CA2271235A1 - Glaucoma-associated protein and corresponding nucleic acid and their therapeutic and diagnostic uses - Google Patents

Glaucoma-associated protein and corresponding nucleic acid and their therapeutic and diagnostic uses Download PDF

Info

Publication number
CA2271235A1
CA2271235A1 CA002271235A CA2271235A CA2271235A1 CA 2271235 A1 CA2271235 A1 CA 2271235A1 CA 002271235 A CA002271235 A CA 002271235A CA 2271235 A CA2271235 A CA 2271235A CA 2271235 A1 CA2271235 A1 CA 2271235A1
Authority
CA
Canada
Prior art keywords
glc1a
gene
glc
nucleic acid
protein
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
CA002271235A
Other languages
French (fr)
Inventor
Edwin M. Stone
Val Sheffield
Wallace L. M. Alward
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.)
University of Iowa Research Foundation UIRF
Original Assignee
University Of Iowa Research Foundation
Edwin M. Stone
Val Sheffield
Wallace L. M. Alward
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/748,479 external-priority patent/US5925748A/en
Priority claimed from US08/791,347 external-priority patent/US5885776A/en
Priority claimed from US08/822,999 external-priority patent/US6271026B1/en
Application filed by University Of Iowa Research Foundation, Edwin M. Stone, Val Sheffield, Wallace L. M. Alward filed Critical University Of Iowa Research Foundation
Publication of CA2271235A1 publication Critical patent/CA2271235A1/en
Abandoned legal-status Critical Current

Links

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
    • C07K14/46Peptides 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Ophthalmology & Optometry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Diagnostics for glaucoma, which are based on detection of non-wildtype GLC1A
genes or proteins in a subject, glaucoma therapeutics and assays for identifying agents that modulate the activity of GLC1A and that are therefore useful in treating or preventing glaucoma are described.

Description

WO 98/20131 PCT/US97/20702 _ ' GLAUCOMA-ASSOCIATED PROTEIN AND CORRESPONDING NUCLEIC ACID AND THEIR
THERAPEUTIC AND
DIAGNOSTIC USES
1. Background of the Invention In the United States, glaucoma is the second leading cause of legal blindness overall and the leading cause of blindness in African-American individuals (Hitler, R and H.A. Kahn, ( 1975) Am. J. Ophthalmol 80: 62 and Kahn, H.A. and H.B.
Moorhead (1973) US Public Health Service Publication NIH73-427, 120). Primary open glaucoma (POAG) is the most common form of glaucoma affecting 1-2% of the population over age forty (J.M. Tielsch et al., ( 1990) Arch Ophthalmol. 108:
286).
I S Nearly 12,000 people in the United States are blinded annually by this disorder (H.B.
Moorhead ( 1973) US Public Health Service Publication NIH73-427, 1202-4; J.M.
Tielsch et al., (1990) Arch Ophthalmol. 108: 286 and J.M. Tielsch, in Transactions of the New Orleans Academy of Ophthalmology, Ball, S.F. Franklin R.M., Eds (Kugler, Amsterdam, 1993), pp61-68).
One method of identifying genes involved in multifactorial disorders is to study Mendelian diseases with a similar phenotype. Juvenile open angle glaucoma (JOAG) is a term used to refer to a subset of POAG which has an earlier age of onset and a highly penetrant autosomal dominant mode of inheritance (A.T. Johnson et al., (1993) Ophthalmology 100:524). On clinical examination, patients with juvenile onset open angle glaucoma are identical to patients with later onset disease in that both exhibit elevated intraocular pressure and optic nerve cupping in the presence of a biomicroscopically normal trabecular meshwork. Isolation of genes involved in JOAG
. are needed to develop therapeutics and diagnostics for glaucoma.

WO 98/20131 PCT/US97/20702 _
-2-2. Summary of the Invention In one aspect, the invention features isolated GLC 1 A nucleic acid molecules. The disclosed molecules can be non-coding, (e.g. probe, antisense or ribozyme molecules) or can encode a functional polypeptide (e.g. a polypeptide which specifically modulates, e.g., by acting as either an agonist or antagonist, at least one bioactivity of the human GLC I A polypeptide).
In further embodiments, the nucleic acid molecule is a GLC 1 A nucleic acid that is at least 70%, preferably 80%, more preferably 85%, and even more preferably at least 95% homologous in sequence to the nucleic acids shown as SEQ ID
Nos: 1 or 2 or to the complement of the nucleic acids shown as SEQ ID Nos: 1 or 2. In another embodiment, the nucleic acid molecule encodes a polypeptide that is at least 92% and more preferably at least 95% similar in sequence to the polypeptide shown in ~ s SEQ ID No: 3.
The invention also provides probes and primers comprising substantially purified oligonucleotides, which correspond to a region of nucleotide sequence which hybridizes to at least 6 consecutive nucleotides of the sequences set forth as SEQ ID
20 Nos: I or 2 or complements of the sequences set forth as SEQ ID Nos: I or 2, or naturally occurring mutants thereof. In preferred embodiments, the probe/primer further includes a label group attached thereto, which is capable of being detected.
For expression, the subj ect GLC 1 A nucleic acids can include a 25 transcriptional regulatory sequence, e.g. at least one of a transcriptional promoter (e.g., for constitutive expression or inducible expression) or transcriptional enhancer or suppressor sequence, which regulatory sequence is operably linked to the GLC 1 A gene sequence. Such regulatory sequences in conjunction with a GLC I A nucleic acid molecule can be useful vectors for gene expression. This invention also describes host 3o cells transfected with said expression vector whether prokaryotic or eukaryotic and in vitro (e.g. cell culture) and in vivo (e.g. transgenic) methods for producing GLC I A
proteins by employing said expression vectors.
-3-In another aspect, the invention features isolated GLC 1 A polypeptides, preferably substantially pure preparations e.g. of plasma purified or recombinantly produced GLC 1 A polypeptides. In one embodiment, the polypeptide is identical to or similar to a GLC 1 A protein represented in SEQ ID No: 3. Related members of the vertebrate and particularly the mammalian GLC 1 A family are also within the scope of the invention. Preferably, a GLC 1 A polypeptide has an amino acid sequence at least about 92% homologous and preferably at least about 95%, 96%, 97%, 98% or 99%
homologous to the polypeptide represented in SEQ ID No: 3. In a preferred embodiment, the GLC 1 A polypeptide that is encoded by a nucleic acid which hybridizes with a nucleic acid sequence represented in one of SEQ ID Nos: 1 or 2. The subject GLC 1 A proteins also include modified protein, which are resistant to post-translational modification, as for example, due to mutations which alter modification sites (such as tyrosine, threonine, serine or aspargine residues), or which prevent glycosylation of the protein, or which prevent interaction of the protein with intracellular proteins involved in signal transduction.
The GLC 1 A polypeptide can comprise a full length protein, such as represented in SEQ ID No: 3, or it can comprise a fragment corresponding to one or more particular motifs/domains, or to arbitrary sizes, e.g., at least 5, 10, 25, S0, 100, 150, 175, 200, 225, 250,275, 300, 325, 350, 375, 400, 425, 450, 475, 480, 485, 490, 495, or 500 amino acids in length.
Another aspect of the invention features chimeric molecules (e.g. fusion proteins) comprised of a GLC 1 A protein. For instance, the GLC 1 A protein can be provided as a recombinant fusion protein which includes a second polypeptide portion, e.g., a second polypeptide having an amino acid sequence unrelated (heterologous) to the GLC 1 A polypeptide (e. g. the second polypeptide portion is glutathione-S-transferase, an enzymatic activity such as alkaline phosphatase or an epitope tag).
3o Yet another aspect of the present invention concerns an immunogen comprising a GLC 1 A polypeptide in an immunogenic preparation, the immunogen being capable of eliciting an immune response specific for a GLC 1 A
polypeptide; e.g. a humoral response, an antibody response and/or cellular response. In preferred WO 98/20131 PCT/US97/20702 _ '
-4-embodiments, the immunogen comprises an antigenic determinant, e.g. a unique determinant, from the protein represented in SEQ ID No: 3.
A still further aspect of the present invention features antibodies and antibody preparations specifically reactive with an epitope of the GLC 1 A
protein. In preferred embodiments the antibody specifically binds to at least one epitope represented in SEQ ID No: 3 The invention also features transgenic non-human animals which include (and preferably express) a heterologous form of a GLC 1 A gene described herein, or which misexpress an endogenous GLC 1 A gene (e.g., an animal in which expression of one or more of the subject GLC 1 A proteins is disrupted). Such a transgenic animal can serve as an animal model for studying cellular and tissue disorders comprising mutated or mis-expressed GLC 1 A alleles or for use in drug screening. Alternatively, such a transgenic animal can be useful for expressing recombinant GLC 1 A
polypeptides.
In yet another aspect, the invention provides assays, e.g., for screening test compounds to identify inhibitors, or alternatively, potentiators, of an interaction between a GLC 1 A protein and, for example, a virus, an extracellular ligand of the GLC 1 A protein, or an intracellular protein which binds to the GLC 1 A
protein. An exemplary method includes the steps of (i) combining a GLC 1 A polypeptide or bioactive fragments thereof, a GL,C 1 A target molecule (such as a GLC I A
ligand or a GLC 1 A substrate), and a test compound, e.g., under conditions wherein, but for the test compound, the GLC 1 A protein and target molecule are able to interact; and (ii) detecting the formation of a complex which includes the GLC 1 A protein and the target polypeptide either by directly quantitating the complex, by measuring inductive effects of the GLC 1 A protein, or, in the instance of a substrate, measuring the conversion to product. A statistically significant change, such as a decrease, in the interaction of the GLC 1 A and target molecule in the presence of a test compound (relative to what is detected in the absence of the test compound) is indicative of a modulation (e.g., inhibition or potentiation of the interaction between the GLC I A protein and the target molecule).
Yet another aspect of the present invention concerns a method for modulating GLC 1 A bioactivity, (e.g., by potentiating or disrupting a GLC 1 A

WO 98/20131 PCT/LTS97/20702 _
-5-bioactivity). In general, whether carried out in vivo, in vitro, or in situ, the method comprises treating the cell with an effective amount of a GLC 1 A therapeutic so as to alter, relative to the cell in the absence of treatment, a GLC 1 A
bioactivity. Accordingly, the method can be carried out with GLC 1 A therapeutics such as peptide and peptidomimetics or other molecules identified in the above-referenced drug screens which agonize or antagonize the effects of GLC 1 A (e.g., signalling from a protein or ligand binding of a GLC 1 A protein). Other GLC 1 A therapeutics include antisense constructs for inhibiting expression of GLC 1 A proteins, and dominant negative mutants of GLC 1 A proteins which competitively inhibit ligand interactions 1 o upstream and signal transduction downstream of the wild-type GLC 1 A
protein.
A further aspect of the present invention provides a method of determining if a subject is at risk for glaucoma or other disorder resulting from a mutant GLC 1 A gene. The method includes detecting, in a tissue of the subject, the presence or 15 absence of a genetic lesion characterized by at least one of (i) a mutation of a gene encoding a GLC 1 A protein, e.g., a gene represented in one of SEQ ID Nos: 1 or 2 or a homolog thereof; or (ii) the mis-expression of a GLC 1 A gene. In preferred embodiments, detecting the genetic lesion includes ascertaining the existence of at least one of: a deletion of one or more nucleotides from a GLC 1 A gene; an addition of one or 2o more nucleotides to the gene, a substitution of one or more nucleotides of the gene, a gross chromosomal rearrangement of the gene; an alteration in the level of a messenger RNA transcript of the gene (e.g., due to a promoter mutation); the presence of a non-wild type splicing pattern of a messenger RNA transcript of the gene; a non-wild type level of the protein; and/or an aberrant level of soluble GLC 1 A protein.
For example, detecting the genetic lesion can include (i) providing a probe/primer comprised of an oligonucleotide which hybridizes to a sense or antisense sequence of a GLC 1 A gene or naturally occurring mutants thereof, or 5' or 3' flanking sequences naturally associated with the GLC 1 A gene; (ii) contacting the probe/primer to 3o an appropriate nucleic acid containing sample; and {iii) detecting, by hybridization of the probe/primer to the nucleic acid, the presence or absence of the genetic lesion; e.g.
wherein detecting the lesion comprises utilizing the probe/primer to determine the nucleotide sequence of the GLC 1 A gene and, optionally, of the flanking nucleic acid WO 98/20131 PCT/US97/20702 _ _6_ , sequences. For instance, the primer can be employed in a polymerase chain reaction (PCR) or in a ligation chain reaction (LCR). In alternate embodiments, the level of a GLC 1 A protein is detected in an immunoassay using an antibody which is specifically immunoreactive with the GLC 1 A protein.
Other features and advantages of the invention will be apparent from the following detailed description and claims.
to 3. Brief Description of the Figures Figure 1 is a DNA sequence of the human GLC 1 A gene. The coding sequence, in conjunction with the 5' and 3' untranslated regions (UTRs) (SEQ.
ID. No. 1 ) is provided. Intron 1 is provided as SEQ ID No. 9 and intron 2 is provided as SEQ ID
I S No. 10, respectively. DNA encoding protein and comprising the three exon sequences is provided as SEQ ID No. 2. The encoded protein is presented as SEQ. ID. No. 3.
Figure 2 shows the Yeast Artificial Chromosomes (YACS) comprising the minimum tiling path contigs. Solid circles indicate the position of markers (STSs), 2o which were shown to be contained within each YAC. Disease intervals based on recombination within families and shared haplotypes between families are indicated with brackets.
Figure 3A and B shows two polymerase chain reaction (PCR) amplimers 25 of the GLC 1 A gene found to harbor sequence changes in patients with glaucoma. The wildtype amplimer shown in Figure 3A is provided as SEQ ID No. 11 and the mutation containing amplimer (CAC-TYR 430 His) is provided as SEQ ID No. 12. The wildtype amplimer shown in Figure 3B is provided as SEQ ID No. 13 and the mutation containing amplimer (GTC-GLY357 VAL and TAG-GLN361 STOP) is provided as SEQ ID No.
30 14.
Figure 4 schematically depicts the segregation of the GLY364VAL
mutation in a four generation family affected with open angle glaucoma. Black symbols in the pedigree drawing indicate individuals with documented evidence of open angle WO 98/20131 PCT/US97/20702 _ ' _7_ glaucoma. White symbols indicate spouses that are clinically unaffected. A
photograph of a silver-stained SSCP gel is shown below the pedigree drawing and is aligned so that each gel lane lies directly below the pedigree symbol of the family member whose DNA
was analyzed in that lane. For simplicity, clinically unaffected family members were not included.
Figure 5 is a representative chromatogram generated by fluorescent dye-primer sequencing of cloned PCR amplification products from an affected individual reveals a C to T transition which would be expected to result in a premature stop at to codon 368 (GLN368STOP). Sequences in the forward and reverse directions for bath the mutant and normal genes are shown: (A) normal, forward (SEQ ID No 15); (B) mutant, forward (SEQ ID No 16); (C) normal, reverse (SEQ ID No 17); and (D) mutant, reverse (SEQ ID No 18).
t 5 4. Detailed Description of the Invention 4.1. General As reported herein, a genetic locus associated with JOAG was identified 20 on chromosome 1q21-q31 by genetic linkage analysis. Observed recombinations between the glaucoma phenotype and highly polymorphic genetic markers in two large JOAG kindreds allowed the interval containing GLC 1 A gene to be narrowed to a 3 cM
region of chromosome 1 q between markers D 153665 and D 153664. Further evaluation of marker haplotypes revealed that each of three pairs of glaucoma families shared 25 alleles of the same eight contiguous markers suggesting that the GLC 1 A
gene lies within a narrower interval defined by D 1 S 1619 and D I 53664.
Several genes mapping to the GLC 1 A region of chromosome I were considered as candidates for the disease-causing gene. Three genes (LAMC 1 (H.C.
3o Watkins et. al., ( 1993) Hum. Mol. Genet. 2: 1084), NPRI (D.G. Lowe et al., ( 1990) Genomics 8:304), and CNR2 (S. Munro et al., ( 1993) Nature 365:61 ), were excluded from the candidate region by genetic linkage analysis using intragenic polymorphic markers. Five additional candidate genes were determined to lie within the observed WO 98/20131 PCT/US97/20702 _ -g_ recombinant interval by YAC STS content mapping: selectin E (M.P. Bevilacqua et al., (1989) Science 2;13: 1160) (GenBank accession no. M24736); selectin L (T.F.
Tedder et al., ( 1989) J. Exp. Med 170:123) (GenBank accession no. M25280); TXGR 1 (S.
Miura et al., ( 1991 ) Mol. Cell Biol 11:1313) (GenBank accession no. MD90224; APT1 LG 1 (T.
Takahashi et al., (1994) Int. Immunol. 6, 156; and TIGR (Trabecular meshwork Induced Glucocorticoid Response Protein) (J.R. Polansky et al., ( 1989) Prog.
Clin. Biol.
Res 12: 113; J. Escribano et al., ( 1995) J. Biochem. 118: 921; International Patent Application Publication No. WO 96/14411 ) (GenBank accession nos. 895491, 895447, 895443, 847209). However, two of these genes (selectin E, and selectin L) were found to lie outside of the shared haplotype interval with this approach. The remaining genes (APT 1 LG 1, TXGP-1, and TIGR) were found to map within the narrowest JOAG
interval by both YAC STS content and radiation hybrid mapping.
Two of these genes (APT1 LGl and TIGR) were screened for t 5 mutations in families with JOAG. Primers were selected from the available sequence (T.
Takahashi et al., ( 1994) Int. Immunol. 6, 1567, J. Escribano et al., ( 1995) J. Biochem.
118:921; International Patent Application Publication No. WO 96/14411) (GenBank accession nos. 895491, 895447, 895443, 847209) and overlapping PCR
amplification products were evaluated by single strand conformation polymorphism analysis (B.J.
2o Balsam et al., ( 1991 ) Anal. Biochem. 196: 80) and direct DNA sequencing.
Although the complete cDNA sequence of the APT1 LG 1 and TIGR genes have been published, the presence of intervening sequences permitted only 85 - 90% of their coding sequences to be screened in genomic DNA. Eight unrelated JOAG patients were screened with the APT1LG1 assay but no sequence variants were identified.
The TIGR gene assay was initially used to screen affected members of four different 1 q-linked glaucoma families, and affected members of four smaller families implicated by haplotypic data. Amino-acid-altering mutations were detected in four of eight families. A tyrosine to histidine mutation in codon 437 (Figs.
3A and 4) 3o was detected in all 22 affected members of the original family (V.C.
Sheffield et al., ( 1993) Nature Genet. 4: 47 ) linked to 1 q. A glycine to valine mutation in codon 364 (Fig 3B) was detected in two families including one previously unreported adult-onset open angle glaucoma family with 15 affected members (Fig. 4). A nonsense mutation WO 98/20131 PCT/US97/20702 _ ' -9- . -(glutamine to stop) at codon 368 (Figs. 3B and 5) was detected in two families. The latter mutation would be expected to result in a truncation of the gene product.
The prevalence of mutations in the two PCR amplimers that harbored these three changes was then estimated by screening four different populations:
glaucoma patients with a family history of the disease; unselected primary open angle glaucoma probands seen in a single clinic; the general population (approximated by patients with heritable retinal disease and spouses from families who participated in prior linkage studies); and, unrelated volunteers over the age of 40 with normal intraocular pressures and no personal or family history of glaucoma. PCR
products determined to contain a sequence variation by SSCP were sequenced and compared to sequence generated from an unaffected individual as well as the normal chromosome in each affected individual. Overall, missense or nonsense mutations were found in about 3-5% of unrelated glaucoma patients and in about 0.2% of controls. A Chi-square test ~ 5 revealed this difference to be significant (p<0.001 ). Table 1 sets forth GLC 1 A mutations and polymorphisms identified to date.
Table 1 2o Mutations: 1. GLN368STOP
2. GLY364VAL
3. TYR437HIS
4. ILE477ASN
5. 6 by insertion in codon 396 25 6. VAL495ILE
7. ALA445VAL
8. THR377MET
9. GLN19HIS
10. ARG82CYS
Polymorphisms 1. TYR347TYR
2. THR325THR

