Thus, the 4 kb difference between the two transcript sizes present in testis was not due to alternative splicing between the identified exons nor due to alternative polyadenylation and most likely resides in the 5′ untranslated region of the myosin IXa transcript. Therefore, the additional 4 kb present in the larger transcript is likely located in the 5′ UTR. [0242]
  • However, at least one alternative splicing event does occur within the myosin IXa coding region. During the cloning, two distinct bands were generated from PCR reactions conducted on leukocyte cDNA with primers specific to the head region. Sequence analysis of these bands indicated that a cryptic splice site exists at bases 375-380 and acts as a splice donor site, creating a [0243] truncated exon #2. Identical PCR reactions conducted on cDNA from testis and retina produced only the larger product. This data suggests that a second protein may be present in leukocytes, arising from an alternative splice within the second exon, which truncates the N-terminal extension and eliminates the ATTP binding domain of the myosin head region. No unique transcript for the predicted truncated protein was visible on Northern blots, presumably due to the decreased separation of molecules in the 12.0-12.5 kb size range.
  • Example 3 Developmental Expression of Myo IXa in a Mouse Model
  • A fragment specific to the mouse N-terminal extension of myosin IXa was obtained by conducting a PCR reaction on mouse genomic DNA using primers corresponding to bases 252.271 and bases 608-590 of the human myosin IXa sequence. This probe was prepared as described above and hybridized to mouse poly A[0244] + Northern blots prepared using standard methods from freshly dissected tissues. A single transcript of ˜9 kb was detected. Myosin IXa was first visible in RNA from 11.5 d embryos, absent from 13.5 d embryos and reappeared in 15.5 d embryos. Myosin IXa was also present in RNA obtained from 13.5 d old limb buds and brain. Adult tissues also showed a myo IXa transcript of ˜9 kb expressed in brain, heart, skeletal muscle, retina, leukocytes, lung, kidney, thymus and testis. No message was present in liver, a pattern identical with that seen in adult human tissues.
  • Example 4 Myo IXa Functions as a GTPase Activating Protein
  • Myr5 acts as a GTP activating protein on members of the rho subfamily of GTP binding proteins (Reinhard et al. (1995) [0245] EMBO J. 14:697-704). We determined that myosin IXa is also a functional GAP by making an expression construct that contained the predicted GAP domain (bases 6141 to 8035 of SEQ ID NO: 2) and conducting GAP assays with the small GTP binding proteins rhoA, ras, rac and Cdc42.
  • Example 5 Cloning and Sequence Analysis of CNGC-15 cDNA
  • A partial CNGC-15 cDNA, 1281 nucleotides in length was isolated from a human BAC contigs as described in Example 1. The cDNA sequence is shown in FIG. 3. The ATG codon at nucleotide 36 g initiates an open reading frame that contiues through the rest of the cDNA. Comparison of the nucleotide to Genbank, EST and patent databases showed that this partial cDNA has 76.2% homology to a mouse ion channel BCGN-3 mRNA (Genbank accession number AF064874) and lesser homology to mouse and human ion channels. The longest stretch of 100% homology with sequences in the databases was 44 nucleotides. [0246]
  • 1 3 1 2548 PRT Homo sapien 1 Met Asn Ile Asn Asp Gly Gly Arg Arg Arg Phe Glu Asp Asn Glu His 1 5 10 15 Thr Leu Arg Ile Tyr Pro Gly Ala Ile Ser Glu Gly Thr Ile Tyr Cys 20 25 30 Pro Ile Pro Ala Arg Lys Asn Ser Thr Ala Ala Glu Val Ile Glu Ser 35 40 45 Leu Ile Asn Lys Leu His Leu Asp Lys Thr Lys Cys Tyr Val Leu Ala 50 55 60 Glu Val Lys Glu Phe Gly Gly Glu Glu Trp Ile Leu Asn Pro Thr Asp 65 70 75 80 Cys Pro Val Gln Gln Met Met Leu Trp Pro Arg Met Ala Leu Glu Asn 85 90 95 Arg Leu Ser Gly Glu Asp Tyr Arg Phe Leu Leu Arg Glu Lys Asn Leu 100 105 110 Asp Gly Ser Ile His Tyr Gly Ser Leu Gln Ser Trp Leu Arg Val Thr 115 120 125 Glu Glu Arg Arg Arg Met Met Glu Arg Gly Phe Leu Pro Gln Pro Gln 130 135 140 Gln Lys Asp Phe Asp Asp Leu Cys Ser Leu Pro Asp Leu Asn Glu Lys 145 150 155 160 Thr Leu Leu Glu Asn Leu Arg Asp Arg Phe Lys His Glu Lys Ile Tyr 165 170 175 Thr Tyr Val Gly Ser Ile Leu Ile Val Ile Asn Pro Phe Lys Phe Leu 180 185 190 Pro Ile Tyr Asn Pro Lys Tyr Val Lys Met Tyr Asp Asn His Gln Leu 195 200 205 Gly Lys Pro Glu Pro His Ile Tyr Ala Val Ala Asp Val Ala Tyr His 210 215 220 Ala Met Leu Gln Arg Lys Lys Asn Gln Cys Ile Val Ile Ser Gly Glu 225 230 235 240 Ser Gly Ser Gly Lys Thr Gln Ser Thr Asn Phe Leu Ile His His