WO 98/20131 PCT/US97/20702 _ ' -10- .
3. VAL439VAL
4. LYS398ARG
5. PR013PR0 In summary, genetic linkage analysis and examination of shared haplotypes was used to narrow the 1 q glaucoma disease interval. A gene that lies within this interval and which is known to be expressed in the ciliary body (J.
Escribano et al., ( 1995) J. Biochem 118: 921 ) and trabecular meshwork (J.R. Polansky et al., ( I 989) Prog.
Clin. Biol. Res 12: 113 and International Patent Application Publication No.
WO
l0 96/14411 ) was examined. 13 unrelated patients with glaucoma (including the proband of the family (V.C. Sheffield et al., (1993) Nature Genet. =1:47) whose glaucoma phenotype was originally linked to I q) who each harbored one of three different amino-acid-altering mutations were identified. Collectively, this is compelling evidence that mutations in this gene are responsible for the glaucoma previously linked to 15 chromosome 1 q. In addition, the discovery of a mutation in 15 affected members of an adult-onset family as well as the identification of mutations in 2.9% of a consecutively ascertained group of unselected open angle glaucoma patients, suggests that this gene plays a role in a portion of all open angle glaucoma. It is possible that more than 3% of open angle glaucoma will eventually be shown to be associated with mutations in this 2o gene for two reasons: 1 ) only a portion of the gene was evaluated in this study; and, 2) the screening methods used are not 100% sensitive.
The gene, mutations in which have been linked to the glaucoma phenotype, was sequenced using standard techniques. The DNA sequence, including 5' 25 and 3' untranslated regions and two intron sequences is shown in Figure I .
Importantly, this sequence differs substantially from the TIGR gene sequence reported in International Patent Application No. WO 96/1441 I (GenBank accession nos.
895491, 895447, 895443 and 8947209). In fact, as reported, the TIGR gene sequence does not encode a functional protein.
A summary of the differences between the GLC 1 A gene disclosed herein, and the TIGR gene are presented in Table 2.

WO 98/20131 PCT/LTS97/20702 _ ' Table 2 1. The "C" at by #331 of the GLC 1 A DNA coding sequence (Figure 1 ) is not present in the TIGR
sequence.
2. The 29 bps "AGGGGCTGCAGAGGGAGCTGGGCACCCTG"(SEQ
ID NO. 8) at by #344-372 of the GLC 1 A DNA
coding sequence are not included in the TIGR
sequence. Errors l and 2 cause the TIGR sequence to wrongly predict 4 amino acids and exclude 10 amino acids from the protein sequence.
3. The "C" at by #559 of the GLC 1 A DNA coding ~ 5 sequence is not present in the TIGR sequence.
4. A "T" is wrongly inserted between by #560 and #56i of the GLC1A DNA coding sequence in the TIGR sequence. Errors 3 and 4 cause the TIGR sequence to incorrectly predict a serine 2o amino acid at residue # 187 instead of a glutamine.
The 9 bps "CTCAGGAGT" present at bps 706-714 of the GLC1A
DNA coding sequence are wrongly duplicated and inserted between by 714 and 715 in the TIGR sequence. Consequently, 25 the TIGR DNA sequence incorrectly predicts that 3 amino acids are inserted into the GLC 1 A protein sequence.
6. A "T" is incorrectly inserted between by #841 and #842 of the GLC I A DNA coding sequence in the TIGR sequence.
7. The "G" at by #891 of the GLC 1 A DNA coding sequence is not present in the TIGR sequence. Errors 6 and 7 cause WO 98/20131 PCT/LTS97/20702 _ ' -12- . _ 17 amino acids predicted by the GLC 1 A DNA coding sequence to be out of frame in the TIGR sequence.
8. A "G" at by #979 of the GLC 1 A DNA coding sequence is replaced with a "C" in the TIGR
sequence.
9. A "C" at by #980 of the GLC I A DNA coding sequence is replaced with a "G" in the TIGR sequence. Errors 8. and 9. cause to the TIGR sequence to wrongly predict an arginine amino acid at residue #327 instead of an alanine. The above 9 errors in the TIGR GLC 1 A sequence cause 42 incorrect amino acid predictions.
The identification of this disease gene increases the understanding of the pathophysiology of glaucoma, which in turn facilitates the development of assays for identifying molecules that modulate (e.g. agonize or antagonize) the bioactivity of a functional or mutant TIGR gene or protein. A therapeutically effective amount of these molecules can be administered to a subject with glaucoma or at risk for developing 2o glaucoma to prevent or reduce the severity of the condition.
4.2 Definitions For convenience, the meaning of certain terms and phrases employed in the specification, examples, and appended claims are provided below.
The term "agonist", as used herein, is meant to refer to an agent (e.g., a GLC I A therapeutic) that directly or indirectly enhances, supplements or potentiates a GLC 1 A bioactivity.
3o The term "antagonist", as used herein, is meant to refer to an agent (e.g.
a GLC 1 A therapeutic) that directly or indirectly prevents, minimizes or suppresses a GLC 1 A
bioactivity.

WO 98/20131 PCT/LTS97/20702 _ _13_ "Cells," "host cells" or "recombinant host cells" are terms used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
A "chimeric protein" or "fusion protein" is a fusion of a first amino acid sequence encoding one of the subject polypeptides with a second amino acid sequence t o defining a domain (e.g. polypeptide portion) foreign to and not substantially homologous with any domain of one of the proteins. A chimeric protein may present a foreign domain which is found (albeit in a different protein) in an organism which also expresses the first protein, or it may be an "interspecies", "intergenic", etc. fusion of protein structures expressed by different kinds of organisms. In general, a fusion protein can be represented by the general formula X-GLC 1 A-Y, wherein GLC 1 A represents a portion of the protein which is derived from one of the GLC 1 A proteins, and X and Y are independently absent or represent amino acid sequences which are not related to one of the GLC I A
sequences in an organism, including naturally occurring mutants.
"Complementary" sequences as used herein refer to sequences which have sufficient complementarity to be able to hybridize, forming a stable duplex.
A "delivery complex" shall mean a targeting means (e.g. a molecule that results in higher affinity binding of a gene, protein, polypeptide or peptide to a target cell surface and/or increased cellular uptake by a target cell). Examples of targeting means include: sterols (e.g. cholesterol), lipids (e.g. a cationic lipid, virosome or liposome), viruses (e.g. adenovirus, adeno-associated virus, and retrovirus) or target cell specific binding agents (e.g. ligands recognized by target cell specific receptors). Preferred complexes are sufficiently stable in vivo to prevent significant uncoupling prior to internalization by the 3o target cell. However, the complex is cleavable under appropriate conditions within the cell so that the gene, protein, polypeptide or peptide is released in a functional form.

WO 98/20131 PCT/L1S97120702 _ ' As is well known, genes for a particular polypeptide may exist in single or multiple copies within the genome of an individual. Such duplicate genes may be identical or may have certain modifications, including nucleotide substitutions, additions or deletions, which all still code for polypeptides having substantially the same activity.
The term "DNA
sequence encoding a gg polypeptide" may thus refer to one or more genes within a particular individual. Moreover, certain differences in nucleotide sequences may exist between individual organisms, which are called alleles. Such allelic differences may or may not result in differences in amino acid sequence of the encoded polypeptide yet still encode a protein with the same biological activity.
t0 As used herein, the term "gene" or "recombinant gene" refers to a nucleic acid molecule comprising an open reading frame encoding one of the polypeptides of the present invention, including both exon and (optionally) intron sequences. A
"recombinant gene" refers to nucleic acid encoding a GLC 1 A polypeptide and comprising GLC

encoding exon sequences, though it may optionally include intron sequences which are either derived from a chromosomal GLC 1 A gene or from an unrelated chromosomal gene.
Exemplary recombinant genes encoding the subj ect GLC 1 A polypeptides are represented in the appended Sequence Listing. The term "intron" refers to a DNA sequence present in a given GLC 1 A gene which is not translated into protein and is generally found between exons.
"Homology" or "identity" or "similarity" refers to sequence similarity between two peptides or between two nucleic acid molecules. Homology can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base or amino acid, then the molecules are homologous at that position. A degree of homology between sequences is a function of the number of matching or homologous positions shared by the sequences. An "unrelated" or "non-homologous" sequence shares less than identity, though preferably less than 25 % identity, with one of the GLC 1 A
sequences of the 3o present invention.
The term "interact" as used herein is meant to include detectable interactions between molecules, such as can be detected using, for example, a yeast two hybrid assay.

WO 98/20131 PCT/US97/20702 _ ' The term interact is also meant to include "binding" interactions between molecules.
Interactions may, for example, be protein-protein or protein-nucleic acid in nature.
The term "isolated" as used herein with respect to nucleic acids, such as DNA or RNA, refers to molecules separated from other DNAs, or RNAs, respectively, that are present in the natural source of the macromolecule. For example, an isolated nucleic acid encoding one of the subject GLC 1 A polypeptides preferably includes no more than 10 kilobases (kb) of nucleic acid sequence which naturally immediately flanks the gene in genomic DNA, more preferably no more than Skb of such naturally occurring 1 o flanking sequences, and most preferably less than 1.Skb of such naturally occurring flanking sequence. The term isolated as used herein also refers to a nucleic acid or peptide that is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized. Moreover, an "isolated nucleic acid" is meant to include nucleic acid ~ 5 fragments which are not naturally occurring as fragments and would not be found in the natural state. The term "isolated" is also used herein to refer to polypeptides which are isolated from other cellular proteins and is meant to encompass both purified and recombinant polypeptides.
2o The term "modulation" as used herein refers to both upregulation, (i.e., activation or stimulation), for example by agonizing; and downregulation, (i.e. inhibition or suppression) for example by antagonizing or a GLC 1 A bioactivity.
The "non-human animals" of the invention include mammalians such as 25 rodents, non-human primates, sheep, dog, cow, chickens, amphibians, reptiles, etc. Preferred non-human animals are selected from the rodent family including rat and mouse, most preferably mouse, though transgenic amphibians, such as members of the Xenopus genus, and transgenic chickens can also provide important tools for understanding and identifying agents which can affect, for example, embryogenesis and tissue formation. The term 30 "chimeric animal" is used herein to refer to animals in which the recombinant gene is found, or in which the recombinant gene is expressed in some but not all cells of the animal. The term "tissue-specific chimeric animal" indicates that one of the recombinant GLC 1 A genes is present and/or expressed or disrupted in some tissues but not others.

WO 98/20131 PCT/US97/20703 _ ' As used herein, the term "nucleic acid" refers to polynucleotides such as deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA).
The term should also be understood to include, as equivalents, analogs of either RNA or DNA made from nucleotide analogs, and, as applicable to the embodiment being described, single (sense or antisense) and double-stranded polynucleotides.
As used herein, the term "promoter" means a DNA sequence that regulates expression of a selected DNA sequence operably linked to the promoter, and which effects expression of the selected DNA sequence in cells. The term encompasses "tissue specific"
promoters, i.e. promoters, which effect expression of the selected DNA
sequence only in specific cells (e.g. cells of a specific tissue). The term also covers so-called "leaky"
promoters, which regulate expression of a selected DNA primarily in one tissue, but cause expression in other tissues as well. The term also encompasses non-tissue specific promoters and promoters that constitutively express or that are inducible (i.e. expression levels can be controlled).
The terms "protein", "polypeptide" and "peptide" are used interchangeably herein when referring to a gene product.
The term "recombinant protein" refers to a polypeptide of the present invention which is produced by recombinant DNA techniques, wherein generally, DNA
encoding a GLC 1 A polypeptide is inserted into a suitable expression vector which is in turn used to transform a host cell to produce the heterologous protein. Moreover, the phrase 2s "derived from", with respect to a recombinant GLC 1 A gene, is meant to include within the meaning of "recombinant protein" those proteins having an amino acid sequence of a native GLC 1 A protein, or an amino acid sequence similar thereto which is generated by mutations including substitutions and deletions (including truncation) of a naturally occurring form of the protein.
"Small molecule" as used herein, is meant to refer to a composition, which has a molecular weight of less than about SkD and most preferably less than about 4kD.
Small molecules can be nucleic acids, peptides, polypeptides, peptidometics, carbohydrates, lipids or other organic carbon containing or inorganic molecules. Extensive libraries of WO 98/20131 PCT/US97/20702 _ ' _17_ . _ chemical or biological (e.g., fungal, bacterial or alga! extracts) mixtures are available for screening with the assays of the invention.
As used herein, the term "specifically hybridizes" or "specifically detects"
refers to the ability of a nucleic acid molecule of the invention to hybridize to at least approximately 6, 12, 20, 30, 50, 100, 150, 200, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350, 1400, 1450, I 460, 1470, 1480, I 490 consecutive nucleotides of a vertebrate, preferably GLC 1 A gene, such as a GLC 1 A sequence designated in one of SEQ ID Nos: 1 or 2, or a sequence complementary thereto, or naturally occurring mutants thereof, such that it shows at least I O times more hybridization, preferably at least 50 times more hybridization, and even more preferably at least 100 times more hybridization than it does to a cellular nucleic acid (e.g., mRNA or genomic DNA) encoding a protein other than a vertebrate GLC 1 A
protein as defined herein.
~5 "Transcriptional regulatory sequence" is a generic term used throughout the specification to refer to DNA sequences, such as initiation signals, enhancers, and promoters, which induce or control transcription of protein coding sequences with which they are operably linked. In preferred embodiments, transcription of one of the recombinant 2o GLC 1 A genes is under the control of a promoter sequence (or other transcriptional regulatory sequence) which controls the expression of the recombinant gene in a cell-type in which expression is intended. It will also be understood that the recombinant gene can be under the control of transcriptional regulatory sequences which are the same or which are different from those sequences which control transcription of the naturally-occurring 25 forms of GLC 1 A proteins.
As used herein, the term "transfection" means the introduction of a nucleic acid, e.g., an expression vector, into a recipient cell by nucleic acid-mediated gene transfer.
"Transformation", as used herein, refers to a process in which a cell's genotype is changed 3o as a result of the cellular uptake of exogenous DNA or RNA, and, for example, the transformed cell expresses a recombinant form of a mammalian GLC 1 A
polypeptide or, in the case of anti-sense expression from the transferred gene, the expression of a naturally-occurring form of the GLC 1 A protein is disrupted.

WO 98/20131 PCT/US97/20702 _ ' _ 18_ , As used herein, the term "transgene" means a nucleic acid sequence (encoding, e.g., one of the mammalian GLC I A polypeptides, or pending an antisense transcript thereto), which is partly or entirely heterologous, i.e., foreign, to the transgenic animal or cell into which it is introduced, or, is homologous to an endogenous gene of the transgenic animal or cell into which it is introduced, but which is designed to be inserted, or is inserted, into the animal's genome in such a way as to alter the genome of the cell into which it is inserted (e.g., it is inserted at a location which differs from that of the natural gene or its insertion results in a knockout}. A transgene can include one or more t o transcriptional regulatory sequences and any other nucleic acid, such as introns, that may be necessary for optimal expression of a selected nucleic acid.
A "transgenic animal" refers to any animal, preferably a non-human mammal, bird or an amphibian, in which one or more of the cells of the animal contain heterologous nucleic acid introduced by way of human intervention, such as by transgenic techniques well known in the art. The nucleic acid is introduced into the cell, directly or indirectly by introduction into a precursor of the cell, by way of deliberate genetic manipulation, such as by microinjection or by infection with a recombinant virus. The term genetic manipulation does not include classical cross-breeding, or in vitro fertilization, but 2o rather is directed to the introduction of a recombinant DNA molecule. This molecule may be integrated within a chromosome, or it may be extrachromosomally replicating DNA. In the typical transgenic animals described herein, the transgene causes cells to express a recombinant form of one of the GLC 1 A proteins, e.g, either agonistic or antagonistic forms.
However, transgenic animals in which the recombinant GLC 1 A gene is silent are also contemplated, as for example, the FLP or CRE recombinase dependent constructs described below. Moreover, "transgenic animal" also includes those recombinant animals in which gene disruption of one or more GLC I A genes is caused by human intervention, including both recombination and antisense techniques.
3o The term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of preferred vector is an episome, i.e., a nucleic acid capable of extra-chromosomal replication.
Preferred vectors are those capable of autonomous replication and/expression of nucleic acids to which they are linked. Vectors capable of directing the expression of genes to which they are WO 98/20131 PCT/US97/Z0702 _ - I 9- .
operatively linked are referred to herein as "expression vectors". In general, expression vectors of utility in recombinant DNA techniques are often in the form of "plasmids" which refer generally to circular double stranded DNA loops which, in their vector form are not bound to the chromosome. In the present specification, "plasmid" and "vector"
are used interchangeably as the plasmid is the most commonly used form of vector.
However, the invention is intended to include such other forms of expression vectors which serve equivalent functions and which become known in the art subsequently hereto.
4.3 Nucleic Acids of the Present Invention 1 o As described below, one aspect of the invention pertains to isolated nucleic acids comprising nucleotide sequences encoding GLC 1 A polypeptides, and/or equivalents of such nucleic acids. The term equivalent is understood to include nucleotide sequences encoding functionally equivalent GLC I A polypeptides or functionally equivalent peptides having an activity of a vertebrate GLC 1 A protein such as described herein.
Equivalent nucleotide sequences will include sequences that differ by one or more nucleotide substitutions, additions or deletions, such as allelic variants; and will, therefore, include sequences that differ from the nucleotide sequence of the GLC 1 A gene shown in SEQ ID
Nos: 1 or 2 due to the degeneracy of the genetic code.
2o Preferred nucleic acids are vertebrate GLC 1 A nucleic acids. Particularly preferred vertebrate GLC 1 A nucleic acids are mammalian. Regardless of species, particularly preferred GLC 1 A nucleic acids encode polypeptides that are at least 90%
similar to an amino acid sequence of human GLC I A. Preferred nucleic acids encode a GLC 1 A polypeptide comprising an amino acid sequence at least 90% homologous and more preferably 94% homologous with an amino acid sequence of a vertebrate GLC I A, e.g., such as a sequence shown in one of SEQ ID Nos: 1 or 2. Nucleic acids which encode polypeptides at least about 95%, and even more preferably at least about 98-99% similarity with an amino acid sequence represented in SEQ ID Nos.: I or 2 are also within the scope of the invention. In a particularly preferred embodiment, the nucleic acid of the present 3o invention encodes an amino acid GLC I A sequence shown in one of SEQ ID No:
2. In one embodiment, the nucleic acid is a cDNA encoding a peptide having at least one bioactivity of the subject GLC 1 A polypeptide. Preferably, the nucleic acid includes all or a portion of the nucleotide sequence corresponding to the coding region of SEQ ID Nos: I or 2.