Leu 245 250 255 Thr Ala Leu Ser Gln Lys Gly Phe Ala Ser Gly Val Glu Gln Ile Ile 260 265 270 Leu Gly Ala Gly Pro Val Leu Glu Ala Phe Gly Asn Ala Lys Thr Ala 275 280 285 His Asn Asn Asn Ser Ser Arg Phe Gly Lys Phe Ile Gln Val Asn Tyr 290 295 300 Gln Glu Thr Gly Thr Val Leu Gly Ala Tyr Val Glu Lys Tyr Leu Leu 305 310 315 320 Glu Lys Ser Arg Leu Val Tyr Gln Glu His Asn Glu Arg Asn Tyr His 325 330 335 Val Phe Tyr Tyr Leu Leu Ala Gly Ala Ser Glu Asp Glu Arg Ser Ala 340 345 350 Phe His Leu Lys Gln Pro Glu Glu Tyr His Tyr Leu Asn Gln Ile Thr 355 360 365 Lys Lys Pro Leu Arg Gln Ser Trp Asp Asp Tyr Cys Tyr Asp Ser Glu 370 375 380 Pro Asp Cys Phe Thr Val Glu Gly Glu Asp Leu Arg His Asp Phe Glu 385 390 395 400 Arg Leu Gln Leu Ala Met Glu Met Val Gly Phe Leu Pro Lys Thr Arg 405 410 415 Arg Gln Ile Phe Ser Leu Leu Ser Ala Ile Leu His Leu Gly Asn Ile 420 425 430 Cys Tyr Lys Lys Lys Thr Tyr Arg Asp Asp Ser Ile Asp Ile Cys Asn 435 440 445 Pro Glu Val Leu Pro Ile Val Ser Glu Leu Leu Glu Val Lys Glu Glu 450 455 460 Met Leu Phe Glu Ala Leu Val Thr Arg Lys Thr Val Thr Val Gly Glu 465 470 475 480 Lys Leu Ile Leu Pro Tyr Lys Leu Ala Glu Ala Val Thr Val Arg Asn 485 490 495 Ser Met Ala Lys Ser Leu Tyr Ser Ala Leu Phe Asp Trp Ile Val Phe 500 505 510 Arg Ile Asn His Ala Leu Leu Asn Ser Lys Asp Leu Glu His Asn Thr 515 520 525 Lys Thr Leu Ser Ile Gly Val Leu Asp Ile Phe Gly Phe Glu Asp Tyr 530 535 540 Glu Asn Asn Ser Phe Glu Gln Phe Cys Ile Asn Phe Ala Asn Glu Arg 545 550 555 560 Leu Gln His Tyr Phe Asn Gln His Ile Phe Lys Leu Glu Gln Glu Glu 565 570 575 Tyr Arg Thr Glu Gly Ile Ser Trp His Asn Ile Asp Tyr Ile Asp Asn 580 585 590 Thr Cys Cys Ile Asn Leu Ile Ser Lys Lys Pro Thr Gly Leu Leu His 595 600 605 Leu Leu Asp Glu Glu Ser Asn Phe Pro Gln Ala Thr Asn Gln Thr Leu 610 615 620 Leu Asp Lys Phe Lys His Gln His Glu Asp Asn Ser Tyr Ile Glu Phe 625 630 635 640 Pro Ala Val Met Glu Pro Ala Phe Ile Ile Lys His Tyr Ala Gly Lys 645 650 655 Val Lys Tyr Gly Val Lys Asp Phe Arg Glu Lys Asn Thr Asp His Met 660 665 670 Arg Pro Asp Ile Val Ala Leu Leu Arg Ser Ser Lys Asn Ala Phe Ile 675 680 685 Ser Gly Met Ile Gly Ile Asp Pro Val Ala Val Phe Arg Trp Ala Ile 690 695 700 Leu Arg Ala Phe Phe Arg Ala Met Val Ala Phe Arg Glu Ala Gly Lys 705 710 715 720 Arg Asn Ile His Arg Lys Thr Gly His Asp Asp Thr Ala Pro Cys Ala 725 730 735 Ile Leu Lys Ser Met Asp Ser Phe Ser Phe Leu Gln His Pro Val His 740 745 750 Gln Arg Ser Leu Glu Ile Leu Gln Arg Cys Lys Glu Glu Lys Tyr Ser 755 760 765 Ile Thr Arg Lys Asn Pro Arg Thr Pro Leu Ser Asp Leu Gln Gly Met 770 775 780 Asn Ala Leu Asn Glu Lys Asn Gln His Asp Thr Phe Asp Ile Ala Trp 785 790 795 800 Asn Gly Arg Thr Gly Ile Arg Gln Ser Arg Leu Ser Ser Gly Thr Ser 805 810 815 Leu Leu Asp Lys Asp Gly Ile Phe Ala Asn Ser Thr Ser Ser Lys Leu 820 825 830 Leu Glu Arg Ala His Gly Ile Leu Thr Arg Asn Lys Asn Phe Lys Ser 835 840 845 Lys Pro Ala Leu Pro Lys His Leu Leu Glu Val Asn Ser Leu Lys His 850 855 860 Leu Thr Arg Leu Thr Leu Gln Asp Arg Ile Thr Lys Ser Leu Leu His 865 870 875 880 Leu His Lys Lys Lys Lys Pro Pro Ser Ile Ser Ala Gln Phe Gln Ala 885 890 895 Ser Leu Ser Lys Leu Met Glu Thr Leu Gly Gln Ala Glu Pro Tyr Phe 900 905 910 Val Lys Cys Ile Arg Ser Asn Ala Glu Lys Leu Pro Leu Arg Phe Ser 915 920 925 Asp Val Leu Val Leu Arg Gln Leu Arg Tyr Thr Gly Met Leu Glu Thr 930 935 940 Val Gln Ile Arg Gln Ser Gly Tyr Ser Ser Lys Tyr Ser Phe Gln Asp 945 950 955 960 Phe Val Ser His Phe His Val Leu Leu Pro Arg Asn Ile Ile Pro Ser 965 970 975 Lys Phe Asn Ile Gln Asp Phe Phe Arg Lys Ile Asn Leu Asn Pro Asp 980 985 990 Asn Tyr Gln Val Gly Lys Thr Met Val Phe Leu Lys Glu Gln Glu Arg 995 1000 1005 Gln His Leu Gln Asp Leu Leu His Gln Glu Val Leu Arg Arg Ile Ile 1010 1015 1020 Leu Leu Gln Arg Trp Phe Arg Val Leu Leu Cys Arg Gln His Phe Leu 1025 1030 1035 1040 His Leu Arg Gln Ala Ser Val Ile Ile Gln Arg Phe Trp Arg Asn Tyr 1045 1050 1055 Leu Asn Gln Lys Gln Val Arg Asp Ala Ala Val Gln Lys Asp Ala Phe 1060 1065 1070 Val Met Ala Ser Ala Ala Ala Leu Leu Gln Ala Ser Trp Arg Ala His 1075 1080 1085 Leu Glu Arg Gln Arg Tyr Leu Glu Leu Arg Ala Ala Ala Ile Val Ile 1090 1095 1100 Gln Gln Lys Trp Arg Asp Tyr Tyr Arg Arg Arg His Met Ala Ala Ile 1105 1110 1115 1120 Cys Ile Gln Ala Arg Trp Lys Ala Tyr Arg Glu Ser Lys Arg Tyr Gln 1125 1130 1135 Glu Gln Arg Lys Lys Ile Ile Leu Leu Gln Ser Thr Cys Arg Gly Phe 1140 1145 1150 Arg Ala Arg Gln Arg Phe Lys Ala Leu Lys Glu Gln Arg Leu Arg Glu 1155 1160 1165 Thr Lys Pro Glu Val Gly Leu Val Asn Ile Lys Gly Tyr Gly Ser Leu 1170 1175 1180 Glu Ile Gln Gly Ser Asp Pro Ser Glu Trp Glu Asp Cys Ser Phe Asp 1185 1190 1195 1200 Asn