WO 98/20131 PCT/US97/20702 _ Still other preferred nucleic acids of the present invention encode a GLC 1 A
polypeptide which includes a polypeptide sequence corresponding to all or a portion of amino acid residues of SEQ ID No: 2 e.g., at least 2, 5, 10, 25, 50, 100, 150 or 200 amino acid residues of that region. For example, preferred nucleic acid molecules for use as probes/primer or antisense molecules (i.e. noncoding nucleic acid molecules) can comprise at least about 6, 12, 20, 30, 50, 100, 125, 150 or 200 base pairs in length, whereas coding nucleic acid molecules can comprise about 200, 250, 300, 350, 400, 410, 420, 430, 435 or 440 base pairs.
Another aspect of the invention provides a nucleic acid which hybridizes to a nucleic acid represented by one of SEQ ID Nos: 1 or 2 Appropriate stringency conditions which promote DNA hybridization, for example, 6.0 x sodium chloride/sodium citrate (SSC) at about 45°C, followed by a wash of 2.0 x SSC at 50°C, are known to those skilled in the art or can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N. Y. ( I 989), 6.3.1-6.3.6. For example, the salt concentration in the wash step can be selected from a low stringency of about 2.0 x SSC at SO°C to a high stringency of about 0.2 x SSC at 50°C. In addition, the temperature in the wash step can be increased from low stringency conditions at room temperature, about 22°C, to high stringency conditions at 2o about 65°C. Both temperature and salt may be varied, or either the temperature or the salt concentration may be held constant while the other variable is changed. In a preferred embodiment, a GLC I A nucleic acid of the present invention will bind to one of SEQ ID
Nos 1 or 2 under moderately stringent conditions, for example at about 2.0 x SSC and about 40oC. In a particularly preferred embodiment, aGLC 1 A nucleic acid of the present invention will bind to one of SEQ ID Nos: l, 3 or 4 under high stringency conditions, but will not bind to the nucleic acids shown in SEQ ID Nos: 6, 8, 9 or 11.
Preferred nucleic acids have a sequence at least 75% homologous and more preferably 80% and even more preferably at least 85% homologous with an amino acid sequence of a mammalian GLC 1 A, e.g., such as a sequence shown in one of SEQ
ID Nos:
1 and 2 Nucleic acids at least 90%, more preferably 95%, and most preferably at least about 98-99% homologous with a nucleic sequence represented in one of SEQ ID Nos: 1 and 2 are of course also within the scope of the invention. In preferred embodiments, the nucleic acid is a mammalian GLC 1 A gene and in particularly preferred embodiments, includes all WO 98/20131 PCT/US97/20702 _ or a portion of the nucleotide sequence corresponding to the coding region of one of SEQ
ID Nos: 1 or 2 Nucleic acids having a sequence that differs from the nucleotide sequences shown in one of SEQ ID Nos: 1 or 2 due to degeneracy in the genetic code are also within the scope of the invention. Such nucleic acids encode functionally equivalent peptides (i.e., a peptide having a biological activity of a GLC 1 A polypeptide) but differ in sequence from the sequence shown in the sequence listing due to degeneracy in the genetic code. For example, a number of amino acids are designated by more than one triplet.
Codons that specify the same amino acid, or synonyms (for example, CAU and CAC each encode histidine) may result in "silent" mutations which do not affect the amino acid sequence of a GLC 1 A polypeptide. However, it is expected that DNA sequence polymorphisms that do lead to changes in the amino acid sequences of the subject GLC 1 A
polypeptides will exist among mammalians. One skilled in the art will appreciate that these variations in one or ~ s more nucleotides (e.g., up to about 3-5% of the nucleotides) of the nucleic acids encoding polypeptides having an activity of a mammalian GLC 1 A polypeptide may exist among individuals of a given species due to natural allelic variation.
As indicated by the examples set out below, GLC l A protein-encoding 2o nucleic acids can be obtained from mRNA present in any of a number of eukaryotic cells.
It should also be possible to obtain nucleic acids encoding mammalian GLC 1 A
polypeptides of the present invention from genomic DNA from both adults and embryos. For example, a gene encoding a GLC 1 A protein can be cloned from either a cDNA or a genomic library in accordance with protocols described herein, as well as those generally known to persons 25 skilled in the art. Examples of tissues and/or libraries suitable for isolation of the subject nucleic acids include ocular tissue, among others. A cDNA encoding a GLC 1 A
protein can be obtained by isolating total mRNA from a cell, e.g. a vertebrate cell, a mammalian cell, or a human cell, including embryonic cells. Double stranded cDNAs can then be prepared from the total mRNA, and subsequently inserted into a suitable plasmid or bacteriophage 30 vector using any one of a number of known techniques. The gene encoding a mammalian GLC 1 A protein can also be cloned using established polymerase chain reaction techniques in accordance with the nucleotide sequence information provided by the invention. The WO 98/20131 PCT/US97/20702 _ ' nucleic acid of the invention can be DNA or RNA. A preferred nucleic acid is a cDNA
represented by a sequence selected from the group consisting of SEQ ID Nos:l and 2.
4.3.1. Vectors.
s This invention also provides expression vectors containing a nucleic acid encoding a GLC 1 A polypeptide, operably linked to at least one transcriptional regulatory sequence. "Operably linked" is intended to mean that the nucleotide sequence is linked to a regulatory sequence in a manner which allows expression of the nucleotide sequence.
Regulatory sequences are art-recognized and are selected to direct expression of the subject 1 o mammalian GLC 1 A proteins. Accordingly, the term "transcriptional regulatory sequence"
includes promoters, enhancers and other expression control elements. Such regulatory sequences are described in Goeddel; Gene Expression Technology: Methods in Enrymology 185, Academic Press, San Diego, CA ( 1990). In one embodiment, the expression vector includes a recombinant gene encoding a peptide having an agonistic activity of a subject 15 GLC 1 A polypeptide, or alternatively, encoding a peptide which is an antagonistic form of the GLC 1 A protein. Such expression vectors can be used to transfect cells and thereby produce polypeptides, including fusion proteins, encoded by nucleic acids as described herein. Moreover, the gene constructs of the present invention can also be used as a part of a gene therapy protocol to deliver nucleic acids encoding either an agonistic or 2o antagonistic form of one of the subject GLC 1 A proteins. Thus, another aspect of the invention features expression vectors for in vivo or in vitro transfection and expression of a GLC 1 A polypeptide in particular cell types so as to reconstitute the function of, or alternatively, abrogate the function of GLC 1 A-induced signaling in a tissue.
This could be desirable, for example, when the naturally-occurring form of the protein is misexpressed;
25 or to deliver a form of the protein which alters differentiation of tissue.
Expression vectors may also be employed to inhibit neoplastic transformation.
In addition to viral transfer methods, such as those illustrated above, non-viral methods can also be employed to cause expression of a subject GLC 1 A
polypeptide 3o in the tissue of an animal. Most nonviral methods of gene transfer rely on normal mechanisms used by mammalian cells for the uptake and intracellular transport of macromolecules. In preferred embodiments, non-viral targeting means of the present invention rely on endocytic pathways for the uptake of the subject GLC lA
polypeptide gene WO 98/20131 PCT/US97/20702 _ ' by the targeted cell. Exemplary targeting means of this type include liposomal derived systems, poly-lysine conjugates, and artificial viral envelopes.
4.3.2. Probes and Primers Moreover, the nucleotide sequences determined from the cloning of GLC 1 A
genes from mammalian organisms will further allow for the generation of probes and primers designed for use in identifying and/or cloning GLC I A homologs in other cell types, e.g. from other tissues, as well as GLC I A homologs from other mammalian organisms. For instance, the present invention also provides a probe/primer comprising a substantially 1 o purified oligonucleotide, which oligonucleotide comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least approximately 12, preferably 25, more preferably 40, 50 or 75 consecutive nucleotides of sense or anti-sense sequence selected from the group consisting of SEQ ID No:l and 2, or naturally occurring mutants thereof.
For instance, primers based on the nucleic acid represented in SEQ ID Nos: l and 2 can be used in PCR reactions to clone GLC I A homologs. Preferred primers of the invention are set forth as SEQ ID Nos. 4 and 5; and SEQ ID Nos. 5 and 6.
Likewise, probes based on the subject GLC 1 A sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins. In 2o preferred embodiments, the probe further comprises a label group attached thereto and able to be detected, e.g. the label group is selected from amongst radioisotopes, fluorescent compounds, enzymes, and enzyme co-factors.
As discussed in more detail below, such probes can also be used as a part of a diagnostic test kit for identifying cells or tissue which misexpress a GLC 1 A protein, such as by measuring a level of a GLC 1 A -encoding nucleic acid in a sample of cells from a patient; e.g. detecting GLC I A mRNA levels or determining whether a genomic gene has been mutated or deleted. Briefly, nucleotide probes can be generated from the subject GLC1A genes which facilitate histological screening of intact tissue and tissue 3o samples for the presence (or absence) of GLC1A-encoding transcripts.
Similar to the diagnostic uses of anti-GLC I A antibodies, the use of probes directed to GLC
1 A messages, or to genomic GLC 1 A sequences, can be used for both predictive and therapeutic evaluation of allelic mutations which might be manifest in, for example, neoplastic or hyperplastic WO 98/20131 PCT/US97/20702 _ ' disorders (e.g. unwanted cell growth) or abnormal differentiation of tissue.
Used in conjunction with immunoassays as described herein, the oligonucleotide probes can help facilitate the determination of the molecular basis for a developmental disorder which may involve some abnormality associated with expression (or lack thereof) of a GLC
1 A protein.
For instance, variation in polypeptide synthesis can be differentiated from a mutation in a coding sequence.
4.3.3. Antisense. Ribozvme and Triplex techniques One aspect of the invention relates to the use of the isolated nucleic acid in "antisense" therapy. As used herein, "antisense" therapy refers to administration or in situ generation of oligonucleotide molecules or their derivatives which specifically hybridize (e.g. bind) under cellular conditions, with the cellular mRNA and/or genomic DNA
encoding one or more of the subject GLC 1 A proteins so as to inhibit expression of that protein, e.g. by inhibiting transcription and/or translation. The binding may be by ~ 5 conventional base pair complementarity, or, for example, in the case of binding to DNA
duplexes, through specific interactions in the major groove of the double helix. In general, "antisense" therapy refers to the range of techniques generally employed in the art, and includes any therapy which relies on specific binding to oligonucleotide sequences.
An antisense construct of the present invention can be delivered, for example, as an expression plasmid which, when transcribed in the cell, produces RNA
which is complementary to at least a unique portion of the cellular mRNA which encodes a GLC 1 A protein. Alternatively, the antisense construct is an oligonucleotide probe which is generated ex vivo and which, when introduced into the cell causes inhibition of expression by hybridizing with the mRNA and/or genomic sequences of a GLC 1 A gene. Such oligonucleotide probes are preferably modified oligonucleotides which are resistant to endogenous nucleases, e.g. exonucleases and/or endonucleases, and are therefore stable in vivo. Exemplary nucleic acid molecules for use as antisense oligonucleotides are phosphoramidate, phosphothioate and methylphosphonate analogs of DNA (see also U.S.
3o Patents 5,176,996; 5,264,564; and 5,256,775). Additionally, general approaches to constructing oligomers useful in antisense therapy have been reviewed, for example, by Van der Krol et al. ( 1988) Biotechnigues 6:95 8-976; and Stein et al. ( 1988) Cancer Res 48:2659-2668. With respect to antisense DNA, oligodeoxyribonucleotides derived from the WO 98/20131 PCT/US97/20702 _ ' -25- , translation initiation site, e.g_, between the -10 and +10 regions of the GLC
1 A nucleotide sequence of interest, are preferred.
Antisense approaches involve the design of oligonucleotides (either DNA
or RNA) that are complementary to GLC 1 A mRNA. The antisense oligonucleotides will bind to the GLC 1 A mRNA transcripts and prevent translation. Absolute complementarity, although preferred, is not required. a sequence "complementary" to a portion of an RNA, as referred to herein, means a sequence having sufficient complementarity to be able to hybridize with the RNA, forming a stable duplex; in the case of double-stranded antisense nucleic acids, a single strand of the duplex DNA may thus be tested, or triplex formation may be assayed. The ability to hybridize will depend on both the degree of complementarity and the length of the antisense nucleic acid. Generally, the longer the hybridizing nucleic acid, the more base mismatches with an RNA it may contain and still form a stable duplex (or triplex, as the case may be). One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures to determine the melting point of the hybridized complex.
Oligonucleotides that are complementary to the 5' end of the message, e.g., the 5' untranslated sequence up to and including the AUG initiation codon, should work 2o most efficiently at inhibiting translation. However, sequences complementary to the 3' untranslated sequences of mRNAs have recently been shown to be effective at inhibiting translation of mRNAs as well. (Wagner, R. 1994. Nature 372:333). Therefore, oligonucleotides complementary to either the 5' or 3' untranslated, non-coding regions of a GLC 1 A gene could be used in an antisense approach to inhibit translation of endogenous GLC I A mRNA. Oligonucleotides complementary to the 5' untranslated region of the mRNA should include the complement of the AUG start codon. Antisense oligonucleotides complementary to mRNA coding regions are less efficient inhibitors of translation but could be used in accordance with the invention. Whether designed to hybridize to the 5', 3' or coding region of GLC 1 A mRNA, antisense nucleic acids should be at least six nucleotides 3o in length, and are preferably oligonucleotides ranging from 6 to about 50 nucleotides in length. In certain embodiments, the oligonucleotide is at least 10 nucleotides, at least 17 nucleotides, at least 25 nucleotides, or at least 50 nucleotides.

WO 98/20131 PCT/US97/20702 _ ' Regardless of the choice of target sequence, it is preferred that in vitro studies are first performed to quantitate the ability of the antisense oligonucleotide to quantitate the ability of the antisense oligonucleotide to inhibit gene expression. It is preferred that these studies utilize controls that distinguish between antisense gene inhibition and nonspecific biological effects of oligonucleotides. It is also preferred that these studies compare levels of the target RNA or protein with that of an internal control RNA or protein. Additionally, it is envisioned that results obtained using the antisense oligonucleotide are compared with those obtained using a control oligonucleotide. It is preferred that the control oligonucleotide is of approximately the same length as the test 1 o oligonucleotide and that the nucleotide sequence of the oligonucleotide differs from the antisense sequence no more than is necessary to prevent specific hybridization to the target sequence.
The oligonucleotides can be DNA or RNA or chimeric mixtures or derivatives or modified versions thereof, single-stranded or double-stranded.
The oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone, for example, to improve stability of the molecule, hybridization, etc. The oligonucleotide may include other appended groups such as peptides (e.~,., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, 2o e.g., Letsinger et al., 1989, Proc. Natl. Acad Sci. U.S.A. 86:6553-6556;
Lemaitre et al., 1987, Proc. Natl. Acad. Sci. 84:648-652; PCT Publication No. WO 88/09810, published December 15, 1988) or the blood-brain barrier (see, e.g., PCT Publication No.
WO
89/10134, published April 25, 1988), hybridization-triggered cleavage agents.
(See, e.g., Krol et al., 1988, BioTechniques 6:958-976) or intercalating agents. (See, e.g., Zon, 1988, Pharm. Res. 5:539-549). To this end, the oligonucleotide may be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent, etc.
The antisense oligonucleotide may comprise at least one modified base 3o moiety which is selected from the group including but not limited to 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, WO 98/20131 PCT/US97/20702 _ ' 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uraciI-5- oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine.
1 o The antisense oligonucleotide may also comprise at least one modified sugar moiety selected from the group including but not limited to arabinose, 2-fluoroarabinose, xylulose, and hexose.
In yet another embodiment, the antisense oIigonucleotide comprises at ~ 5 least one modified phosphate backbone selected from the group consisting of a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, and a formacetal or analog thereof.
2o In yet another embodiment, the antisense oligonucleotide is an -anomeric oligonucleotide. An -anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual -units, the strands run parallel to each other (Gautier et al., 1987, Nucl. Acids Res. 15:6625-6641 ). The oligonucleotide is a 2'-0-methylribonucleotide (moue et al., 1987, Nucl. Acids Res. 15:6131-6148), or a 25 chimeric RNA-DNA analogue (moue et al., 1987, FEBS Lett. 215:327-330).
Oligonucleotides of the invention may be synthesized by standard methods known in the art, e.g. by use of an automated DNA synthesizer (such as are commercially available from Biosearch, Applied Biosystems, etc.). As examples, 3o phosphorothioate oligonucleotides may be synthesized by the method of Stein et al.
(1988, Nucl. Acids Res. 16:3209), methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin et al., 1988, Proc.
Natl. Acad.
Sci. U.S.A. 85:7448-745I), etc.