Arg Ile Lys Ala Ile Glu Glu Cys Lys Ser Val Ile Glu Ser Asn 1205 1210 1215 Arg Ile Ser Arg Glu Ser Ser Val Asp Cys Leu Lys Glu Ser Pro Asn 1220 1225 1230 Lys Gln Gln Glu Arg Ala Gln Ser Gln Ser Gly Val Asp Leu Gln Glu 1235 1240 1245 Asp Val Leu Val Arg Glu Arg Pro Arg Ser Leu Glu Asp Leu His Gln 1250 1255 1260 Lys Lys Val Gly Arg Ala Lys Arg Glu Ser Arg Arg Met Arg Glu Leu 1265 1270 1275 1280 Glu Gln Ala Ile Phe Ser Leu Glu Leu Leu Lys Val Arg Ser Leu Gly 1285 1290 1295 Gly Ile Ser Pro Ser Glu Asp Arg Arg Trp Ser Thr Glu Leu Val Pro 1300 1305 1310 Glu Gly Leu Gln Ser Pro Arg Gly Thr Pro Asp Ser Glu Ser Ser Gln 1315 1320 1325 Gly Ser Leu Glu Leu Leu Ser Tyr Glu Glu Ser Gln Lys Ser Lys Leu 1330 1335 1340 Glu Ser Val Ile Ser Asp Glu Gly Asp Leu Gln Phe Pro Ser Pro Lys 1345 1350 1355 1360 Ile Ser Ser Ser Pro Lys Phe Asp Ser Arg Asp Asn Ala Leu Ser Ala 1365 1370 1375 Ser Asn Glu Thr Ser Ser Ala Glu His Leu Lys Asp Gly Thr Met Lys 1380 1385 1390 Glu Met Val Val Cys Ser Ser Glu Ser Ile Thr Cys Lys Pro Gln Leu 1395 1400 1405 Lys Asp Ser Phe Ile Ser Asn Ser Leu Pro Thr Phe Phe Tyr Ile Pro 1410 1415 1420 Gln Gln Asp Pro Leu Lys Thr Asn Ser Gln Leu Asp Thr Ser Ile Gln 1425 1430 1435 1440 Arg Asn Lys Leu Leu Glu Asn Glu Asp Thr Ala Gly Glu Ala Leu Thr 1445 1450 1455 Leu Asp Ile Asn Arg Glu Thr Arg Arg Tyr His Cys Ser Gly Lys Asp 1460 1465 1470 Gln Ile Val Pro Ser Leu Asn Thr Glu Ser Ser Asn Pro Val Leu Lys 1475 1480 1485 Lys Leu Glu Lys Leu Asn Thr Glu Lys Glu Glu Arg Gln Lys Gln Leu 1490 1495 1500 Gln Gln Gln Asn Glu Lys Glu Met Met Glu Gln Ile Arg Gln Gln Thr 1505 1510 1515 1520 Asp Ile Leu Glu Lys Glu Arg Lys Ala Phe Lys Thr Ile Glu Lys Pro 1525 1530 1535 Arg Ile Gly Glu Cys Leu Val Ala Pro Ser Ser Tyr Gln Ser Lys Gln 1540 1545 1550 Arg Val Glu Arg Pro Ser Ser Leu Leu Ser Leu Asn Thr Ser Asn Lys 1555 1560 1565 Gly Glu Leu Asn Val Leu Gly Ser Leu Ser Leu Lys Asp Ala Ala Leu 1570 1575 1580 Ala Gln Lys Asp Ser Ser Ser Ala His Leu Pro Pro Lys Asp Arg Pro 1585 1590 1595 1600 Val Thr Val Phe Phe Glu Arg Lys Gly Ser Pro Cys Gln Ser Ser Thr 1605 1610 1615 Val Lys Glu Leu Ser Lys Thr Asp Arg Met Gly Thr Gln Leu Asn Val 1620 1625 1630 Ala Cys Lys Leu Ser Asn Asn Arg Ile Ser Lys Arg Glu His Phe Arg 1635 1640 1645 Pro Thr Gln Ser Tyr Ser His Asn Ser Asp Asp Leu Ser Arg Glu Gly 1650 1655 1660 Asn Ala Arg Pro Ile Phe Phe Thr Pro Lys Asp Asn Met Ser Ile Pro 1665 1670 1675 1680 Leu Val Ser Lys Glu Ala Leu Asn Ser Lys Asn Pro Gln Leu His Lys 1685 1690 1695 Glu Asp Glu Pro Ala Trp Lys Pro Val Lys Leu Ala Gly Pro Gly Gln 1700 1705 1710 Arg Glu Thr Ser Gln Arg Phe Ser Ser Val Asp Glu Gln Ala Lys Leu 1715 1720 1725 His Lys Thr Met Ser Gln Gly Glu Ile Thr Lys Leu Ala Val Arg Gln 1730 1735 1740 Lys Ala Ser Asp Ser Asp Ile Arg Pro Gln Arg Ala Lys Met Arg Phe 1745 1750 1755 1760 Trp Ala Lys Gly Lys Gln Gly Glu Lys Lys Thr Thr Arg Val Lys Pro 1765 1770 1775 Thr Thr Gln Ser Glu Val Ser Pro Leu Phe Ala Gly Thr Asp Val Ile 1780 1785 1790 Pro Ala His Gln Phe Pro Asp Glu Leu Ala Ala Tyr His Pro Thr Pro 1795 1800 1805 Pro Leu Ser Pro Glu Leu Pro Gly Ser Cys Arg Lys Glu Phe Lys Glu 1810 1815 1820 Asn Lys Glu Pro Ser Pro Lys Ala Lys Arg Lys Arg Ser Val Lys Ile 1825 1830 1835 1840 Ser Asn Val Ala Leu Asp Ser Met His Trp Gln Asn Asp Ser Val Gln 1845 1850 1855 Ile Ile Ala Ser Val Ser Asp Leu Lys Ser Met Asp Glu Phe Leu Leu 1860 1865 1870 Lys Lys Val Asn Asp Leu Asp Asn Glu Asp Ser Lys Lys Asp Thr Leu 1875 1880 1885 Val Asp Val Val Phe Lys Lys Ala Leu Lys Glu Phe Arg Gln Asn Ile 1890 1895 1900 Phe Ser Phe Tyr Ser Ser Ala Leu Ala Met Asp Asp Gly Lys Ser Ile 1905 1910 1915 1920 Arg Tyr Lys Asp Leu Tyr Ala Leu Phe Glu Gln Ile Leu Glu Lys Thr 1925 1930 1935 Met Arg Leu Glu Gln Arg Asp Ser Leu Gly Glu Ser Pro Val Arg Val 1940 1945 1950 Trp Val Asn Thr Phe Lys Val Phe Leu Asp Glu Tyr Met Asn Glu Phe 1955 1960 1965 Lys Thr Ser Asp Cys Thr Ala Thr Lys Val Pro Lys Thr Glu Arg Lys 1970 1975 1980 Lys Arg Arg Lys Lys Glu Thr Asp Leu Val Glu Glu His Asn Gly His 1985 1990 1995 2000 Ile Phe Lys Ala Thr Gln Tyr Ser Ile Pro Thr Tyr Cys Glu Tyr Cys 2005 2010 2015 Ser Ser Leu Ile Trp Ile Met Asp Arg Ala Ser Val Cys Lys Leu Cys 2020 2025 2030 Lys Tyr Ala Cys His Lys Lys Cys Cys Leu Lys Thr Thr Ala Lys Cys 2035 2040 2045 Ser Lys Lys Tyr Asp Pro Glu Leu Ser Ser Arg Gln Phe Gly Val Glu 2050 2055 2060 Leu Ser Arg Leu Thr Ser Glu Asp Arg Thr Val Pro Leu Val Val Glu 2065 2070 2075 2080 Lys Leu Ile Asn Tyr Ile Glu Met His Gly Leu Tyr Thr Glu Gly Ile 2085 2090 2095 Tyr Arg Lys Ser Gly Ser Thr Asn Lys Ile Lys Glu Leu Arg Gln Gly 2100 2105 2110 Leu Asp Thr Asp Ala Glu Ser Val Asn Leu