WO 98/20131 PCT/US97/20702 _ ' While antisense nucleotides complementary to the GLC 1 A coding region sequence could be used, those complementary to the transcribed untranslated region are most preferred.
The antisense molecules should be delivered to cells which express the GLC 1 A in vivo. A number of methods have been developed for delivering antisense DNA or RNA to cells; e,g_, antisense molecules can be injected directly into the tissue site, or modified antisense molecules, designed to target the desired cells (~, antisense to linked to peptides or antibodies that specifically bind receptors or antigens expressed on the target cell surface) can be administered systematically.
However, it is often difficult to achieve intracellular concentrations of the antisense sufficient to suppress translation of endogenous mRNAs. Therefore a ~ 5 preferred approach utilizes a recombinant DNA construct in which the antisense oligonucleotide is placed under the control of a strong pol III or pol II
promoter. The use of such a construct to transfect target cells in the patient will result in the transcription of sufficient amounts of single stranded RNAs that will form complementary base pairs with the endogenous GLC 1 A transcripts and thereby prevent 2o translation of the GLC 1 A mRNA. For example, a vector can be introduced in vivo such that it is taken up by a cell and directs the transcription of an antisense RNA. Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA. Such vectors can be constructed by recombinant DNA technology methods standard in the art. Vectors can be plasmid, 25 viral, or others known in the art, used for replication and expression in mammalian cells.
Expression of the sequence encoding the antisense RNA can be by any promoter known in the art to act in mammalian, preferably human cells. Such promoters can be inducible or constitutive. Such promoters include but are not limited to: the SV40 early promoter region (Bernoist and Chambon, 1981, Nature 290:304-310), the promoter contained in 3o the 3' long terminal repeat of Rous sarcoma virus (Yamamoto et al., 1980, Cell 22:787-797), the herpes thymidine kinase promoter ( Wagner et al., 1981, Proc. Natl.
Acad. Sci.
U.S.A. 78:1441-1445), the regulatory sequences of the metallothionein gene (Brinster et al, 1982, Nature 296:39-42), etc. Any type of plasmid, cosmid, YAC or viral vector can be used to prepare the recombinant DNA construct which can be introduced directly into WO 98/20131 PCT/US97/20702 _ ' the tissue site; e.~, the choroid plexus or hypothalamus. Alternatively, viral vectors can be used which selectively infect the desired tissue; (e.~., for brain, herpesvirus vectors may be used), in which case administration may be accomplished by another route (e.~., systematically).
Ribozyme molecules designed to catalytically cleave GLC1A mRNA
transcripts can also be used to prevent translation of GLC 1 A mRNA and expression of GLC1A. (See, ~, PCT International Publication WO 90/11364, published October 4, 1990; Sarver et al., 1990, Science 247:1222-1225). While ribozymes that cleave mRNA
at site specific recognition sequences can be used to destroy GLC 1 A mRNAs, the use of hammerhead ribozymes is preferred. Hammerhead ribozymes cleave mRNAs at locations dictated by flanking regions that form complementary base pairs with the target mRNA. The sole requirement is that the target mRNA have the following sequence of two bases: 5'-UG-3'. The construction and production of hammerhead ribozymes is well known in the art and is described more fully in Haseloff and Gerlach, 1988, Nature, 334:585-591. There are hundreds of potential hammerhead ribozyme cleavage sites within the nucleotide sequence of human GLC 1 A cDNA.
Preferably the ribozyme is engineered so that the cleavage recognition site is located near the 5' end of the GLC 1 A mRNA; i.e., to increase efficiency and minimize the intracellular 2o accumulation of non-functional mRNA transcripts.
The ribozymes of the present invention also include RNA
endoribonucleases (hereinafter "Cech-type ribozymes") such as the one which occurs naturally in Tetrahymena thermophila (known as the IVS, or L-19 IVS RNA) and which has been extensively described by Thomas Cech and collaborators (Zaug, et al., 1984, Science, 224:574-578; Zaug and Cech, 1986, Science, 231:470-475; Zaug, et al., 1986, Nature, 324:429-433; published International patent application No. WO
88/04300 by University Patents Inc.; Been and Cech, 1986, Cell, 47:207-216). The Cech-type ribozymes have an eight base pair active site which hybridizes to a target RNA
sequence 3o whereafter cleavage of the target RNA takes place. The invention encompasses those Cech-type ribozymes which target eight base-pair active site sequences that are present in GLC 1 A.

WO 98/20131 PCT/US97/20702 _ As in the antisense approach, the ribozymes can be composed of modified oligonucleotides (~ for improved stability, targeting, etc.) and should be delivered to cells which express the GLC 1 A in vivo e.~, hypothalamus and/or the choroid plexus. A
preferred method of delivery involves using a DNA construct "encoding" the ribozyme under the control of a strong constitutive pol III or pol II promoter, so that transfected cells will produce sufficient quantities of the ribozyme to destroy endogenous messages and inhibit translation. Because ribozymes unlike antisense molecules, are catalytic, a lower intracellular concentration is required for efficiency.
Endogenous GLC 1 A gene expression can also be reduced by inactivating or "knocking out" the GLC 1 A gene or its promoter using targeted homologous recombination. (E.~., see Smithies et al., 1985, Nature 317:230-234; Thomas &
Capecchi, 1987, Cell 5 I :503-S 12; Thompson et al., 1989 Cell 5:3 I3-321;
each of which is incorporated by reference herein in its entirety). For example, a mutant, non-functional GLC 1 A (or a completely unrelated DNA sequence) flanked by DNA
homologous to the endogenous GLC 1 A gene (either the coding regions or regulatory regions of the GLC 1 A gene) can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express GLC 1 A in vivo.
Insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the 2o GLC 1 A gene. Such approaches are particularly suited in the agricultural field where modifications to ES (embryonic stem) cells can be used to generate animal offspring with an inactive GLClA (~, see Thomas & Capecchi 1987 and Thompson 1989, s_upra). However this approach can be adapted for use in humans provided the recombinant DNA constructs are directly administered or targeted to the required site in vivo using appropriate viral vectors, e.~, herpes virus vectors for delivery to brain tissue; e.~.., the hypothalamus and/or choroid plexus.
Alternatively, endogenous GLC 1 A gene expression can be reduced by targeting deoxyribonucleotide sequences complementary to the regulatory region of the 3o GLC 1 A gene (i.e., the GLC 1 A promoter and/or enhancers) to form triple helical structures that prevent transcription of the GLC 1 A gene in target cells in the body. (See generally, Helene, C. 1991, Anticancer Drug Des., 6(6):569-84; Helene, C., et al., 1992, Ann, N.Y. Acad. Sci., 660:27-36; and Maher, L.J., 1992, Bioassays 14(12):807-15).

WO 98/20131 PCT/LTS97/20702 _ -31- .
Likewise, the antisense constructs of the present invention, by antagonizing the normal biological activity of one of the GLC 1 A proteins, can be used in the manipulation of tissue, e.g. tissue differentiation, both in vivo and for ex vivo tissue cultures.
Furthermore, the anti-sense techniques (e.g. microinjection of antisense molecules, or transfection with plasmids whose transcripts are anti-sense with regard to a GLC 1 A mRNA or gene sequence) can be used to investigate role of GLC 1 A in developmental events, as well as the normal cellular function of GLC 1 A in adult tissue.
Such techniques can be utilized in cell culture, but can also be used in the creation of transgenic animals, as detailed below.
Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. The mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by an endonucleolytic cleavage. The composition of ribozyme molecules must include one or more sequences complementary to the target gene mRNA, and must include the well known catalytic sequence responsible for mRNA cleavage. For this sequence, see U.S.
Pat. No.
5,093,246, which is incorporated by reference herein in its entirety. As such within the scope of the invention are engineered hammerhead motif ribozyme molecules that 2o specifically and efficiently catalyze endonucleolytic cleavage of RNA
sequences encoding GLC 1 A proteins.
Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the molecule of interest for ribozyme cleavage sites which include the following sequences, GUA, GUU and GUC. Once identified, short RNA
sequences of between 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site may be evaluated for predicted structural features, such as secondary structure, that may render the oligonucleotide sequence unsuitable. The suitability of candidate sequences may also be evaluated by testing their accessibility to hybridization 3o with complementary oligonucleotides, using ribonuclease protection assays.
Nucleic acid molecules to be used in triple helix formation for the inhibition of transcription are preferably single stranded and composed of deoxyribonucleotides. The -3~_ base composition of these oligonucleotides should promote triple helix formation via Hoogsteen base pairing roles, which generally require sizable stretches of either purines or pyrimidines to be present on one strand of a duplex. Nucleotide sequences may be pyrimidine-based, which will result in T'AT and CGC triplets across the three associated strands of the resulting triple helix. The pyrimidine-rich molecules »rnvir~P
hacP
complementarity to a purine-rich region of a single strand of the duplex in a parallel orientation to that strand. In addition, nucleic acid molecules may be chosen that are purine-rich, for example, containing a stretch of G residues. These molecules will form a triple helix with a DNA duplex that is rich in GC pairs, in which the majority of the purine residues are located on a single strand of the targeted duplex, resulting in CGC triplets across the three strands in the triplex.
Alternatively, the potential sequences that can be targeted for triple helix formation may be increased by creating a so called "switchback" nucleic acid molecule.
Switchback molecules are synthesized in an alternating 5'-3', 3'-5' manner, such that they base pair with first one strand of a duplex and then the other, eliminating the necessity for a sizable stretch of either purines or pyrimidines to be present on one strand of a duplex.
Antisense RNA and DNA, ribozyme, and triple helix molecules of the invention may be prepared by any method known in the art for the synthesis of DNA and RNA molecules. These include techniques for chemically synthesizing oligodeoxyribonucleotides and oligoribonucleotides well known in the art such as for example solid phase phosphoramidite chemical synthesis. Alternatively, RNA
molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors which incorporate suitable RNA polymerise promoters such as the T7 or SP6 polymerise promoters. Alternatively, antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into cell lines.
Moreover, various well-known modifications to nucleic acid molecules may be introduced as a means of increasing intracellular stability and half life.
Possible modifications include but are not limited to the addition of flanking sequences of ribonucleotides or deoxyribonucleotides to the 5' and/or 3' ends of the molecule or the use WO 98/20131 PCT/LTS97/20702 _ of phosphorothioate or 2' O-methyl rather than phosphodiesterase linkages within the oligodeoxyribonucleotide backbone.
4.4. Polypeptides of the Present Invention The present invention also makes available isolated GLC 1 A polypeptides which are isolated from, or otherwise substantially free of other cellular proteins, especially other signal transduction factors and/or transcription factors which may normally be associated with the GLC 1 A polypeptide. The term "substantially free of other cellular proteins" (also referred to herein as "contaminating proteins") or "substantially pure or 1 o purified preparations" are defined as encompassing preparations of GLC 1 A
polypeptides having less than about 20% (by dry weight) contaminating protein, and preferably having less than about 5% contaminating protein. Functional forms of the subject polypeptides can be prepared, for the first time, as purified preparations by using a cloned gene as described herein. By "purified", it is meant, when referring to a peptide or DNA or RNA
sequence, ~ 5 that the indicated molecule is present in the substantial absence of other biological macromolecules, such as other proteins. The term "purified" as used herein preferably means at least 80% by dry weight, more preferably in the range of 95-99% by weight, and most preferably at least 99.8% by weight, of biological macromolecules of the same type present (but water, buffers, and other small molecules, especially molecules having a 2o molecular weight of less than 5000, can be present). The term "pure" as used herein preferably has the same numerical limits as "purified" immediately above.
"Isolated" and "purified" do not encompass either natural materials in their native state or natural materials that have been separated into components (e.g., in an acrylamide gel) but not obtained either as pure (e.g. lacking contaminating proteins, or chromatography reagents such as denaturing 25 agents and polymers, e.g. acrylamide or agarose) substances or solutions.
In preferred embodiments, purified GLC 1 A preparations will lack any contaminating proteins from the same animal from which GLC1A is normally produced, as can be accomplished by recombinant expression of, for example, a human GLC 1 A protein in a non-human cell.
3o Full length proteins or fragments corresponding to one or more particular motifs and/or domains or to arbitrary sizes, for example, at least 5, 10, 25, 50, 75, 100, 125, 1 SO amino acids in length are within the scope of the present invention.

WO 98/20131 PCT1US97/20702 _ -34- .
For example, isolated GLC I A polypeptides can include all or a portion of an amino acid sequences corresponding to a GLC I A polypeptide represented in SEQ ID
No. 3. Isolated peptidyl portions of GLC I A proteins can be obtained by screening peptides recombinantly produced from the corresponding fragment of the nucleic acid encoding such peptides. In addition, fragments can be chemically synthesized using techniques known in the art such as conventional Merrifield solid phase f Moc or t-Boc chemistry.
For example, a GLC 1 A polypeptide of the present invention may be arbitrarily divided into fragments of desired length with no overlap of the fragments, or preferably divided into overlapping fragments of a desired length. The fragments can be produced (recombinantly or by 1 o chemical synthesis) and tested to identify those peptidyl fragments which can function as either agonists or antagonists of a wild-type (e.g., "authentic") GLC 1 A
protein.
Another aspect of the present invention concerns recombinant forms of the GLC I A proteins. Recombinant polypeptides preferred by the present invention, in addition to native GLC 1 A proteins, are at least 92% homologous and more preferably 94%
homologous and most preferably 95% homologous with an amino acid sequence represented by SEQ ID Nos: 3. Polypeptides which are at least about 98-99%
homologous with a sequence selected from the group consisting of SEQ ID No: 3 are also within the scope of the invention. In a preferred embodiment, a GLC 1 A protein of the present 2o invention is a GLC 1 A protein. In a particularly preferred embodiment a GLC 1 A protein comprises the coding sequence of one of SEQ ID No.: 3. In particularly preferred embodiments, a GLC I A protein has a GLC I A bioactivity.
The present invention further pertains to recombinant forms of one of the subj ect GLC 1 A polypeptides which are encoded by genes derived from a mammalian organism, and which have amino acid sequences evolutionarily related to the GLC I A
proteins represented in SEQ ID Nos: 3. Such recombinant GLC 1 A polypeptides preferably are capable of functioning in one of either role of an agonist or antagonist of at least one biological activity of a wild-type ("authentic") GLC I A protein of the appended sequence listing. The term "evolutionarily related to", with respect to amino acid sequences of GLC I A proteins, refers to both polypeptides having amino acid sequences which have arisen naturally, and also to mutational variants of GLC 1 A polypeptides which are derived, for example, by combinatorial mutagenesis. Such evolutionarily derived GLC I A
polypeptides preferred by the present invention have a GLC 1 A bioactivity and are at least WO 98/20131 PCT/US97/20702 _ ' 92% homologous and more preferably 94% homologous and most preferably 98-99%
homologous with the amino acid sequence set forth as SEQ ID Nos: 3. In a particularly preferred embodiment, a GLC 1 A protein comprises the amino acid coding sequence of SEQ ID No: 3.
In general, polypeptides referred to herein as having an activity (e.g., are "bioactive"} of a GLC I A protein are defined as polypeptides which include an amino acid sequence corresponding (e.g., identical or homologous) to all or a portion of the amino acid sequences of a GLC 1 A protein shown in SEQ ID No: 3 and which mimic or antagonize all or a portion of the biological/biochemical activities of a naturally occurring GLC 1 A protein.
In preferred embodiments, the biochemical activities are related to gene expression, pituitary development, and abdominal development related to umbilical and vitelline artery expression.
Other biological activities of the subj ect GLC 1 A proteins are described herein or will be reasonably apparent to those skilled in the art. According to the present invention, a polypeptide has biological activity if it is a specific agonist or antagonist of a naturally-occurring form of a GLC I A protein.
The present invention further pertains to methods of producing the subject GLC 1 A polypeptides. For example, a host cell transfected with a nucleic acid vector directing expression of a nucleotide sequence encoding the subject polypeptides can be cultured under appropriate conditions to allow expression of the peptide to occur. The cells may be harvested, lysed and the protein isolated. A cell culture includes host cells, media and other byproducts. Suitable media for cell culture are well known in the arr ThP
recombinant GLC I A polypeptide can be isolated from cell culture medium, host cells, or both using techniques known in the art for purifying proteins including ion-exchange chromatography, gel filtration chromatography, ultrafiltration, electrophoresis, and immunoaffinity purification with antibodies specific for such peptide. In a preferred 3o embodiment, the recombinant GLC 1 A polypeptide is a fusion protein containing a domain which facilitates its purification, such as GST fusion protein or poly(His) fusion protein.

WO 98/20131 PCT/US97/20702 _ ' Moreover, it will be generally appreciated that, under certain circumstances, it may be advantageous to provide homologs of one of the subject GLC 1 A
polypeptides which function in a limited capacity as one of either a GLC 1 A agonist (mimetic) or a GLC 1 A antagonist, in order to promote or inhibit only a subset of the biological activities of the naturally-occurring form of the protein. Thus, specific biological effects can be elicited by treatment with a homolog of limited function, and with fewer side effects relative to treatment with agonists or antagonists which are directed to all of the biological activities of naturally occurring forms of GLC 1 A proteins.
1 o Homologs of each of the subj ect GLC 1 A proteins can be generated by mutagenesis, such as by discrete point mutation(s), or by truncation. For instance, mutation can give rise to homologs which retain substantially the same, or merely a subset, of the biological activity of the GLC I A polypeptide from which it was derived.
Alternatively, antagonistic forms of the protein can be generated which are able to inhibit the function of the naturally occurring form of the protein, such as by competitively binding to a downstream or upstream member of the biochemical pathway, which includes the protein. In addition, agonistic forms of the protein may be generated which are constitutively active. Thus, the human GLC 1 A protein and homologs thereof provided by the subject invention may be either positive or negative regulators of gene expression.
The recombinant GLC 1 A polypeptides of the present invention also include homologs of the authentic GLC 1 A proteins, such as versions of those protein which are resistant to proteolytic cleavage, as for example, due to mutations which alter ubiquitination or other enzymatic targeting associated with the protein.
GLC 1 A polypeptides may also be chemically modified to create GLC 1 A
derivatives by forming covalent or aggregate conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like. Covalent derivatives of GLC1A proteins can be prepared by linking the chemical moieties to functional groups on 3o amino acid sidechains of the protein or at the N-terminus or at the C-terminus of the polypeptide.
Modification of the structure of the subject GLC 1 A polypeptides can be for such purposes as enhancing therapeutic or prophylactic efficacy, stability (e.g., ex vivo shelf WO 98/20131 PCTJUS97/20702 _ -3'7- , life and resistance to proteolytic degradation in vivo), or post-translational modifications (e.g., to alter phosphorylation pattern of protein). Such modified peptides, when designed to retain at least one activity of the naturally-occurring form of the protein, or to produce specific antagonists thereof, are considered functional equivalents of the GLC