Asp Asp Tyr Asn Ile His 2115 2120 2125 Val Ile Ala Ser Val Phe Lys Gln Trp Leu Arg Asp Leu Pro Asn Pro 2130 2135 2140 Leu Met Thr Phe Glu Leu Tyr Glu Glu Phe Leu Arg Ala Met Gly Leu 2145 2150 2155 2160 Gln Glu Arg Lys Glu Thr Ile Arg Gly Val Tyr Ser Val Ile Asp Gln 2165 2170 2175 Leu Ser Arg Thr His Leu Asn Thr Leu Glu Arg Leu Ile Phe His Leu 2180 2185 2190 Val Arg Ile Ala Leu Gln Glu Asp Thr Asn Arg Met Ser Ala Asn Ala 2195 2200 2205 Leu Ala Ile Val Phe Ala Pro Cys Ile Leu Arg Cys Pro Asp Thr Thr 2210 2215 2220 Asp Pro Leu Gln Ser Val Gln Asp Ile Ser Lys Thr Thr Thr Cys Val 2225 2230 2235 2240 Glu Leu Ile Val Val Glu Gln Met Asn Lys Tyr Lys Ala Arg Leu Lys 2245 2250 2255 Asp Ile Ser Ser Leu Glu Phe Ala Glu Asn Lys Ala Lys Thr Arg Leu 2260 2265 2270 Ser Leu Ile Arg Arg Ser Met Gly Lys Gly Arg Ile Arg Arg Gly Asn 2275 2280 2285 Tyr Pro Gly Pro Ser Ser Pro Val Val Val Arg Leu Pro Ser Val Ser 2290 2295 2300 Asp Val Ser Glu Glu Thr Leu Thr Ser Glu Ala Ala Met Glu Thr Asp 2305 2310 2315 2320 Ile Thr Glu Gln Gln Gln Ala Ala Met Gln Gln Glu Glu Arg Val Leu 2325 2330 2335 Thr Glu Gln Ile Glu Asn Leu Gln Lys Glu Lys Glu Glu Leu Thr Phe 2340 2345 2350 Glu Met Leu Val Leu Glu Pro Arg Ala Ser Asp Asp Glu Thr Leu Glu 2355 2360 2365 Ser Glu Ala Ser Ile Gly Thr Ala Asp Ser Ser Glu Asn Leu Asn Met 2370 2375 2380 Glu Ser Glu Tyr Ala Ile Ser Glu Lys Ser Glu Arg Ser Leu Ala Leu 2385 2390 2395 2400 Ser Ser Leu Lys Thr Ala Gly Lys Ser Glu Pro Ser Ser Lys Leu Arg 2405 2410 2415 Lys Gln Leu Lys Lys Gln Gln Asp Ser Leu Asp Val Val Asp Ser Ser 2420 2425 2430 Val Ser Ser Leu Cys Leu Ser Asn Thr Ala Ser Ser His Gly Thr Arg 2435 2440 2445 Lys Leu Phe Gln Ile Tyr Ser Lys Ser Pro Phe Tyr Arg Ala Ala Ser 2450 2455 2460 Gly Asn Glu Ala Leu Gly Met Glu Gly Pro Leu Gly Gln Thr Lys Phe 2465 2470 2475 2480 Leu Glu Asp Lys Pro Gln Phe Ile Ser Arg Gly Thr Phe Asn Pro Glu 2485 2490 2495 Lys Gly Lys Gln Lys Leu Lys Asn Val Lys Asn Ser Pro Gln Lys Thr 2500 2505 2510 Lys Glu Thr Pro Glu Gly Thr Val Met Ser Gly Arg Arg Lys Thr Val 2515 2520 2525 Asp Pro Asp Cys Thr Ser Asn Gln Gln Leu Ala Leu Phe Gly Asn Asn 2530 2535 2540 Glu Phe Met Val 2545 2 8473 DNA Homo sapien 2 gcgtccgctc gcccggaccc tgaggctgct gggcccaccc tcccggaacc gtccgaccct 60 cggtggcctc ggctcgttct gccatctccg gtcctaccct ggggcggagg gtggaaggca 120 gcttccgtcg aagaggaggg ggctgcggtg gccaccgcgg cggagcccga gttattttac 180 caagaaaatg gtttgcacga ctttgaacat atactatcca tgctgatggg acaggatcca 240 atatgaatat aaatgatgga ggaagacgac gctttgaaga taatgaacat acattacgga 300 tatatcctgg ggctatttca gaagggacaa tctactgtcc gattcctgcc agaaaaaact 360 ccacagctgc tgaggtgatt gagtctctta taaacaaact tcatcttgac aaaacaaaat 420 gttacgttct agcagaggta aaggaatttg gtggagaaga atggattctc aatccaacag 480 attgtccagt tcagcaaatg atgctgtggc cccgaatggc tctggaaaat cgcttaagtg 540 gagaagacta ccgcttcctt ctgagagaga aaaaccttga tggatcaatc cattatggta 600 gcctgcagtc atggctacgg gtaacagaag aacgtcgcag gatgatggaa cggggttttc 660 ttccacagcc tcaacagaaa gactttgatg atttatgtag tttacctgat ttgaatgaga 720 aaactctctt agaaaaccta cgagatcgct ttaagcatga aaaaatttat acctatgttg 780 gcagtattct aatagttatt aacccattca agtttcttcc tatttataac cccaaatatg 840 tcaaaatgta tgataaccac caactgggaa aacctgagcc ccacatttat gctgtggctg 900 atgtagctta tcatgccatg cttcagcgca aaaagaatca gtgcatcgtg atttcaggag 960 agagtggttc tgggaagact caaagcacaa actttcttat tcaccacctt actgctctca 1020 gtcagaaagg atttgccagt ggagtagaac agattattct tggagctgga ccagtacttg 1080 aggcctttgg aaatgcaaag acagctcata ataacaattc aagtcgtttt gggaagttta 1140 ttcaagtaaa ttaccaggaa acaggcactg tacttggtgc ctatgttgaa aaatatctac 1200 tggagaagtc cagactcgtt tatcaggagc ataatgaacg gaactatcat gtattctatt 1260 acctcctggc aggagcaagt gaagatgaga gatcagcatt ccatcttaag caaccagagg 1320 aatatcatta tctcaatcag ataacaaaga aacccctcag acagagctgg gatgattatt 1380 gctatgactc tgagccggat tgcttcacgg tggaaggaga agatttgaga catgactttg 1440 agcgcctaca acttgccatg gaaatggtag gatttcttcc caagacacga agacagattt 1500 tctctcttct ctcagccata ctacatttgg gtaatatctg ttacaaaaag aagacatacc 1560 gggatgactc cattgatatc tgtaatcctg aagttctgcc tattgtctca gaattattag 1620 aggttaaaga agagatgcta tttgaagcat tagttacaag gaagacggtg acagtgggag 1680 aaaagcttat tttgccatac aagttggcag aggctgtgac agtgaggaac tccatggcta 1740 agtctctgta tagtgccctg tttgactgga tagtttttcg aattaatcat gcacttctga 1800 atagtaaaga tttagagcat aataccaaga cattgtctat tggtgttctt gatatttttg 1860 ggtttgaaga ttatgaaaat aacagctttg aacagttctg tattaatttt gctaatgaac 1920 gtttacagca ctactttaat cagcatatct ttaaattgga acaagaggaa tatagaactg 1980 aaggtatcag ctggcacaac atagattaca ttgataatac ctgctgcata aatcttatta 2040 gcaaaaaacc aacaggactg cttcatcttt