polypeptides described in more detail herein. Such modified peptides can be produced, for instance, by amino acid substitution, deletion, or addition.
For example, it is reasonable to expect that an isolated replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, a threonine with a serine, or a similar replacement of an amino acid with a structurally related amino acid (i.e.
isosteric and/or isoelectric mutations) will not have a major effect on the biological activity of the resulting molecule. Conservative replacements are those that take place within a family of amino acids that are related in their side chains. Genetically encoded amino acids are can be divided into four families: ( 1 ) acidic = aspartate, glutamate;
(2) basic = lysine, arginine, histidine; (3) nonpolar = alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan; and (4) uncharged polar = gIycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine. In similar fashion, the amino acid repertoire can be grouped as ( 1 ) acidic = aspartate, glutamate; (2) basic = lysine, arginine histidine, (3) aliphatic = glycine, alanine, valine, leucine, isoleucine, serine, threonine, with serine and 2o threonine optionally be grouped separately as aliphatic-hydroxyl; (4) aromatic =
phenylalanine, tyrosine, tryptophan; (5) amide = asparagine, glutamine; and (6) sulfur -containing = cysteine and methionine. (see, for example, Biochemistry, 2nd ed., Ed. by L.
Stryer, WH Freeman and Co.: 1981 ). Whether a change in the amino acid sequence of a peptide results in a functional GLC 1 A homolog (e.g. functional in the sense that the resulting polypeptide mimics or antagonizes the wild-type form) can be readily determined by assessing the ability of the variant peptide to produce a response in cells in a fashion similar to the wild-type protein, or competitively inhibit such a response.
Polypeptides in which more than one replacement has taken place can readily be tested in the same manner.
3o This invention further contemplates a method for generating sets of combinatorial mutants of the subject GLC I A proteins as well as truncation mutants, and is especially useful for identifying potential variant sequences (e.g. homologs) that are functional in modulating gene expression. The purpose of screening such combinatorial WO 98/20131 PCT/LTS97/20702 _ libraries is to generate, for example, novel GLC 1 A homologs which can act as either agonists or antagonist, or alternatively, possess novel activities all together.
Likewise, GLC 1 A homologs can be generated by the present combinatorial approach to selectively inhibit gene expression. For instance, mutagenesis can provide GLC I A homologs which are able to bind other signal pathway proteins (or DNA) yet prevent propagation of the signal, e.g. the homologs can be dominant negative mutants.
Moreover, manipulation of certain domains of GLC 1 A by the present method can provide domains more suitable for use in fusion proteins.
In one embodiment, the variegated library of GLC 1 A variants is generated by combinatorial mutagenesis at the nucleic acid level, and is encoded by a variegated gene library. For instance, a mixture of synthetic oligonucleotides can be enzymatically ligated into gene sequences such that the degenerate set of potential GLC 1 A
sequences are expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g. for phage display) containing the set of GLC 1 A sequences therein.
There are many ways by which such libraries of potential GLC 1 A homologs can be generated from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be carried out in an automatic DNA synthesizer, and the synthetic genes then ligated into an appropriate expression vector. The purpose of a degenerate set of genes is to provide, in one mixture, all of the sequences encoding the desired set of potential GLC 1 A sequences. The synthesis of degenerate oligonucleotides is well known in the art (see for example, Narang, SA ( 1983) Tetrahedron 39:3; Itakura et al. ( 198 I ) Recombinant DNA, Proc 3rd Cleveland Sympos. Macromolecules, ed.
AG
Walton, Amsterdam: Elsevier ppg. 273-289; Itakura et al. { 1984) Annu. Rev.
Biochem.
53:323; Itakura et al. ( 1984) Science 198:1056; Ike et al. ( 1983) Nucleic Acid Res. I I :477.
Such techniques have been employed in the directed evolution of other proteins (see, for example, Scott et al. ( 1990) Science 249:386-390; Roberts et al. ( 1992) PNAS
89:2429-2433; Devlin et al. ( 1990) Science 249: 404-406; Cwirla et al. ( 1990) PNAS
87: 6378-6382;
as well as U.S. Patents Nos. 5,223,409, 5,198,346, and 5,096,815).
Likewise, a library of coding sequence fragments can be provided for a GLC 1 A clone in order to generate a variegated population of GLC 1 A
fragments for WO 98/20131 PCT/ITS97/20702 _ -39_ . -screening and subsequent selection of bioactive fragments. A variety of techniques are known in the art for generating such libraries, including chemical synthesis.
In one embodiment, a library of coding sequence fragments can be generated by (i) treating a double stranded PCR fragment of a GLC I A coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule; (ii) denaturing the double stranded DNA; (iii) renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products; (iv) removing single stranded portions from reformed duplexes by treatment with S 1 nuclease; and (v) ligating the resulting fragment library into an expression vector. By this exemplary method, an expression library can be derived which codes for N-terminal, C-terminal and internal fragments of various sizes.
A wide range of techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA
libraries for gene products having a certain property. Such techniques will be generally adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of GLC1A homologs. The most widely used techniques for screening large gene libraries typically comprises cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing 2o the combinatorial genes under conditions in which detection of a desired activity facilitates relatively easy isolation of the vector encoding the gene whose product was detected. Each of the illustrative assays described below are amenable to high through-put analysis as necessary to screen large numbers of degenerate GLC 1 A sequences created by combinatorial mutagenesis techniques.
Combinatorial mutagenesis has a potential to generate very large libraries of mutant proteins, e.g., in the order of 1026 molecules. Combinatorial libraries of this size may be technically challenging to screen even with high throughput screening assays. To overcome this problem, a new technique has been developed recently, recrusive ensemble 3o mutagenesis (REM), which allows one to avoid the very high proportion of non-functional proteins in a random library and simply enhances the frequency of functional proteins, thus decreasing the complexity required to achieve a useful sampling of sequence space. REM
is an algorithm which enhances the frequency of functional mutants in a library when an WO 98/20131 PCT/US97/20702 _ ' appropriate selection or screening method is employed (Arkin and Yourvan, 1992, PNAS
USA 89:7811-7815; Yourvan et al., 1992, Parallel Problem Solving from Natzrre, 2., In Maenner and Manderick, eds., Elsevir Publishing Co., Amsterdam, pp. 401-410;
Delgrave et al., 1993, Protein Engineering 6(3):327-331).
The invention also provides for reduction of the GLC 1 A proteins to generate mimetics, e.g. peptide or non-peptide agents, which are able to disrupt binding of a mammalian GLC 1 A polypeptide of the present invention with either upstream or downstream components. Thus, such mutagenic techniques as described above are also 1 o useful to map the determinants of the GLC 1 A proteins which participate in protein-protein interactions involved in, for example, binding of the subject GLC 1 A
polypeptide to proteins which may function upstream (including both activators and repressors of its activity) or to proteins or nucleic acids which may function downstream of the GLC 1 A
polypeptide, whether they are positively or negatively regulated by it. To illustrate, the critical residues of a subject GLC1A polypeptide which are involved in molecular recognition of a component upstream or downstream of a GLC 1 A can be determined and used to generate GLC 1 A-derived peptidomimetics which competitively inhibit binding of the authentic GLC 1 A protein with that moiety. By employing, for example, scanning mutagenesis to map the amino acid residues of each of the subject GLC 1 A proteins which are involved in 2o binding other extracellular proteins, peptidomimetic compounds can be generated which mimic those residues of the GLC 1 A protein which facilitate the interaction.
Such mimetics may then be used to interfere with the normal function of a GLC 1 A protein.
For instance, non-hydrolyzable peptide analogs of such residues can be generated using benzodiazepine (e.g., see Freidinger et al. in Peptides: Chemistry and Biology, G.R. Marshall ed., ESCOM
Publisher: Leiden, Netherlands, 1988), azepine (e.g., see Huffman et al. in Peptides:
Chemistry and Biology, G.R. Marshall ed., ESCOM Publisher: Leiden, Netherlands, 1988), substituted gamma lactam rings (Garvey et al. in Peptides: Chemistry and Biology, G.R.
Marshall ed., ESCOM Publisher: Leiden, Netherlands, 1988), keto-methylene pseudopeptides (Ewenson et al. ( 1986) J Med Chem 29:295; and Ewenson et al.
in Peptides:
3o Structure and Function (Proceedings of the 9th American Peptide Symposium) Pierce Chemical Co. Rockland, IL, 1985 ), b-turn dipeptide cores (Nagai et al. ( 1985) Tetrahedron Lett 26:647; and Sato et al. ( 1986) J Chem Soc Perkin Trans 1:1231 ), and b-aminoalcohols (Gordon et al. ( 1985) Biochem Biophys Res Commun 126:419; and Dann et al. ( 1986) Biochem Biophys Res Commun 134:71 ).

WO 98/20131 PCT/US97/20702 _ ' _41 _ .
4.4.1. Cells expressing recombinant GLClA polYpeptides This invention also pertains to a host cell transfected to express a recombinant form of the subject GLC 1 A polypeptides. The host cell may be any prokaryotic or eukaryotic cell. Thus, a nucleotide sequence derived from the cloning of GLC 1 A proteins, encoding all or a selected portion of the full-length protein, can be used to produce a recombinant form of a GLC 1 A polypeptide via microbial or eukaryotic cellular processes. Ligating the polynucleotide sequence into a gene construct, such as an expression vector, and transforming or transfecting into hosts, either eukaryotic (yeast, I o avian, insect or mammalian) or prokaryotic (bacterial) cells, are standard procedures used in producing other well-known proteins, e. g. MAP kinase, pg. 53, WT 1, PTP
phosphotases, SRC, and the like. Similar procedures, or modifications thereof, can be employed to prepare recombinant GLC 1 A polypeptides by microbial means or tissue-culture technology in accord with the subject invention.
IS
The recombinant GLC 1 A genes can be produced by ligating nucleic acid encoding a GLC 1 A protein, or a portion thereof, into a vector suitable for expression in either prokaryotic cells, eukaryotic cells, or both. Expression vectors for production of recombinant forms of the subj ect GLC 1 A polypeptides include plasmids and other vectors.
2o For instance, suitable vectors for the expression of a GLC 1 A polypeptide include plasmids of the types: pBR322-derived plasmids, pEMBL-derived plasmids, pEX-derived plasmids, pBTac-derived plasmids and pUC-derived plasmids for expression in prokaryotic cells, such as E. coli.
25 A number of vectors exist for the expression of recombinant proteins in yeast. For instance, YEP24, YIPS, YEPS 1, YEP52, pYES2, and YRP 17 are cloning and expression vehicles useful in the introduction of genetic constructs into S.
cerevisiae (see, for example, Broach et al. (1983) in Experimental Manipulation of Gene Expression, ed.
M. Inouye Academic Press, p. 83, incorporated by reference herein}. These vectors can 3o replicate in E. coli due the presence of the pBR322 ori, and in S.
cerevisiae due to the replication determinant of the yeast 2 micron plasmid. In addition, drug resistance markers such as ampicillin can be used. In an illustrative embodiment, a GLC 1 A
polypeptide is WO 98/20131 PCT/US97/20702 _ ' -42- .
produced recombinantly utilizing an expression vector generated by sub-cloning the coding sequence of one of the GLC 1 A genes represented in SEQ ID Nos: 1 and 2.
The preferred mammalian expression vectors contain both prokaryotic sequences, to facilitate the propagation of the vector in bacteria, and one or more eukaryotic transcription units that are expressed in eukaryotic cells. The pcDNAI/amp, pcDNAI/neo, pRc/CMV, pSV2gpt, pSV2neo, pSV2-dhfr, pTk2, pRSVneo, pMSG, pSVT7, pko-neo and pHyg derived vectors are examples of mammalian expression vectors suitable for transfection of eukaryotic cells. Some of these vectors are modified with sequences from to bacterial plasmids, such as pBR322, to facilitate replication and drug resistance selection in both prokaryotic and eukaryotic cells. Alternatively, derivatives of viruses such as the bovine papillomavirus (BPV-1 ), or Epstein-Barr virus (pHEBo, pREP-derived and p205) can be used for transient expression of proteins in eukaryotic cells. The various methods employed in the preparation of the plasmids and transformation of host organisms are well known in the art. For other suitable expression systems for both prokaryotic and eukaryotic cells, as well as general recombinant procedures, see Molecular Cloning A
Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1989) Chapters 16 and 17.
2o In some instances, it may be desirable to express the recombinant GLC 1 A
polypeptide by the use of a baculovirus expression system. Examples of such baculovirus expression systems include pVL-derived vectors (such as pVL1392, pVL1393 and pVL941 ), pAcUW-derived vectors (such as pAcUW 1 ), and pBlueBac-derived vectors (such as the 13-gal containing pBlueBac III).
When it is desirable to express only a portion of a GLC 1 A protein, such as a form lacking a portion of the N-terminus, i.e. a truncation mutant which lacks the signal peptide, it may be necessary to add a start codon (ATG) to the oligonucleotide fragment containing the desired sequence to be expressed. It is well known in the art that a 3o methionine at the N-terminal position can be enzymatically cleaved by the use of the enzyme methionine aminopeptidase (MAP). MAP has been cloned from E. coli (Ben-Bassat et al. (1987) J. Bacteriol. 169:751-757) and Salmonella typhimurium and its in vitro activity has been demonstrated on recombinant proteins (Miller et al. ( 1987) PNAS 84:2718-1722). Therefore, removal of an N-terminal methionine, if desired, can be achieved either WO 98/20131 PCT/US97/20702 _ ' _43_ in vivo by expressing GLC 1 A-derived polypeptides in a host which produces MAP (e.g., E. coli or CM89 or S. cerevisiae), or in vitro by use of purified MAP (e.g., procedure of Miller et al., supra).
In other embodiments transgenic animals, described in more detail below could be used to produce recombinant proteins.
4.4.2 Fusion proteins and Immuno eg-nsns In another embodiment, the coding sequences for the polypeptide can be incorporated as a part of a fusion gene including a nucleotide sequence encoding a different polypeptide. This type of expression system can be useful under conditions where it is desirable to produce an immunogenic fragment of a GLC 1 A protein. For example, the VP6 capsid protein of rotavirus can be used as an immunologic carrier protein for portions of the GLC 1 A polypeptide, either in the monomeric form or in the form of a viral particle. The nucleic acid sequences corresponding to the portion of a subject GLC 1 A
protein to which antibodies are to be raised can be incorporated into a fusion gene construct which includes coding sequences for a late vaccinia virus structural protein to produce a set of recombinant viruses expressing fusion proteins comprising GLC 1 A epitopes as part of the virion. It has been demonstrated with the use of immunogenic fusion proteins utilizing the Hepatitis B
2o surface antigen fusion proteins that recombinant Hepatitis B virions can be utilized in this role as well. Similarly, chimeric constructs coding for fusion proteins containing a portion of a GLC 1 A protein and the poliovirus capsid protein can be created to enhance immunogenicity of the set of polypeptide antigens (see, for example, EP
Publication No:
0259149; and Evans et al. (1989) Nature 339:385; Huang et al. (1988) J. Virol.
62:3855;
and Schlienger et al. ( 1992) J. Virol. 66:2).
The Multiple Antigen Peptide system for peptide-based immunization can also be utilized to generate an immunogen, wherein a desired portion of a GLCIA
polypeptide is obtained directly from organo-chemical synthesis of the peptide onto an oligomeric branching lysine core (see, for example, Posnett et al. ( 1988) JBC
263:1719 and Nardelli et al. ( 1992) J. Immunol. 148:914). Antigenic determinants of GLC 1 A proteins can also be expressed and presented by bacterial cells.

WO 98/20131 PCT/US97/20702 _ ' In addition to utilizing fusion proteins to enhance immunogenicity, it is widely appreciated that fusion proteins can also facilitate the expression of proteins, and accordingly, can be used in the expression of the GLC 1 A polypeptides of the present invention. For example, GLC 1 A polypeptides can be generated as glutathione-S-transferase (GST-fusion) proteins. Such GST-fusion proteins can enable easy purification of the GLC 1 A polypeptide, as for example by the use of glutathione-derivatized matrices (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al. (N.Y.:
John Wiley & Sons, 1991 )).
In another embodiment, a fusion gene coding for a purification leader sequence, such as a poly-(His)/enterokinase cleavage site sequence at the N-terminus of the desired portion of the recombinant protein, can allow purification of the expressed fusion protein by affinity chromatography using a Ni '-~ metal resin. The purification leader sequence can then be subsequently removed by treatment with enterokinase to provide the purified protein (e.g., see Hochuli et al. ( 1987) J. Chromatography 411:177;
and Janknecht et al. PNAS 88:8972).
Techniques for making fusion genes are known to those skilled in the art.
Essentially, the joining of various DNA fragments coding for different polypeptide 2o sequences is performed in accordance with conventional techniques, employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers.
Alternatively, PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed to generate a chimeric gene sequence (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al. John Wiley & Sons:
1992).
3o 4.4.3. Antibodies Another aspect of the invention pertains to an antibody specifically reactive with a GLC 1 A protein. For example, by using immunogens derived from a GLC 1 A protein, e.g. based on the cDNA sequences, anti-protein/anti-peptide antisera or monoclonal antibodies can be made by standard protocols (See, for example, Antibodies:

WO 98/20131 PCTlUS97/20702 _ ' A Laboratory Manual ed. by Harlow and Lane (Cold Spring Harbor Press: 1988)).
A
mammal, such as a mouse, a hamster or rabbit can be immunized with an immunogenic form of the peptide (e.g., a GLC 1 A poIypeptide or an antigenic fragment which is capable of eliciting an antibody response, or a fusion protein as described above).
Techniques for conferring immunogenicity on a protein or peptide include conjugation to carriers or other techniques well known in the art. An immunogenic portion of a GLC1A protein can be administered in the presence of adjuvant. The progress of immunization can be monitored by detection of antibody titers in plasma or serum.
Standard ELISA or other immunoassays can be used with the immunogen as antigen to 1 o assess the levels of antibodies. In a preferred embodiment, the subject antibodies are immunospecific for antigenic determinants of a GLC 1 A protein of a mammal, e.g.
antigenic determinants of a protein represented by SEQ ID No: 2 or closely related homologs (e.g. at least 92% homologous, and more preferably at least 94%
homologous).
is Following immunization of an animal with an antigenic preparation of a GLC 1 A polypeptide, anti-GLC 1 A antisera can be obtained and, if desired, polyclonal anti-GLC 1 A antibodies isolated fram the serum. To produce monoclonal antibodies, antibody-producing cells (lymphocytes) can be harvested from an immunized animal 2o and fused by standard somatic cell fusion procedures with immortalizing cells such as myeloma cells to yield hybridoma cells. Such techniques are well known in the art, and include, for example, the hybridoma technique (originally developed by Kohler and Milstein, ( 1975) Nature, 256: 495-497), the human B cell hybridoma technique (Kozbar et al., (1983) Immunology Today, 4: 72), and the EBV-hybridoma technique to produce 25 human monoclonal antibodies (Cole et al., (1985) Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc. pp. 77-96). Hybridoma cells can be screened immunochemically for production of antibodies specifically reactive with a GLC

polypeptide of the present invention and monoclonal antibodies isolated from a culture comprising such hybridoma cells.
The term antibody as used herein is intended to include fragments thereof which are also specifically reactive with one of the subj ect mammalian GLC 1 A
polypeptides. Antibodies can be fragmented using conventional techniques and the WO 98/20131 PCT/LTS97/20702 _ ' -46- ' fragments screened for utility in the same manner as described above for whole antibodies. For example, F(ab)~ fragments can be generated by treating antibody with pepsin. The resulting F(ab)~ fragment can be treated to reduce disulfide bridges to produce Fab fragments. The antibody of the present invention is further intended to include bispecific and chimeric molecules having affinity for a GLC 1 A
protein conferred by at least one CDR region of the antibody.
Antibodies which specifically bind GLC1A epitopes can also be used in immunohistochemicaI staining of tissue samples in order to evaluate the abundance and i 0 pattern of expression of each of the subject GLC 1 A polypeptides. Anti-antibodies can be used diagnostically in immuno-precipitation and immuno-blotting to detect and evaluate GLC 1 A protein levels in tissue as part of a clinical testing procedure.
For instance, such measurements can be useful in predictive valuations of the onset or progression of proliferative disorders. Likewise, the ability to monitor GLC 1 A protein levels in an individual can allow determination of the efficacy of a given treatment regimen for an individual afflicted with such a disorder. The level of GLC 1 A
polypeptides may be measured from cells in bodily fluid, such as in samples of cerebral spinal fluid or amniotic fluid, or can be measured in tissue, such as produced by biopsy.
Diagnostic assays using anti-GLC 1 A antibodies can include, for example, immunoassays designed to aid in early diagnosis of a degenerative disorder, particularly ones which are manifest at birth. Diagnostic assays using anti-GLC 1 A
polypeptide antibodies can also include immunoassays designed to aid in early diagnosis and phenotyping neoplastic or hyperplastic disorders.
Another application of anti-GLC 1 A antibodies of the present invention is in the immunological screening of cDNA libraries constructed in expression vectors such as gtl 1, gtl8-23, ZAP, and ORFB. Messenger libraries of this type, having coding sequences inserted in the correct reading frame and orientation, can produce fusion proteins. For instance, gtl 1 will produce fusion proteins whose amino termini consist of 13-galactosidase amino acid sequences and whose carboxy termini consist of a foreign polypeptide.
Antigenic epitopes of a GLC 1 A protein, e.g. other orthologs of a particular GLC 1 A protein or other paralogs from the same species, can then be detected with antibodies, as, for example, reacting nitrocellulose filters lifted from infected plates with anti-antibodies. Positive phage detected by this assay can then be isolated from the infected WO 98/20131 PCT/CTS97/20702 _ ' _47_ plate. Thus, the presence of GLC 1 A homologs can be detected and cloned from other animals, as can alternate isoforms (including splicing variants) from humans.
4.5 Methods of Treating Disease There may be a variety of pathological conditions for which GLC 1 A
therapeutics of the present invention can be used in treatment. One clear example is various forms of glaucoma.
1 o A "GLC 1 A therapeutic," whether an antagonist or agonist of wild type GLC IA, can be, as appropriate, any of the preparations described above, including isolated polypeptides, gene therapy constructs, antisense molecules, peptidomimetics, small molecules, non-nucleic acid, non-peptidic or agents identified in the drug assays provided herein.
Since, in some cases, genes may be upregulated in a disease state and in other cases may be downregulated, it will be desirable to activate and/or potentiate or suppress and/or downmodulate GLC 1 A bioactivity depending on the condition to be treated using the techniques compounds and methods described herein. Some genes may be underexpressed in certain disease states. The activity of GLC 1 A gene products may he in some way impaired, leading to the development of disease symptoms. Such down-regulation of GLC 1 A gene expression or decrease in the activity of a GLC 1 A
protein may have a causative or exacerbating effect on the disease state.
Among the approaches which may be used to ameliorate disease symptoms involving the misexpression of a GLC 1 A gene are, for example, antisense, ribozyme, and triple helix molecules described above. Compounds that compete with a GLC 1 A
protein for binding to upstream or downstream elements in a signaling cascade will antagonize a GLC 1 A protein , thereby inducing a therapeutic effect. Examples of suitable compounds 3o include the antagonists or homologues described in detail above. In other instances, the increased expression or bioactivity of a GLC I A protein may be desirable and may be accomplished by, for example the use of the GLC 1 A agonists or mimetics or by gene replacement therapy, as described herein.

WO 98/20131 PCT/LTS97/20702 _ ' _48_ .
Compounds identified as increasing or decreasing GLC I A gene expression or protein activity can be administered to a subject at therapeutically effective dose to treat or ameliorate symptoms associated with glaucoma.
4.5.1. Effective Dose Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LDSp (the dose lethal to 50% of the population) and the EDSp (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LDSp/ED50.
Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage t 5 to uninfected cells and, thereby, reduce side effects.
The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the EDSp with little or 2o no toxicity. The dosage may vary within this range denendinu vmnn rhP
~inca~P f.".", employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the ICSp (i.e., the concentration of the test compound 25 which achieves a half maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
Levels in plasma may be measured, for example, by high performance liquid chromatography.
5.2. Formulation and Use 30 Pharmaceutical compositions for use in accordance with the present invention may be formulated in conventional manner using one or more physiologically acceptable carriers or excipients. Thus, the compounds and their physiologically acceptable salts and solvates may be formulated for administration by, for example, injection, WO 98/20131 PCT/US97/20702 _ ' inhalation or insufflation (either through the mouth or the nose) or oral, buccal, parenteral or rectal administration.
For such therapy, the oligomers of the invention can be formulated for a variety of loads of administration, including systemic and topical or localized administration. Techniques and formulations generally may be found in Remmington's Pharmaceutical Sciences, Meade Publishing Co., Easton, PA. For systemic administration, injection is preferred, including intramuscular, intravenous, intraperitoneal, and subcutaneous. For injection, the oligomers of the invention can be formulated in liquid I o solutions, preferably in physiologically compatible buffers such as Hank's solution or Ringer's solution. In addition, the oligomers may be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms are also included.
For oral administration, the pharmaceutical compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or 2o wetting agents (e. g.) sodium lauryl sulphate). The tablets may be coated by methods well known in the art. Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives or hydrogenated edible fats);
emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, flavoring, coloring and sweetening agents as appropriate.
Preparations for oral administration may be suitably formulated to give controlled release of the active compound.

WO 98/20131 PCT/US97/20702 _ ' _50_ For buccal administration the compositions may take the form of tablets or lozenges formulated in conventional manner.
For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g. gelatin I o for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
The compounds may be formulated for parenteral administration by injection. e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in mufti-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g.) sterile pyrogen-free water, 2o before use.
The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable 3o polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
Systemic administration can also be by transmucosal or transdermal means.
For transmucosal or transdermal administration, penetrants appropriate to the barrier to be WO 98/20131 PCT/US97/20702 _ ' permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration bile salts and fusidic acid derivatives.
In addition, detergents may be used to facilitate permeation. Transmucosal administration may be through nasal sprays or using suppositories. For topical administration, the oligomers of the invention are formulated into ointments, salves, gels, or creams as generally known in the art.
In clinical settings, the gene delivery systems for the therapeutic GLC 1 A
gene can be introduced into a patient by any of a number of methods, each of which is familiar in the art. For instance, a pharmaceutical preparation of the gene delivery system can be introduced systemically, e.g. by intravenous injection, and specific transduction of the protein in the target cells occurs predominantly from specificity of transfection provided by the gene delivery vehicle, cell-type or tissue-type expression due to the transcriptional regulatory sequences controlling expression of the receptor gene, or a combination thereof.
~ 5 In other embodiments, initial delivery of the recombinant gene is more limited with introduction into the animal being quite localized. For example, the gene delivery vehicle can be introduced by catheter (see U.S. Patent 5,328,470) or by stereotactic injection (e.g.
Chen et al. ( 1994) PNAS 91: 3054-3057). A GLC 1 A gene, such as any one of the sequences represented in the group consisting of SEQ ID NO: 1 or 2, or a sequence homologous 2o thereto can be delivered in a gene therapy construct by electroporation using techniques described, for example, by Dev et al. ((1994) Cancer Treat Rev 20:105-115).
Gene therapy vectors comprised of viruses that provide specific effective and highly localized treatment of eye diseases are described in Published International Patent Application No. WO
95/34580 to U. Eriksson et al.
The pharmaceutical preparation of the gene therapy construct can consist essentially of the gene delivery system in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery system can be produced intact from recombinant cells, e.g. retroviral 3o vectors, the pharmaceutical preparation can comprise one or more cells which produce the gene delivery system.

WO 98/20131 PCT/LJS97/207U2 _ ' The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack.
The pack or dispenser device may be accompanied by instructions for administration.
Diagnostic and Prognostic Assay The following mutations have been identified in glaucoma patients: ( 1 ) GLN368STOP, (2) GLY364VAL, (3) TYR437HIS, (4) ILE477ASN, (5) a 6 by insertion to in codon 396, (6) VAL495ILE, (7) ALA445VAL, (8) THR377MET, (9) GLN19HIS,
(10) ARG82CYS. In addition, the following polymorphisms have been identified in normal individuals: (1) TYR347TYR, (2) THR325THR, (3) VAL439VAL, (4) ARG398LYS and (5) Prol3Pro.
The prevalence of the sequence changes reported herein, coupled with the prevalence of glaucoma in the general population, suggests that mutations in the GLC 1 A
gene cause glaucoma in nearly one hundred thousand patients in the United States.
Therefore, the GLC 1 A gene is the most common molecularly recognizable form of blindness. For comparison, only two thousand people in the United States would be 2o expected to harbor mutations in the rhodopsin gene, which is the most common form of molecularly recognizable retinitis pigmentosa. The discovery of specific glaucoma-causing mutations allows for identification of subjects at high risk for this form of glaucoma before significant visual loss has occurred, so that they can be directed toward sight-saving therapy.
In the diagnostic and prognostic assays described herein, in addition to the GLC 1 A nucleic acid molecules and polypeptides described above, the present invention provides for the use of nucleic acids comprising at least a portion of the nucleic acid sequence shown in SEQ ID Nos: 1 or 2 or polypeptides comprising at least a portion of the amino acid sequence shown in SEQ ID No: 3.
The present method provides a method for determining if a subject is at risk for glaucoma. In preferred embodiments, the methods can be characterized as comprising detecting, in a sample of cells from the subject, the presence or absence of a genetic lesion characterized by at least one of (i) an alteration affecting the integrity of a gene encoding WO 98/20131 PCT/US97/20702 ' _5 i_ . -a GLC I A-protein, or (ii) the mis-expression of the GLC 1 A gene. To illustrate, such genetic lesions can be detected by ascertaining the existence of at least one of (i) a deletion of one or more nucleotides from a GLC I A gene, (ii) an addition of one or more nucleotides to a GLC I A gene, (iii) a substitution of one or more nucleotides of a GLC I A
gene, (iv) a gross chromosomal rearrangement of a GLC 1 A gene, (v) a gross alteration in the level of a messenger RNA transcript of a GLC 1 A gene, (vii) aberrant modification of a GLC I A gene, such as of the methylation pattern of the genomic DNA, (vii) the presence of a non-wild type splicing pattern of a messenger RNA transcript of a GLC I A gene, (viii) a non-wild type level of a GLC I A-protein, (ix) allelic loss of a GLC I A gene, and (x) inappropriate t o post-translational modification of a GLC 1 A-protein. As set out below, the present invention provides a large number of assay techniques for detecting lesions in a GLC I A
gene, and importantly, provides the ability to discern between different molecular causes underlying GLC I A-dependent aberrant cell growth, proliferation andlor differentiation.
In an exemplary embodiment, there is provided a nucleic acid composition comprising a (purified) oligonucleotide probe including a region of nucleotide sequence which is capable of hybridizing to a sense or antisense sequence of a GLC 1 A
gene, such as represented by any of SEQ ID Nos: l and 2, or naturally occurring mutants thereof, or S' or 3' flanking sequences or intronic sequences naturally associated with the subject GLC 1 A
2o genes or naturally occurring mutants thereof. The nucleic acid of a cell is rendered accessible for hybridization, the probe is exposed to nucleic acid of the sample, and the hybridization of the probe to the sample nucleic acid is detected. Such techniques can be used to detect lesions at either the genomic or mRNA level, including deletions, substitutions, etc., as well as to determine mRNA transcript levels.
As set out above, one aspect of the present invention relates to diagnostic assays for determining, in the context of cells isolated from a patient, if mutations have arisen in one or more GLC 1 A of the sample cells. The present method provides a method for determining if a subject is at risk for a disorder characterized by aberrant cell 3o proliferation and/or differentiation. In preferred embodiments, the method can be generally characterized as comprising detecting, in a sample of cells from the subj ect, the presence or absence of a genetic lesion characterized by an alteration affecting the integrity of a gene encoding a GLC 1 A. To illustrate, such genetic lesions can be detected by ascertaining the WO 98/20131 PCT/US97/20702 ' existence of at least one of (i) a deletion of one or more nucleotides from a GLC 1 A-gene, (ii) an addition of one or more nucleotides to a GLC 1 A-gene, (iii) a substitution of one or more nucleotides of a GI_C 1 A-gene, and (iv) the presence of a non-wild type splicing pattern of a messenger RNA transcript of a GLC 1 A-gene. As set out below, the present invention provides a large number of assay techniques for detecting lesions in genes, and importantly, provides the ability to discern between different molecular causes underlying GLC 1 A-dependent aberrant cell growth, proliferation and/or differentiation.
In certain embodiments, detection of the lesion comprises utilizing the i o probe/primer in a polymerase chain reaction (PCR) (see, e.g. U.S. Patent Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al. ( 1988) Science 241:1077-1080; and Nakazawa et al. ( 1994) PNAS 91:360-364), the latter of which can be particularly useful for detecting point mutations in the GLC 1 A-gene (see Abravaya et al. ( 1995) Nuc Acid Res 23:675-682).
In a merely illustrative embodiment, the method includes the steps of (i) collecting a sample of cells from a patient, (ii) isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, (iii) contacting the nucleic acid sample with one or more primers which specifically hybridize to a GLC 1 A gene under conditions such that hybridization and amplification of the GLC 1 A-gene (if present) occurs, and (iv) detecting the presence or 2o absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.
Alternative amplification methods include: self sustained sequence replication (Guatelli, J.C. et al., 1990, Proc. Natl. Acad. Sci. USA 87:1874-1878), transcriptional amplification system (Kwoh, D.Y. et al., 1989, Proc. Natl.
Acad. Sci. USA
86:1173-1177), Q-Beta Replicase (Lizardi, P.M. et al., 1988, Bio/Technology 6:1197), or any other nucleic acid amplification method, followed by the detection of the amplified 3o molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.

WO 98/20131 PCT/LTS97/20702 ' -55- ' In a preferred embodiment of the subject assay, mutations in a GLC 1 A gene from a sample cell are identified by alterations in restriction enzyme cleavage patterns. For example, sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis. Moreover, the use of sequence specific ribozymes (see, for example. U.S.
Patent No. 5,498,531 ) can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
In yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence the GLC 1 A gene and detect mutations by comparing the sequence of the sample GLC 1 A with the corresponding wild-type (control) sequence. Exemplary sequencing reactions include those based on techniques developed by Maxim and Gilbert (Proc. Natl Acad Sci USA ( 1977) 74:560) or Sanger (Sanger et al (1977) Proc. Nat. Acad. Sci 74:5463). It is also contemplated that any of a variety of automated sequencing procedures may be utilized when performing the subject assays (Biotechnigues (1995) 19:448), including by sequencing by mass spectrometry (see, for example PCT publication WO 94/16101; Cohen et al. (1996) Adv Chromatogr 36:127-162;
and Griffin et al. (1993) Appl Biochem Biotech~tol 38:147-159). 1t will be evident to one skilled in the art that, for certain embodiments, the occurrence of only one, two or three of 2o the nucleic acid bases need be determined in the sequencing reaction. For instance, A-tract or the like, e.g., where only one nucleic acid is detected, can be carried out.
In a further embodiment, protection from cleavage agents (such as a nuclease, hydroxylamine or osmium tetroxide and with piperidine) can be used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes (Myers, et al. (1985) Science 230:1242). In general, the art technique of "mismatch cleavage" starts by providing heteroduplexes of formed by hybridizing (labelled) RNA or DNA containing the wild-type GLC 1 A sequence with potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent which cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with S 1 nuclease to enzymatically digesting the mismatched regions. In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated WO 98/20131 PCT/US97/20702 ' with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation.
See, for example, Cotton et al ( 1988) Proc. Natl Acad Sci USA 85:4397; Saleeba et al ( 1992) Methods Enzymod. 217:286-295. In a preferred embodiment, the control DNA or RNA can be labeled for detection.
In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA
(so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in GLC 1 A cDNAs obtained from samples of cells. For example, the mutt enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA
glycosylase from HeLa cells cleaves T at G/T mismatches (Hsu et al. ( 1994) Carcinogenesis 15:1657-1662).
According to an exemplary embodiment, a probe based on a GLC 1 A sequence, e.g., a wild-type GLC 1 A sequence, is hybridized to a cDNA or other DNA product from a test cell(s).
The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, for example, U.S.
Patent No. 5,459,039.
In other embodiments, alterations in electrophoretic mobility is used to identify mutations in GLC 1 A genes. For example, single strand conformation polymorphism (SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids (Orita et al. (1989} Proc Natl.
Acad. Sci USA
86:2766, see also Cotton (1993) Mutat Res 285:125-144; and Hayashi (1992) Genet Anal Tech Appl 9:73-79). Single-stranded DNA fragments of sample and control GLC 1 A nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labelled or detected with labelled probes. The sensitivity of the assay may be enhanced by 3o using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In a preferred embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al. ( 1991 ) Trends Genet 7:5).