tggatgaaga aagcaacttt ccacaggcta 2100 caaatcaaac attgctagac aagtttaagc atcaacatga agataattct tacatcgaat 2160 ttccagccgt gatggagcct gctttcatta taaaacatta tgctggaaaa gtaaaatatg 2220 gggtaaagga tttccgggaa aaaaatacag atcatatgcg cccagacatt gtagctcttc 2280 tgagaagcag caagaatgca tttatctctg ggatgattgg aattgatcct gtagctgttt 2340 tccgatgggc aattctccga gcttttttca gagccatggt tgctttcagg gaagctggga 2400 aaagaaacat tcacagaaaa actggacatg atgatacagc gccatgtgca attttgaaaa 2460 gtatggatag ttttagcttt ctccaacacc cagtccacca gaggagctta gagattctgc 2520 agagatgcaa ggaagagaag tacagtataa cccggaaaaa tcccagaaca cctctttctg 2580 atctccaggg catgaatgct ctaaatgaaa aaaaccaaca tgatacattt gatattgcct 2640 ggaatggcag aactgggatt cgccagagca gactatcaag tggcacctcc ttgcttgata 2700 aagatggaat atttgctaat tcaactagca gcaaactcct ggagagagcc catggaattc 2760 tcacgagaaa caaaaatttc aaatccaagc ctgcccttcc aaagcacttg ctagaagtaa 2820 attctttaaa gcacctgaca agactgacac tacaagatcg cattaccaag tctcttcttc 2880 atttacacaa gaagaaaaaa cctcccagca tcagtgccca gtttcaggca tcattaagca 2940 agctaatgga aacacttggt caagcagaac catattttgt aaaatgcatt cgctctaatg 3000 ctgaaaagct gcccttaagg ttcagtgatg tcttggtact tagacagctt cgatacaccg 3060 ggatgctgga aacagttcaa attcgccaat caggatacag ctccaaatat tctttccagg 3120 attttgtgag ccacttccat gtacttcttc cccgaaatat tattccatcc aaatttaaca 3180 ttcaggattt cttcaggaaa ataaatctta atccagataa ttatcaagtt ggaaaaacca 3240 tggtctttct aaaggagcag gaacgacagc acttacaaga tctgcttcac caagaggtgc 3300 tccgcagaat catattgttg cagcgatggt tcagggtctt gctgtgtagg cagcatttcc 3360 tccatctgag acaagcatct gttattatcc agagattctg gaggaattac ctaaatcaga 3420 agcaagtcag agatgcagct gtgcagaagg atgcttttgt tatggctagt gcagctgctc 3480 ttctccaagc ttcctggcgt gctcacttag agaggcagcg gtacttggag ttacgggctg 3540 cagccatcgt tatccagcag aaatggagag attactatag gcgcaggcac atggctgcta 3600 tttgcataca agcaagatgg aaagcctaca gggaaagtaa aaggtaccaa gaacaaagga 3660 aaaaaattat ccttttgcaa tcaacatgta gaggattcag agcaagacaa agatttaaag 3720 ctttaaaaga acaaaggcta agagaaacaa agccagaagt tggattggtg aatattaagg 3780 gatatggatc tctggaaatt cagggttcag acccttcaga atgggaggat tgttcttttg 3840 acaacagaat aaaagccata gaggaatgta aatctgtaat agagagtaat cgaattagcc 3900 gtgaaagttc agtggactgc ttgaaggagt caccaaacaa gcagcaggag agagcccaaa 3960 gccagagtgg tgtggacttg caggaagatg tgcttgtaag agagagaccc aggtccttgg 4020 aggatctcca tcagaaaaaa gtaggccggg ctaagagaga aagtaggaga atgagagaac 4080 tagagcaagc tatatttagc ttagaattgc tgaaagttcg ttctcttggt ggtatttctc 4140 cttcagagga tcgcagatgg tctacagaat tggtgcctga aggccttcag tctccacggg 4200 gtacacctga tagtgagagc tctcaaggaa gcttggaact tctgagctat gaggaaagcc 4260 aaaagagcaa actagagtct gtcatttcag atgaaggaga cttgcagttt ccatcaccta 4320 agatatccag cagtccaaaa tttgattcac gggacaatgc cctcagtgcc tcaaatgaga 4380 ctagcagtgc agagcatttg aaggatggaa ctatgaagga aatggtggtc tgcagttctg 4440 agtctattac ctgtaaacca cagctgaaag actccttcat ttcaaatagt ctacctactt 4500 ttttttatat cccccaacaa gacccactga aaacaaattc ccaactagac acaagtatcc 4560 aaagaaacaa actattggaa aatgaagaca cagcggggga agctcttact ttggatatca 4620 acagggaaac tagaaggtat cactgctcag gaaaagatca gattgttcct tctttgaata 4680 cagagtcttc taatcctgtg cttaagaagt tagaaaagct aaacactgag aaggaagaaa 4740 ggcaaaaaca gttgcagcaa cagaatgaaa aagagatgat ggaacagatt cgccagcaaa 4800 cagatatttt agagaaggag cgcaaagcct tcaagacaat tgaaaagcca agaattggag 4860 agtgtttggt ggcaccatct tcctatcagt caaagcaaag agtagagagg ccatcctctc 4920 tcctcagctt aaatacctca aataagggag aacttaatgt actggggtcc ctatcattaa 4980 aagatgcagc tcttgcccaa aaagacagtt cctctgctca cttaccccca aaggaccgac 5040 ctgtcaccgt gttctttgaa agaaaaggaa gtccatgcca atctagtact gtcaaggaat 5100 tatccaagac agacagaatg ggcacccagc tgaatgtagc ctgtaaactc tcaaataatc 5160 gcatttcaaa aagagaacac tttaggccaa ctcagtctta cagccacaat tctgatgacc 5220 tttccagaga gggaaatgct aggcccattt tcttcactcc aaaggataat atgagtattc 5280 cccttgtcag caaagaagcc ttaaacagta aaaatcctca actccataaa gaagatgaac 5340 cagcatggaa acctgtgaag ttagctgggc caggccaaag agagacatca cagcgatttt 5400 cgtcagttga tgaacaagca aaacttcata agactatgtc tcaaggagag attaccaagt 5460 tggcagtgag acagaaggct tcagattcag atataagacc tcagagagct aagatgagat 5520 tctgggccaa agggaaacaa ggggagaaga agactaccag agtgaaacct actacccagt 5580 cagaggtttc gccactcttt gcaggcacag atgtgattcc agctcatcag tttccagatg 5640 aattagctgc atatcaccca acacctcctt tgagcccaga actgcccggc agttgccgga 5700 aggaattcaa agagaacaaa gaaccttctc caaaggctaa gcgcaagcga agtgtgaaga 5760 ttagcaacgt ggctttggat tctatgcatt ggcaaaatga ctctgtccag atcatagcaa 5820 gtgtcagtga tttaaaaagc atggatgaat ttcttctgaa aaaggtgaat gacctagata 5880 atgaagacag caagaaggat acactagtgg atgttgtatt taaaaaagcc ctgaaggaat 5940 ttcggcagaa