WO 98/20131 PCT/US97/20702 _ ' In yet another embodiment the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al ( 1985) Nature 313:495). When DGGE
is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 by of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing agent gradient to identify differences in the mobility of control and sample DNA (Rosenbaum and Reissner (1987) Biophys Chem 265:12753).
Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al. ( 1986) Nature 324:163 ); Saiki et al ( 1989) Proc. Natl Acad. Sci USA 86:6230}. Such allele specific oligonucleotide hybridization techniques may be used to test one mutation per reaction when oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labelled target DNA.
Alternatively, allele specific amplification technology which depends on selective PCR amplification may be used in conjunction with the instant invention.
Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization) (Gibbs et al ( 1989) Nucleic Acids Res. 17:2437-2448) or at the extreme 3' end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner ( 1993) Tibtech 11:238. In addition it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection (Gasparini et al ( 1992) Mol. Cell Probes 6:1 ). It is anticipated that in certain 3o embodiments amplification may also be performed using Taq ligase for amplification {Barany ( 199 i ) Proc. Natl. Acad Sci USA 88:189). In such cases, ligation will occur only if there is a perfect match at the 3' end of the 5' sequence making it possible to detect the WO 98/20131 PCT/US97/20702 ' _58_ .
presence of a known mutation at a specific site by looking for the presence or absence of amplification.
For mutations that produce premature termination of protein translation, the protein truncation test (PTT) offers an efficient diagnostic approach (Roest, et. al., ( 1993) Hum. Mol. Genet. 2:1719-21; van der Luijt, et. aL, ( 1994) Genomics 20:1-4).
For PTT, RNA is initially isolated from available tissue and reverse-transcribed, and the segment of interest is amplified by PCR. The products of reverse transcription PCR are then used as a template for nested PCR amplification with a primer that contains an RNA
polymerase promoter and a sequence for initiating eukaryotic translation. After amplification of the region of interest, the unique motifs incorporated into the primer permit sequential in vitro transcription and translation of the PCR products. Upon sodium dodecyl sulfate-polyacrylamide gel electrophoresis of translation products, the appearance of truncated polypeptides signals the presence of a mutation that causes premature termination of translation. In a variation of this technique, DNA (as opposed to RNA) is used as a PCR
template when the target region of interest is derived from a single exon.
Another embodiment of the invention provides for a nucleic acid composition comprising a (purified) oligonucleotide probe including a region of nucleotide 2o sequence which is capable of hybridizing to a sense or antisense sequence of a GLC 1 A-gene, or naturally occurring mutants thereof, or 5' or 3' flanking sequences or intronic sequences naturally associated with the subject GLC 1 A-genes or naturally occurring mutants thereof. The nucleic acid of a cell is rendered accessible for hybridization, the probe is exposed to nucleic acid of the sample, and the hybridization of the probe to the sample nucleic acid is detected. Such techniques can be used to detect lesions at either the genomic or mRNA level, including deletions, substitutions, etc., as well as to determine mRNA transcript levels. Such oligonucleotide probes can be used for both predictive and therapeutic evaluation of allelic mutations which might be manifest in, for example, neoplastic or hyperplastic disorders (e.g. aberrant cell growth).
The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose WO 98/20131 PCT/US97/20702 _ ' -59- ' patients exhibiting symptoms or family history of a disease or illness involving a GLC 1 A
gene.
Any cell type or tissue, preferably monocytes, endothelial cells, or smooth muscle cells, in which the GLC 1 A is expressed may be utilized in the diagnostics described below. For example, a subject's bodily fluid (e.g. blood) can be obtained by known techniques (e.g. venipuncture). Alternatively, nucleic acid tests can be performed on dry samples (e.g. hair or skin). Fetal nucleic acid samples can be obtained from maternal blood as described in International Patent Application No. WO 91 /07660 to Bianchi.
Alternatively, amniocytes or chorionic villi may be obtained for performing prenatal testing.
Diagnostic procedures may also be performed in situ directly upon tissue sections (fixed and/or frozen) of patient tissue obtained from biopsies or resections, such that no nucleic acid purification is necessary. Nucleic acid reagents may be used as probes t 5 and/or primers for such in situ procedures (see, for example, Nuovo, G.J., 1992, PCR in situ hybridization: protocols and applications, Raven Press, NY).
In addition to methods which focus primarily on the detection of one nucleic acid sequence, profiles may also be assessed in such detection schemes.
Fingerprint profiles 2o may be generated, for example, by utilizing a differential display procedure, Northern analysis and/or RT-PCR.
Antibodies directed against wild type or mutant GLC 1 A proteins, which are discussed, above, may also be used in disease diagnostics and prognostics.
Such diagnostic 25 methods, may be used to detect abnormalities in the level of GLC 1 A
protein expression, or abnormalities in the structure andlor tissue, cellular, or subcellular location of GLC 1 A
protein. Structural differences may include, for example, differences in the size, electronegativity, or antigenicity of the mutant GLC 1 A protein relative to the normal GLC 1 A protein. Protein from the tissue or cell type to be analyzed may easily be detected 30 or isolated using techniques which are well known to one of skill in the art, including but not limited to western blot analysis. For a detailed explanation of methods for carrying out western blot analysis, see Sambrook et a1, 1989, supra, at Chapter 18. The protein detection and isolation methods employed herein may also be such as those described in Harlow and WO 98/20131 PCT/US97/20702 ' Lane, for example, (Harlow, E. and Lane, D., 1988, "Antibodies: A Laboratory Manual", Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York), which is incorporated herein by reference in its entirety.
This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled antibody (see below) coupled with light microscopic, flow cytometric, or fluorimetric detection. The antibodies (or fragments thereof) useful in the present invention may, additionally, be employed histologically, as in immunofluorescence or immunoelectron microscopy, for in situ detection of GLC

1 o proteins. In situ detection may be accomplished by removing a histological specimen from a patient, and applying thereto a labeled antibody of the present invention.
The antibody (or fragment) is preferably applied by overlaying the labeled antibody (or fragment) onto a biological sample. Through the use of such a procedure, it is possible to determine not only the presence of the GLC 1 A protein, but also its distribution in the examined tissue. Using ~ 5 the present invention, one of ordinary skill will readily perceive that any of a wide variety of histological methods (such as staining procedures) can be modified in order to achieve such in situ detection.
Often a solid phase support or carrier is used as a support capable of 2o binding an antigen or an antibody. Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite. The nature of the carrier can be either soluble to some extent or insoluble for the purposes of the present invention.
The support material may have virtually any possible structural configuration so long as the coupled 25 molecule is capable of binding to an antigen or antibody. Thus, the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tube, or the external surface of a rod. Alternatively, the surface may be flat such as a sheet, test strip, etc. Preferred supports include polystyrene beads. Those skilled in the art will know many other suitable carriers for binding antibody or antigen, or will be able to ascertain the same 3o by use of routine experimentation.
One means for labeling an anti-GLC 1 A protein specific antibody is via linkage to an enzyme and use in an enzyme immunoassay (EIA) (Volley, "The Enzyme Linked Immunosorbent Assay (ELISA)", Diagnostic Horizons 2:1-7, 1978, Microbiological WO 98/20131 PCT/US97/20702 ' -61- ' Associates Quarterly Publication, Walkersville, MD; Voller, et al., J. Clin.
Pathol. 31:507-520 ( 1978); Butler, Meth. Enzymol. 73:482-523 ( 1981 ); Maggio, (ed.) Enzyme Immunoassay, CRC Press, Boca Raton, FL, 1980; Ishikawa, et al., (eds.) Enzyme Immunoassay, Kgaku Shoin, Tokyo, 1981 ). The enzyme which is bound to the antibody will react with an appropriate substrate, preferably a chromogenic substrate, in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorimetric or by visual means. Enzymes which can be used to detectably label the antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate, dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase. The detection can be accomplished by colorimetric methods which employ a chromogenic substrate for the enzyme. Detection may also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards.
Detection may also be accomplished using any of a variety of other immunoassays. For example, by radioactively labeling the antibodies or antibody 2o fragments, it is possible to detect fingerprint gene wild type or mutant peptides through the use of a radioimmunoassay (RIA) (see, for example, Weintraub, B., Principles of Radioimmunoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March, 1986, which is incorporated by reference herein).
The radioactive isotope can be detected by such means as the use of a gamma counter or a scintillation counter or by autoradiography.
It is also possible to label the antibody with a fluorescent compound. When the fluorescently labeled antibody is exposed to light of the proper wave length, its presence can then be detected due to fluorescence. Among the most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o_-phthaldehyde and fluorescamine.

WO 98/20131 PCT/US97/20702 ' The antibody can also be detestably labeled using fluorescence emitting metals such as 152Eu, or others of the lanthanide series. These metals can be attached tn the antibody using such metal chelating groups as diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA).
The antibody also can be detestably labeled by coupling it to a chemiluminescent compound. The presence of the chemiluminescent-tagged antibody is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction. Examples of particularly useful chemiluminescent labeling compounds I o are luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.
Likewise, a bioluminescent compound may be used to label the antibody of the present invention. Bioluminescence is a type of chemiluminescence found in biological systems in, which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence. Important bioluminescent compounds for purposes of labeling are luciferin, luciferase and aequorin.
2o Moreover, it will be understood that any of the above methods for detecting alterations in a GLC 1 A gene or gene product can be used to monitor the course of treatment or therapy.
4.7. Drug Screening Assay Based on the identification of specific GLC 1 A mutations in human beings with glaucoma, one of ski 11 in the art can use any of a number of standard assays to screen for drugs, which will interfere with or otherwise prevent the development of glaucoma. By addressing the molecular basis of glaucoma, these agents are expected to be superior to existing therapies.
For example, identification of the precise phenotype associated with these mutations can be used to identify functionally important regions of the protein. These specific mutations can then be used in other experiments which will include overexpression in cell lines and the creation of transgenic animals. Ideally, one could WO 98/ZOI31 PCT/US97/2070Z _ ' identify mutations which reproducibly cause glaucoma at very different times in the person's life and then be able to show that these mutations had similar differences of effect in a cellular expression system or a transgenic animal.
Although GLC 1 A likely causes glaucoma through its actions within the trabucular meshwork or ciliary body, it will be important to characterize the expression of this gene throughout the eye and indeed throughout the body to make sure that an important part of the pathogenesis of GLC 1 A-linked glaucoma is not caused by an indirect mechanism. Therefor identifying tissue distribution and the developmental time 1 o course of gene expression (at both protein and DNA levels) using standard procedures will be important.
In addition, identification of proteins that interact with the GLC I A gene product and genes encoding the proteins can performed, since proteins that interact with 15 GLC I A gene product will be important targets for involvement in the pathogenesis of various types of glaucoma.
Further, studies will be undertaken to discover whether mutations known to cause glaucoma in human beings alter protein trafficking in tissue culture as well as 2o animal models, since one mechanism through which mutations in the GLC 1 A
gene could cause disease would be to alter the expression of other important gene products.
This can occur by affecting overall protein trafficking within the cell caused for example by increased removal of mutant pxoteins at the level of the endoplasmic reticulum.
25 In addition to the pathogenic studies described above, the precise location and sequence of the introns within the GLC I A gene can be identified so that improved PCR-based screening assays can be developed to fully assay the coding sequences of the GLC 1 A in patients with glaucoma. Also, sequences that participate in the regulation of the GLC I A gene will be important to characterize. For example, some glaucoma-3o causing mutations in the GLC 1 A gene exist in upstream sequences which alter the expression of the gene either positively or negatively. Further, classes of genes that are similarly regulated may be important in glaucoma (for example genes that share upstream regulatory sequences with the GLC 1 A gene).

WO 98/20131 PCT/US97/20702 _ Further understanding of the pathogenesis of glaucoma is useful for identifying new classes of drugs which can be useful in the treatment of glaucoma. For example, preliminary data suggest that the GLC 1 A gene is induced by exposure of cells to steroids. Therefore, drugs which are capable of blocking this steroid effect can be tested for the ability to prevent or delay the development of glaucoma.
As further described below, in vitro assays which are suitable for very high throughput screening of compounds can be performed. As the simplest example of this approach, one could use antibodies to the GLC 1 A gene product to develop a simple ELISA assay for the induction of the GLC 1 A gene product and then perform this assay in a 96 well microtiter plate format to screen a large number of drugs for the efficacy in blocking the steroid induction of the gene product. In this way, automated methods could be used to screen several thousand potentially therapeutic compounds for efficacy.
Also, knowledge of the structure/function of the GLC 1 A gene immediately suggests other genes which might be involved in glaucoma. Such clues will come from studies of homology, evolution, evaluation of structural motifs within the gene, and genetic studies using analyses designed to identify genes causing polygenic disease.
In the original linkage study described herein, it was recognized that 3 of 22 obligate carriers of the glaucoma gene failed to manifest a severe glaucoma phenotype. This information suggests that other genes are capable of mitigating the effect of the GLC 1 A mutation. One powerful way to search for such mitigator genes is to express a glaucoma-causing gene in different backgrounds. This can be done by creating transgenic animals and then breeding the glaucoma-causing gene on different genetic mouse strains. If the phenotype is altered in different strains these animals can be back crossed in such a way that the mitigating gene can be identified.
Some of the assays mentioned above, will now be described in further detail below.
4.7.1 Cell-free assays WO 98/20131 PCTlUS97/20702 ' In many drug screening programs which test libraries of compounds and natural extracts, high throughput assays are desirable in order to maximize the number of compounds surveyed in a given period of time. Assays which are performed in cell-free systems, such as may be derived with purified or semi-purified proteins, are often preferred as "primary" screens in that they can be generated to permit rapid development and relatively easy detection of an alteration in a molecular target which is mediated by a test compound. Moreover, the effects of cellular toxicity and/or bioavailability of the test compound can be generally ignored in the in vitro system, the assay instead being focused primarily on the effect of the drug on the molecular target as may be manifest in an alteration of binding affinity with upstream or downstream elements.
Accordingly, in an exemplary screening assay of the present invention, the compound of interest is contacted with proteins which may function upstream (including both activators and repressors of its activity) or to proteins or nucleic acids which may function downstream of the polypeptide, whether they are positively or negatively regulated by it. To the mixture of the compound and the upstream or downstream element is then added a composition containing a GLC 1 A polypeptide. Detection and quantification of complexes of GLC 1 A
with it's upstream or downstream elements provide a means for determining a compound's efficacy at inhibiting (or potentiating) complex formation between GLC 1 A and a GLC 1 A-binding element. The efficacy of the compound can be assessed by generating dose response curves 2o from data obtained using various concentrations of the test compound.
Moreover, a control assay can also be performed to provide a baseline for comparison. In the control assay, isolated and purified GLC 1 A polypeptide is added to a composition containing the GLC 1 A-binding element, and the formation of a complex is quantitated in the absence of the test compound.
Complex formation between the GLC I A polypeptide and a GLC 1 A binding element may be detected by a variety of techniques. Modulation of the formation of complexes can be quantitated using, for example, detectably labeled proteins such as radiolabeled, fluorescently labeled, or enzymatically labeled GLC 1 A
polypeptides, by immunoassay, or by chromatographic detection.
Typically, it will be desirable to immobilize either GLC 1 A or its binding protein to facilitate separation of complexes from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. Binding of GLC 1 A to an WO 98/20131 PCT/US97/20?02 _ upstream or downstream element, in the presence and absence of a candidate agent, can be accomplished in any vessel suitable for containing the reactants. Examples include microtitre plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein can be provided which adds a domain that allows the protein to be bound to a matrix. For example, glutathione-S-transferase/GLC 1 A (GST/GLC 1 AA) fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtitre plates, which are then combined with the cell lysates, e.g.
an 35S-labeled, and the test compound, and the mixture incubated under conditions conducive to complex formation, e.g. at physiological conditions for salt and pH, though 1 o slightly more stringent conditions may be desired. Following incubation, the beads are washed to remove any unbound label, and the matrix immobilized and radiolabel determined directly (e.g. beads placed in scintilant), or in the supernatant after the complexes are subsequently dissociated. Alternatively, the complexes can be dissociated from the matrix, separated by SDS-PAGE, and the level of GLC 1 A-binding protein found in the bead fraction quantitated from the gel using standard electrophoretic techniques such as described in the appended examples.
Other techniques for immobilizing proteins on matrices are also available for use in the subj ect assay. For instance, either GLC 1 A or its cognate binding protein can be 2o immobilized utilizing conjugation of biotin and streptavidin. For instance, biotinylated GLC 1 A molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
Alternatively, antibodies reactive with GLC 1 A but which do not interfere with binding of upstream or downstream elements can be derivatized to the wells of the plate, and GLC 1 A
trapped in the wells by antibody conjugation. As above, preparations of a GLC
I A-binding protein and a test compound are incubated in the GLC 1 A-presenting wells of the plate, and the amount of complex trapped in the well can be quantitated. Exemplary methods for detecting such complexes, in addition to those described above for the GST-immobilized 3o complexes, include immunodetection of complexes using antibodies reactive with the GLC 1 A binding element, or which are reactive with GLC 1 A protein and compete with the binding element; as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the binding element, either intrinsic or extrinsic activity. In the WO 98/20131 PCT/US97/20702 _ instance of the latter, the enzyme can be chemically conjugated or provided as a fusion protein with the GLC 1 A-BP. To illustrate, the GLC 1 A-BP can be chemically cross-linked or genetically fused with horseradish peroxidase, and the amount of polypeptide trapped in the complex can be assessed with a chromogenic substrate of the enzyme, e.g. 3,3'-diamino-benzadine terahydrochloride or 4-chloro-1-napthol. Likewise, a fusion protein comprising the polypeptide and glutathione-S-transferase can be provided, and complex formation quantitated by detecting the GST activity using 1-chloro-2,4-dinitrobenzene (Habig et al (1974) J Biol Chem 249:7130).
For processes which rely on immunodetection for quantitating one of the proteins trapped in the complex, antibodies against the protein, such as anti-antibodies, can be used. Alternatively, the protein to be detected in the complex can be "epitope tagged" in the form of a fusion protein which includes, in addition to the GLC 1 A
sequence, a second polypeptide for which antibodies are readily available (e.g. from ~ 5 commercial sources). For instance, the GST fusion proteins described above can also be used for quantification of binding using antibodies against the GST moiety.
Other useful epitope tags include myc-epitopes (e.g., see Ellison et al. ( 1991 ) J Biol Chem 266:21150-21157) which includes a 10-residue sequence from c-myc, as well as the pFLAG
system (International Biotechnologies, Inc.) or the pEZZ-protein A system (Pharamacia, NJ).
4.7.2. Cell based assay In addition to cell-free assays, such as described above, the readily available source of mutant and functional GLC 1 A nucleic acids and proteins provided by the present invention also facilitates the generation of cell-based assays for identifying small molecule agonists/antagonists and the like. For example, cells can be caused to overexpress a recombinant GLC 1 A protein in the presence and absence of a test agent of interest, with the assay scoring for modulation in GLC 1 A responses by the target cell mediated by the test agent. As with the cell-free assays, agents which produce a statistically significant change in GLC 1 A -dependent responses (either inhibition or potentiation) can be identified. In an 3o illustrative embodiment, the expression or activity of a GLC1A is modulated in cells and the effects of compounds of interest on the readout of interest (such as tissue differentiation, proliferation, tumorigenesis) are measured. For example, the expression of genes which are up- or down-regulated in response to a GLC 1 A-dependent signal cascade can be assayed.