tatcttcagc ttttattcat ctgcattggc gatggatgat gggaaaagca 6000 tacggtataa agacctctat gcactatttg aacagattct ggaaaagacg atgaggcttg 6060 agcagcgtga ttcactgggt gaatctccag tgagagtttg ggtcaacact tttaaagtgt 6120 ttttagatga atatatgaat gaattcaaga cttcagattg cacagccaca aaggtgccaa 6180 aaacagaaag aaagaaaaga aggaaaaagg aaactgattt ggtggaagaa cacaatggtc 6240 acatctttaa agccacccaa tatagcatcc ctacatactg tgaatactgt tcttctttga 6300 tatggataat ggaccgagcc tctgtttgca aattatgcaa gtatgcttgc cataagaagt 6360 gctgtctgaa aaccacagcc aagtgctcta aaaagtatga tccagagctg tcatctcgac 6420 aatttggggt tgaactgtcc cgtttgacca gtgaagaccg aactgttcct ttagtagtgg 6480 aaaagctcat aaactacatt gaaatgcatg gactgtatac agaaggtatt tatcgaaagt 6540 ctggttcgac taataaaatc aaggagcttc ggcagggtct agatacagat gctgagagtg 6600 taaatctaga tgactataac atacacgtca ttgcaagtgt attcaaacaa tggcttcgag 6660 atttgcccaa tcctctcatg acctttgaac tctatgagga atttcttcga gctatgggcc 6720 ttcaggagag gaaggagaca atccgtggtg tatactctgt gattgatcaa ctctcccgaa 6780 ctcatctcaa tacactggaa cgcctcatct ttcatctagt caggattgct ctgcaggaag 6840 acactaatcg aatgtctgct aatgctttgg ccattgtgtt tgcgccctgc attctccgct 6900 gccctgacac cactgaccca ctacaaagtg tacaggacat cagtaagact accacttgtg 6960 tggaactgat tgttgtggaa caaatgaata aatacaaggc tcgtctcaaa gatatcagta 7020 gcttggaatt tgctgagaat aaggcaaaga ccaggttgtc actgattcgt agatcaatgg 7080 gaaaggggcg tattcgtcga ggaaactatc caggtccatc gtctcctgtt gtagttcggt 7140 tgccttctgt gtctgatgtc tcagaggaga ccttgactag tgaggcagcc atggagactg 7200 acatcacaga acagcagcaa gcagctatgc agcaggagga gagagtactg actgagcaga 7260 ttgagaacct acagaaggag aaggaggagc taacatttga gatgcttgta ctggaacccc 7320 gtgcctctga tgatgaaacc cttgagtctg aggcctccat tgggactgct gatagctcag 7380 agaatttgaa tatggagtct gaatatgcta tctctgagaa atcagaaaga agcttagccc 7440 ttagctccct gaagacagct ggcaagtctg aaccttccag caagttgcga aagcaactta 7500 aaaagcagca agactcttta gatgtcgtgg actcttcggt ctcctcttta tgtctgtcta 7560 acacggcatc atctcatggg accagaaaac tatttcagat ttattccaaa tctccattct 7620 accgagctgc ctcaggtaat gaggccctgg gaatggaagg accattgggc cagaccaaat 7680 tcctggaaga caagcctcag ttcatcagca gaggaacctt caacccggaa aagggcaaac 7740 aaaaattaaa gaatgtgaaa aactcacctc agaaaaccaa agagacccca gaggggacag 7800 tcatgtctgg ccgcagaaaa actgtggacc cagactgcac ctccaaccaa cagctagcac 7860 tctttggaaa taatgaattt atggtctgaa ccggcagatg tgtgtccctc cgtggctaca 7920 gagtggtaaa caaatctcac ctttggggct gcgtttcatc acctcgtcca caatagtcaa 7980 tcctaattgt ggtcctgcct cttttctaag catatggcta agactgtatg tgctgaattc 8040 ctgggcctcc tgcagaagca gaaagcctgc tggggatggt gccagctgtg ccttggctgt 8100 tgtatttgaa ttgagatttt tactatacaa agccacctag ggcctgggga tttgggtcag 8160 ttgtagttgc ctctccccca ccctcttttc ccttcccaaa ggtgggtgtt gaactagggg 8220 ggatattgct gtcctgaggg accctctcat ttctgacatt tgaagaaaac gtataaatct 8280 ttcttaaccg tgaaagcaaa agcctttggg tttattttgg gatagttagg agctagggta 8340 gaatataatt tttttccaaa aacttactta caaacaaaaa gcctaatccc tctattttaa 8400 gatttctgaa aaaacactcc atgttatatt ctggggaaag caaaaacaaa aaaaaaaaaa 8460 aaaaaaaaaa aaa 8473 3 1307 DNA Homo sapien misc_feature (1)...(1307) n = A,T,C or G 3 gaattcgggc ttccatcctt aataggaact caatnntagg gctngggcgg ccgcccgggc 60 aggtgcgcga acaggagagg gtcaagtcgg ccggattttg gattatccac ccctacagtg 120 acttcagatt ttactgggac ctgaccatgc tgctgctgat ggtgggaaac ctgattatca 180 ttcctgtggg catcaccttc ttcaaggatg agaacaccac accctggatt gtcttcaatg 240 tggtgtcaga cacattcttc ctcatcgact tggtcttcaa cttccgcaca gggatcgtgg 300 tggaggacaa cacagagatc atcctggacc cgcagcggat taaaatgaag tacctgaaaa 360 gctggttcat ggtagatttc atttcctcca tcccccgtgg aaaacatctt cctcattgtg 420 gagacaggca tcgactcgga ggtctacaag actgcccggg ccctgcgcat tgtccgcttc 480 acgaagatcc tcagcctctt acgcctgtta cgcctctccc gcctcattcg atatattcac 540 cagtgggaag agatcttcca catgacctac gacctggcca gcgccgtggt gcgcatcgtg 600 aacctcatcg gcatgatgct cctgctctgc cactgggacg gctgcctgca gttcctggta 660 cccatgctac aggacttccc tgacgactgc tgggtgtcca tcaacaacat ggtgaacaac 720 tcctggggga agcagtactc ctacgcgctc ttcaaggcca tgagccacat gctgtgcatc 780 ggctacgggc ggcaggcgcc cgtgggcatc tccgacgtct ggctcaccat gctcagcatg 840 atcgtgggtg ccacctgcta cgccatgttc attggccacg ccactgccct catccagtcc 900 ctggactcct cccggcgcca gtaccaggaa aagtacaagc aggtggagca gtacatgtcc 960 tttcacaagc tcccgcccga cacccggcag cgcatccacg actactacga gcaccgctac 1020 cagggcaaga tgttcgacga ggagagcacc ctgggcgagc taagcgagcc cctgcgggag 1080 gagatcatca actttaactg tcggaagctg gtggcctcca tgccactgtt tgccaatgcg 1140 gaccccaact tcgtgacgtc catgctgacc aagctgcgtt tcgaggtctt ccagcctggg 1200 gactacatca tccgggaagg caccattggc aagaagatgt acttcatcca gcatggcgtg 1260 gtcagcgtgc tcaccaaggg caacaaggag accaagaagc cgaattc 1307

Claims (31)

That which is claimed:
1. A native Myosin IXa polypeptide substantially free of other human proteins.
2. An active fragment or derivative of a Myosin IXa polypeptide.
3. A Myosin IXa polypeptide comprising an amino acid sequence selected from the group consisting of:
a) SEQ IDNO: 1;
b) residues 1-146 of SEQ ID NO: 1;
c) residues 147-719 of SEQ ID NO: 1;
d) residues 239-246 of SEQ ID NO: 1;
e) residues 720-835 of SEQ ID NO: 1;
f) residues 872-1166 of SEQ ID NO: 1;
g) residues 2074-2219 of SEQ ID NO: 1;
h) a polypeptide having at least about 61% homology to SEQ ID NO: 1;
i) a polypeptide at least about 95% homology to an amino acid sequence of b), c), d), e), f) or g); and
j) a polypeptide of at least about 90 contiguous residues of SEQ ID NO: 1.
4. A fusion protein comprising the Myosin IXa polypeptide of claim 3, or fragment thereof, and a polypeptide of interest.
5. A native CNGC-15 polypeptide substantially free of other human proteins.
6. An active fragment or derivative of a CNGC-15 polypeptide.
7. A CNGC-15 polypeptide comprising an amino acid sequence selected from the group consisting of:
a) a polypeptide encoded by SEQ ID NO: 3;
b) a polypeptide encoded by nucleotides 369-1307 of SEQ ID NO: 3;
c) a polypeptide having at least about 80% homology to the polypeptide encoded by nucleotides 369-1307 of SEQ ID NO: 3; and
d) a polypeptide of at least about 20 contiguous residues of the polypeptide encoded by nucleotides 369-1307 of SEQ ID NO: 3.
8. A fusion protein comprising the CNGC-15 polypeptide of claim 7, or fragment thereof, and a polypeptide of interest.
9. A nucleotide sequence encoding the fusion protein of claim 4.
10. A nucleotide sequence encoding the fusion protein of claim 8.
11. An isolated nucleotide sequence selected from the group consisting of:
a) a nucleotide sequence encoding a native Myosin IXa polypeptide;
b) a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 1;
c) the nucleotide sequence of SEQ ID NO: 2;
d) a nucleotide sequence comprising at least about 390 contiguous bases of SEQ ID NO: 2;
e) a nucleotide sequence comprising nucleotides 243-7085 of SEQ ID NO: 2;
f) a nucleotide sequence comprising nucleotides 243-680 of SEQ ID NO: 2;
g) a nucleotide sequence comprising nucleotides 683-2399 of SEQ ID NO: 2;
h) a nucleotide sequence comprising nucleotides 959-977 of SEQ ID NO: 2;
i) a nucleotide sequence comprising nucleotides 2402-2747 of SEQ ID NO: 2;
j) a nucleotide sequence comprising nucleotides 3158-3740 of SEQ ID NO: 2; and
k) a nucleotide sequence comprising nucleotides 6473-6899 of SEQ ID NO: 2;
12. An isolated nucleotide sequence selected from the group consisting of:
a) a nucleotide sequence encoding a native CNGC-15 polypeptide;
b) a nucleotide sequence encoding the polypeptide encoded by nucleotides 369-1307 of SEQ ID NO: 3;
c) the nucleotide sequence of SEQ ID NO: 3;
d) a nucleotide sequence comprising at least 45 contiguous nucleotides of the sequence of SEQ ID NO: 3; and
e) a nucleotide sequence that hybridizes under stringent conditions to a denatured DNA having a nucleotide sequence of a), b), c) or d).
13. An expression vector comprising the nucleotide sequence of claim 9 wherein said nucleotide sequence is operably linked to a promoter.
14. An expression vector comprising the nucleotide sequence of claim 10 wherein said nucleotide sequence is operably linked to a promoter.
15. An expression vector comprising the nucleotide sequence of claim 11 wherein said nucleotide sequence is operably linked to a promoter.
16. An expression vector comprising the nucleotide sequence of claim 12 wherein said nucleotide sequence is operably linked to a promoter.
17. A transformed cell having stably incorporated into its genome the expression vector of claim 13.
18. A transformed cell having stably incorporated into its genome the expression vector of claim 14.
19. A transformed cell having stably incorporated into its genome the expression vector of claim 15.
20. A transformed cell having stably incorporated into its genome the expression vector of claim 16.
21. A method for producing a Myosin IXa polypeptide or a derivative thereof, comprising:
a) providing a host cell stably transformed with an expression vector comprising a promoter operably linked to a nucleotide sequence encoding said Myosin IXa polypeptide or derivative thereof; and
b) culturing the host cell under conditions which allow expression of said polypeptide.
22. A method for producing a CNGC-15 polypeptide or a derivative thereof, comprising:
a) providing a host cell stably transformed with an expression vector comprising a promoter operably linked to a nucleotide sequence encoding said CNGC-15 polypeptide or derivative thereof; and
b) culturing the host cell under conditions which allow expression of said polypeptide.
23. A method for detecting an unconventional Myosin polynucleotide comprising:
a) providing a nucleic acid probe which hybridizes a nucleotide sequence of claim 11;
b) contacting the probe with a sample of polynucleotides under hybridizing conditions to form a duplex; and
c) detecting said duplexes.
24. A method for detecting a CNGC polynucleotide comprising:
a) providing a nucleic acid probe which hybridizes a nucleotide sequence of claim 12;
b) contacting the probe with a sample of polynucleotides under hybridizing conditions to form a duplex; and
c) detecting said duplexes.
25. An antibody specific for a CNGC-15 or Myosin IXa polypeptide.
26. A method for detecting a CNGC-15 polypeptide comprising:
a) providing an antibody that binds to a CNGC-15 polypeptide;
b) contacting the antibody to a sample under binding conditions to form a duplex; and
c) detecting said duplexes.
27. A method for detecting a Myosin IXa polypeptide comprising:
a) providing an antibody that binds to a Myosin IXa polypeptide;
b) contacting the antibody to a sample under binding conditions to form a duplex; and
c) detecting said duplexes.
28. A method for treating Bardet-Biedl Syndrome, Usher Syndrome, hearing loss, retinis pigmentosa, obesity, hypogonadism, sterility, polydactyly, brachydactyly, syndactyly, mental retardation, renal abnormalities, hypertension, diabetes and cardiovascular abnormalities, comprising administering a therapeutically effective amount of a CNGC-15 polypeptide to a subject in need of such treatment.
29. A method for treating Bardet-Biedl Syndrome, Usher Syndrome, hearing loss, retinis pigmentosa, obesity, hypogonadism, sterility, polydactyly, brachydactyly, syndactyly, mental retardation, renal abnormalities, hypertension, diabetes and cardiovascular abnormalities, comprising administering a therapeutically effective amount of a Myosin-IXA polypeptide to a subject in need of such treatment.
30. A composition comprising a CNGC-15 polypeptide and a pharmaceutically acceptable carrier.
31. A composition comprising a Myosin IXa polypeptide and a pharmaceutically acceptable carrier.
US09/851,682 1997-10-15 2001-05-08 Myosin IXa and cyclic nucleotide gated channel-15 (CNGC-15) polynucleotides, polypeptides, compositions, methods, and uses thereof Abandoned US20020091248A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US09/851,682 US20020091248A1 (en) 1997-10-15 2001-05-08 Myosin IXa and cyclic nucleotide gated channel-15 (CNGC-15) polynucleotides, polypeptides, compositions, methods, and uses thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US6285897P 1997-10-15 1997-10-15
US6224197P 1997-10-17 1997-10-17
US6895397P 1997-12-30 1997-12-30
US09/172,422 US6300485B1 (en) 1997-10-15 1998-10-14 Myosin IXa and cyclic nucleotide gated channel-15 (CNGC-15) polynucleotides, polypeptides, compositions, methods, and uses thereof
US09/851,682 US20020091248A1 (en) 1997-10-15 2001-05-08 Myosin IXa and cyclic nucleotide gated channel-15 (CNGC-15) polynucleotides, polypeptides, compositions, methods, and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/172,422 Division US6300485B1 (en) 1997-10-15 1998-10-14 Myosin IXa and cyclic nucleotide gated channel-15 (CNGC-15) polynucleotides, polypeptides, compositions, methods, and uses thereof