WO 98/20131 PCT/US97/20702 _ ' _68_ In preferred embodiments, the regulatory regions of such genes, e.g., the 5' flanking promoter and enhancer regions, are operably linked to a detectable marker (such as luciferase) which encodes a gene product that can be readily detected.
Exemplary cells or cell lines may be derived from ocular tissue (e.g.
trabecular meshwork or ciliary body epithelia); as well as generic mammalian cell lines such as HeLa cells and COS cells, e.g., COS-7 (ATCC# CRL-1651 ). Further, the transgenic animals discussed herein may be used to generate cell lines containing one or more cell types involved in glaucoma, that can be used as cell culture models for this disorder. While 1 o primary cultures derived from the glaucomatous transgenic animals of the invention may be utilized, the generation of continuous cell lines is preferred. For examples of techniques which may be used to derive a continuous cell line from the transgenic animals, see Small et al., 1985, Mol. Cell Biol. 5:642-648.
1 S Using these cells, the effect of a test compound on a variety of end points can be tested including cell proliferation, migration, phagocytosis, adherence and/or biosynthesis (e.g. of extracellular matrix components). The cells can then be examined for phenotypes associated with glaucoma, including, but not limited to changes in cellular morphology, cell proliferation) cell migration, and cell adhesion.
In the event that the GLC 1 A proteins themselves, or in complexes with other proteins, are capable of binding DNA and modifying transcription of a gene, a transcriptional based assay could be used, for example, in which a GLC 1 A
responsive regulatory sequence is operably linked to a detectable marker gene.
Monitoring the influence of compounds on cells may be applied not only in basic drug screening, but also in clinical trials. In such clinical trials, the expression of a panel of genes may be used as a "read out" of a particular drug's therapeutic effect.
3a In yet another aspect of the invention, the subj ect GLC 1 A polypeptides can be used to generate a "two hybrid" assay (see, for example, U.S. Patent No.
5,283,317;
Zervos et al. ( 1993) Cell 72:223-232; Madura et al. ( 1993) J Biol Chem 268:12046-12054;
Bartel et al. (1993) Biotechniques 14:920-924; Iwabuchi et al. (1993) Oncogene 8:1693-1696; and Brent W094/10300), for isolating coding sequences for other cellular proteins which bind to or interact with GLC 1 A ("GLC 1 A-binding proteins" or "GLC 1 A-bp.
Briefly, the two hybrid assay relies on reconstituting in vivo a functional transcriptional activator protein from two separate fusion proteins. In particular, the method makes use of chimeric genes which express hybrid proteins. To illustrate, a first hybrid gene comprises the coding sequence for a DNA-binding domain of a transcriptional activator fused in frame to the coding sequence for a GLC 1 A polypeptide. The second hybrid protein encodes a transcriptional activation domain fused in frame to a sample gene 1 o from a cDNA library. If the bait and sample hybrid proteins are able to interact, e.g., form a GLC 1 A-dependent complex, they bring into close proximity the two domains of the transcriptional activator. This proximity is sufficient to cause transcription of a reporter gene which is operably linked to a transcriptional regulatory site responsive to the transcriptional activator, and expression of the reporter gene can be detected and used to I 5 score for the interaction of the GLC 1 A and sample proteins.
4_.8 Transgenic animals 4.8.1. Animal-based s, stems 2o Another aspect of the present invention concerns transgenic animals which are comprised of cells (of that animal) which contain a transgene of the present invention and which preferably (though optionally) express an exogenous GLC 1 A protein in one or more cells in the animal. A GLC 1 A transgene can encode the wild-type form of the protein, or can encode homologs thereof, including both agonists and antagonists, as well as 25 antisense constructs. In preferred embodiments, the expression of the transgene is restricted to specific subsets of cells, tissues or developmental stages utilizing, for example, cis-acting sequences that control expression in the desired pattern. In the present invention, such mosaic expression of a GLC 1 A protein can be essential for many forms of lineage analysis and can additionally provide a means to assess the effects of, for example, lack of GLC 1 A
3o expression which might grossly alter development in small patches of tissue within an otherwise normal embryo. Toward this and, tissue-specific regulatory sequences and conditional regulatory sequences can be used to control expression of the transgene in certain spatial patterns. Moreover, temporal patterns of expression can be provided by, for WO 98/20131 PCT/US97/20702 ' example, conditional recombination systems or prokaryotic transcriptional regulatory sequences.
Genetic techniques, which allow for the expression of transgenes can be regulated via site-specific genetic manipulation in vivo, are known to those skilled in the art.
For instance, genetic systems are available which allow for the regulated expression of a recombinase that catalyzes the genetic recombination of a target sequence. As used herein, the phrase "target sequence" refers to a nucleotide sequence that is genetically recombined by a recombinase. The target sequence is flanked by recombinase recognition sequences 1 o and is generally either excised or inverted in cells expressing recombinase activity.
Recombinase catalyzed recombination events can be designed such that recombination of the target sequence results in either the activation or repression of expression of one of the subj ect GLC 1 A proteins. For example, excision of a target sequence which interferes with the expression of a recombinant GLC 1 A gene, such as one which encodes an antagonistic homolog or an antisense transcript, can be designed to activate expression of that gene. This interference with expression of the protein can result from a variety of mechanisms, such as spatial separation of the GLC 1 A gene from the promoter element or an internal stop codon. Moreover, the transgene can be made wherein the coding sequence of the gene is flanked by recombinase recognition sequences and is initially transfected into cells in a 3' 2o to 5' orientation with respect to the promoter element. In such an instance, inversion of the target sequence will reorient the subj ect gene by placing the 5' end of the coding sequence in an orientation with respect to the promoter element which allow for promoter driven transcriptional activation.
The transgenic animals of the present invention all include within a plurality of their cells a transgene of the present invention, which transgene alters the phenotype of the "host cell" with respect to regulation of cell growth, death and/or differentiation. Since it is possible to produce transgenic organisms of the invention utilizing one or more of the transgene constructs described herein, a general description will be given of the production of transgenic organisms by referring generally to exogenous genetic material.
This general description can be adapted by those skilled in the art in order to incorporate specific transgene sequences into organisms utilizing the methods and materials described below.

WO 98/20131 PCT/US97/20702 _ ' _71_ .
In an illustrative embodiment, either the crelloxP recombinase system of bacteriophage P 1 (Lakso et al. ( 1992) PNAS 89:6232-6236; Orban et al. ( 1992) PNAS
89:6861-6865) or the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al. (1991 ) Science 251:1351-1355; PCT publication WO 92/15694) can be used to generate s in vivo site-specific genetic recombination systems. Cre recombinase catalyzes the site-specific recombination of an intervening target sequence located between loxP
sequences. loxP sequences are 34 base pair nucleotide repeat sequences to which the Cre recombinase binds and are required for Cre recombinase mediated genetic recombination.
The orientation of loxP sequences determines whether the intervening target sequence is 1 o excised or inverted when Cre recombinase is present (Abremski et al. ( 1984) J. Biol. Chem.
259:1509-1514); catalyzing the excision of the target sequence when the loxP
sequences are oriented as direct repeats and catalyzes inversion of the target sequence when loxP
sequences are oriented as inverted repeats.
15 Accordingly, genetic recombination of the target sequence is dependent on expression of the Cre recombinase. Expression of the recombinase can be regulated by promoter elements which are subject to regulatory control, e.g., tissue-specific, developmental stage-specific, inducible or repressible by externally added agents. This regulated control will result in genetic recombination of the target sequence only in cells 2o where recombinase expression is mediated by the promoter element. Thus, the activation expression of a recombinant GLC 1 A protein can be regulated via control of recombinase expression.
Use of the crelloxP recombinase system to regulate expression of a 25 recombinant GLC 1 A protein requires the construction of a transgenic animal containing transgenes encoding both the Cre recombinase and the subject protein. Animals containing both the Cre recombinase and a recombinant GLC 1 A gene can be provided through the construction of "double" transgenic animals. A convenient method for providing such animals is to mate two transgenic animals each containing a transgene, e.g., a GLC 1 A gene 3o and recombinase gene.
One advantage derived from initially constructing transgenic animals containing a GLC 1 A transgene in a recombinase-mediated expressible format derives from WO 98/20131 PCTlUS97/20702 _ -77_ the likelihood that the subject protein, whether agonistic or antagonistic, can be deleterious upon expression in the transgenic animal. In such an instance, a founder population, in which the subject transgene is silent in all tissues, can be propagated and maintained.
Individuals of this founder population can be crossed with animals expressing the recombinase in, for example, one or more tissues and/or a desired temporal pattern. Thus, the creation of a founder population in which, for example, an antagonistic GLC I A
transgene is silent will allow the study of progeny from that founder in which disruption of GLC 1 A mediated induction in a particular tissue or at certain developmental stages would result in, for example, a lethal phenotype.
to Similar conditional transgenes can be provided using prokaryotic promoter sequences which require prokaryotic proteins to be simultaneous expressed in order to facilitate expression of the GLC 1 A transgene. Exemplary promoters and the corresponding trans-activating prokaryotic proteins are given in U.S. Patent No. 4,833,080.
Moreover, expression of the conditional transgenes can be induced by gene therapy-like methods wherein a gene encoding the trans-activating protein, e.g. a recombinase or a prokaryotic protein, is delivered to the tissue and caused to be expressed, such as in a cell-type specific manner. By this method, a GLC 1 AA transgene could remain silent into adulthood until "turned on" by the introduction of the trans-activator.
In an exemplary embodiment, the "transgenic non-human animals" of the invention are produced by introducing transgenes into the germline of the non-human animal. Embryonal target cells at various developmental stages can be used to introduce transgenes. Different methods are used depending on the stage of development of the embryonal target cell. The specific lines) of any animal used to practice this invention are selected for general good health, good embryo yields, good pronuclear visibility in the embryo, and good reproductive fitness. In addition, the haplotype is a significant factor. For example, when transgenic mice are to be produced, strains such as C57BL/6 or FVB lines 3o are often used (Jackson Laboratory, Bar Harbor, ME). Preferred strains are those with H-2b, H-2d or H-2q haplotypes such as C57BL/6 or DBA/l. The Iine(s) used to practice this invention may themselves be transgenics, and/or may be knockouts (i.e., obtained from animals which have one or more genes partially or completely suppressed) .

WO 98/20131 PCT/US97I20702 _ ' -73- .
In one embodiment, the transgene construct is introduced into a single stage embryo. The zygote is the best target for micro-injection. In the mouse, the male pronucleus reaches the size of approximately 20 micrometers in diameter which allows reproducible injection of 1-2pl of DNA solution. The use of zygotes as a target for gene s transfer has a major advantage in that in most cases the injected DNA will be incorporated into the host gene before the first cleavage (Brinster et al. (1985) PNAS
82:4438-4442). As a consequence, all cells of the transgenic animal will carry the incorporated transgene. This will in general also be reflected in the efficient transmission of the transgene to offspring of the founder since 50% of the germ cells will harbor the transgene.
to Normally, fertilized embryos are incubated in suitable media until the pronuclei appear. At about this time, the nucleotide sequence comprising the transgene is introduced into the female or male pronucleus as described below. In some species such as mice, the male pronucleus is preferred. It is most preferred that the exogenous genetic ~ s material be added to the male DNA complement of the zygote prior to its being processed by the ovum nucleus or the zygote female pronucleus. It is thought that the ovum nucleus or female pronucleus release molecules which affect the male DNA complement, perhaps by replacing the protamines of the male DNA with histones, thereby facilitating the combination of the female and male DNA complements to form the diploid zygote.
Thus, it is preferred that the exogenous genetic material be added to the male complement of DNA or any other complement of DNA prior to its being affected by the female pronucleus. For example, the exogenous genetic material is added to the early male pronucleus, as soon as possible after the formation of the male pronucleus, which is when 2s the male and female pronuclei are well separated and both are located close to the cell membrane. Alternatively, the exogenous genetic material could be added to the nucleus of the sperm after it has been induced to undergo decondensation. Sperm containing the exogenous genetic material can then be added to the ovum or the decondensed sperm could be added to the ovum with the transgene constructs being added as soon as possible 3o thereafter.
Introduction of the transgene nucleotide sequence into the embryo may be accomplished by any means known in the art such as, for example, microinjection, WO 98/20131 PCT/US97/20702 ' electroporation, or lipofection. Following introduction of the transgene nucleotide sequence into the embryo, the embryo may be incubated in oitro for varying amounts of time, or reimplanted into the surrogate host, or both. In vitro incubation to maturity is within the scope of this invention. One common method in to incubate the embryos in vitro for about 1-7 days, depending on the species, and then reimplant them into the surrogate host.
For the purposes of this invention a zygote is essentially the formation of a diploid cell which is capable of developing into a complete organism.
Generally, the zygote will be comprised of an egg containing a nucleus formed, either naturally or artif cially, by 1 o the fusion of two haploid nuclei from a gamete or gametes. Thus, the gamete nuclei must be ones which are naturally compatible, i.e., ones which result in a viable zygote capable of undergoing differentiation and developing into a functioning organism.
Generally, a euploid zygote is preferred. If an aneuploid zygote is obtained, then the number of chromosomes should not vary by more than one with respect to the euploid number of the organism from which either gamete originated.
In addition to similar biological considerations, physical ones also govern the amount (e.g., volume) of exogenous genetic material which can be added to the nucleus of the zygote or to the genetic material which forms a part of the zygote nucleus. If no 2o genetic material is removed, then the amount of exogenous genetic material which can be added is limited by the amount which will be absorbed without being physically disruptive.
Generally, the volume of exogenous genetic material inserted will not exceed about 10 picoliters. The physical effects of addition must not be so great as to physically destroy the viability of the zygote. The biological limit of the number and variety of DNA
sequences will vary depending upon the particular zygote and functions of the exogenous genetic material and will be readily apparent to one skilled in the art, because the genetic material, including the exogenous genetic material, of the resulting zygote must be biologically capable of initiating and maintaining the differentiation and development of the zygote into a functional organism.
The number of copies of the transgene constructs which are added to the zygote is dependent upon the total amount of exogenous genetic material added and will be the amount which enables the genetic transformation to occur. Theoretically only one copy is required; however, generally, numerous copies are utilized, for example, 1,000-20,000 WO 98/20131 PCT/US97/20702 ' copies of the transgene construct, in order to insure that one copy is functional. As regards the present invention, there will often be an advantage to having more than one functioning copy of each of the inserted exogenous DNA sequences to enhance the phenotypic expression of the exogenous DNA sequences.
s Any technique which allows for the addition of the exogenous genetic material into nucleic genetic material can be utilized so long as it is not destructive to the cell, nuclear membrane or other existing cellular or genetic structures. The exogenous genetic material is preferentially inserted into the nucleic genetic material by microinjection.
Io Microinjection of cells and cellular structures is known and is used in the art.
Reimplantation is accomplished using standard methods. Usually, the surrogate host is anesthetized, and the embryos are inserted into the oviduct.
The number of embryos implanted into a particular host will vary by species, but will usually be 15 comparable to the number of off spring the species naturally produces.
Transgenic offspring of the surrogate host may be screened for the presence and/or expression of the transgene by any suitable method. Screening is often accomplished by Southern blot or Northern blot analysis, using a probe that is complementary to at least 2o a portion of the transgene. Western blot analysis using an antibody against the protein encoded by the transgene may be employed as an alternative or additional method for screening for the presence of the transgene product. Typically, DNA is prepared from tail tissue and analyzed by Southern analysis or PCR for the transgene.
Alternatively, the tissues or cells believed to express the transgene at the highest levels are tested for the presence and 2s expression of the transgene using Southern analysis or PCR, although any tissues or cell types may be used for this analysis.
Alternative or additional methods for evaluating the presence of the transgene include, without limitation, suitable biochemical assays such as enzyme and/or 3o immunological assays, histological stains for particular marker or enzyme activities, flow cytometric analysis, and the like. Analysis of the blood may also be useful to detect the presence of the transgene product in the blood, as well as to evaluate the effect of the transgene on the levels of various types of blood cells and other blood constituents.

WO 98/20131 PCT/US97t20702 _ _76_ Progeny of the transgenic animals may be obtained by mating the transgenic animal with a suitable partner, or by in vitro fertilization of eggs and/or sperm obtained from the transgenic animal. Where mating with a partner is to be performed, the partner may or may not be transgenic and/or a knockout; where it is transgenic, it may contain the same or a different transgene, or both. Alternatively, the partner may be a parental line. Where in vitro fertilization is used, the fertilized embryo may be implanted into a surrogate host or incubated in vitro, or both. Using either method, the progeny may be evaluated for the presence of the transgene using methods described above, or other appropriate methods.
The transgenic animals produced in accordance with the present invention will include exogenous genetic material. As set out above, the exogenous genetic material will, in certain embodiments, be a DNA sequence which results in the production of a GLC 1 A protein (either agonistic or antagonistic), and antisense transcript, or a GLC 1 A
mutant. Further, in such embodiments the sequence will be attached to a transcriptional control element, e.g., a promoter, which preferably allows the expression of the transgene product in a specific type of cell.
Retroviral infection can also be used to introduce transgene into a non-human animal. The developing non-human embryo can be cultured in vitro to the blastocyst stage.
During this time, the blastomeres can be targets for retroviral infection (Jaenich, R. ( 1976) PNAS 73:1260-1264). Efficient infection of the blastomeres is obtained by enzymatic treatment to remove the zona pellucida (Manipulating the Mouse Embryo, Hogan eds. (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1986). The viral vector system used to introduce the transgene is typically a replication-defective retrovirus carrying the transgene (Jahner et al. ( 1985) PNAS 82:6927-6931; Van der Putten et al. ( 1985) PNAS
82:6148-6152). Transfection is easily and efficiently obtained by culturing the blastomeres on a monolayer of virus-producing cells (Van der Putten, supra; Stewart et al.
( 1987) EMBO J. 6:383-388). Alternatively, infection can be performed at a later stage. Virus or 3o virus-producing cells can be injected into the blastocoele (Jahner et al. ( 1982) Nature 298:623-628). Most of the founders will be mosaic for the transgene since incorporation occurs only in a subset of the cells which formed the transgenic non-human animal. Further, the founder may contain various retroviral insertions of the transgene at different positions in the genome which generally will segregate in the offspring. In addition, it is also possible -77- .
to introduce transgenes into the germ line by intrauterine retroviral infection of the midgestation embryo (Jahner et al. ( 1982) supra).
A third type of target cell for transgene introduction is the embryonal stem cell (ES). ES cells are obtained from pre-implantation embryos cultured in vitro and fused with embryos (Evans et al. ( 1981 ) Nature 292:154-156; Bradley et al. ( 1984) Nature 309:255-258; Gossler et al. (1986) PNAS 83: 9065-9069; and Robertson et al.
(1986) Nature 322:445-448). Transgenes can be efficiently introduced into the ES cells by DNA
transfection or by retrovirus-mediated transduction. Such transformed ES cells can thereafter be combined with blastocysts from a non-human animal. The ES cells thereafter colonize the embryo and contribute to the germ line of the resulting chimeric animal. For review see Jaenisch, R. (1988) Science 240