Publications (1)

Publication Number Publication Date
US20020091248A1 true US20020091248A1 (en) 2002-07-11

Family

ID=27370236

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/172,422 Expired - Fee Related US6300485B1 (en) 1997-10-15 1998-10-14 Myosin IXa and cyclic nucleotide gated channel-15 (CNGC-15) polynucleotides, polypeptides, compositions, methods, and uses thereof
US09/851,682 Abandoned US20020091248A1 (en) 1997-10-15 2001-05-08 Myosin IXa and cyclic nucleotide gated channel-15 (CNGC-15) polynucleotides, polypeptides, compositions, methods, and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/172,422 Expired - Fee Related US6300485B1 (en) 1997-10-15 1998-10-14 Myosin IXa and cyclic nucleotide gated channel-15 (CNGC-15) polynucleotides, polypeptides, compositions, methods, and uses thereof

Country Status (3)

Country Link
US (2) US6300485B1 (en)
AU (1) AU1098699A (en)
WO (1) WO1999019489A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014164472A1 (en) * 2013-03-10 2014-10-09 The Board Of Regents Of The University Of Texas System Biomarkers for chagas disease related cardiomyopathy

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU5886700A (en) * 1999-06-30 2001-01-31 Chiron Corporation Human chromosome 15 and 16 bardet-biedl syndrome polynucleotides and polypeptides and methods of use
US7094882B2 (en) 2000-04-21 2006-08-22 Yeda Research And Development Co. Ltd. Growth factor which acts through erb b-4 rtk
AU2002365195A1 (en) * 2001-07-12 2003-07-30 Incyte Genomics, Inc. Intracellular signaling molecules

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014164472A1 (en) * 2013-03-10 2014-10-09 The Board Of Regents Of The University Of Texas System Biomarkers for chagas disease related cardiomyopathy
US9696323B2 (en) 2013-03-10 2017-07-04 The Board Of Regents Of The University Of Texas System Biomarkers for chagas disease related cardiomyopathy

Also Published As

Publication number Publication date
US6300485B1 (en) 2001-10-09
WO1999019489A1 (en) 1999-04-22
AU1098699A (en) 1999-05-03

Similar Documents

Publication Publication Date Title
US7393942B2 (en) Nucleic acids encoding mutant forms of Fas ligand
DE69833572T2 (en) Regulated metastatic breast and colon cancer genes
DE69938190T2 (en) GENES WITH CHANGED EXPRESSION IN METASTIC BREAST OR THICK-DARM CANCER CELLS
RU2284332C2 (en) Neisseria antigenic peptides
JP2009165468A (en) Staphylococcus aureus protein and nucleic acid encoding the same
JP2003504062A (en) Antigenic meningococcal peptide
CA2327259A1 (en) Human transcriptional regulator molecules
WO1999034000A1 (en) IKKβ REGULATES TRANSCRIPTION FACTORS
US20020034800A1 (en) Bone marrow secreted proteins and polynucleotides
US6300485B1 (en) Myosin IXa and cyclic nucleotide gated channel-15 (CNGC-15) polynucleotides, polypeptides, compositions, methods, and uses thereof
WO1999033999A1 (en) Mitogen-activated protein kinase kinase 7 (mkk7)
EP1961813B1 (en) Human cyclin-dependent kinase (hPNQALRE)
EP0865483A2 (en) SIGNALING INOSITOL POLYPHOSPHATE 5-PHOSPHATASES (SIPs)
JP2002508658A (en) Detection of decrease in wild-type huBUB1 gene
US5948640A (en) Mammalian additional sex combs (mammalian Asx) acts as a tumor suppressor
US5914266A (en) Mammallian sex comb on midleg (mammalian SCM) acts as a tumor suppressor
WO1999033990A1 (en) HUMAN CXC CHEMOKINE (Tim-1)
EP0960120A1 (en) MAMMALIAN SEX COMB ON MIDLEG (MAMMALIAN Scm) ACTS AS AN ONCOGENE
CA2340277A1 (en) Human rna-associated proteins
WO1999000499A1 (en) Human fadd-interacting protein (fip)
US6174679B1 (en) CIF150/hTAFII150 is necessary for cell cycle progression
WO1999033986A1 (en) MAMMALIAN ENHANCER OF POLYCOMB (epc) ACTS AS A TUMOR SUPPRESSOR
WO1999029850A1 (en) HUMAN KISMET PROTEIN (hkis) ACTS AS AN ONCOGENE
WO1999033985A2 (en) C1F150/hTAFII150 IS NECESSARY FOR CELL CYCLE PROGRESSION
WO1998057985A2 (en) Human rip-interacting factor (rif)

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION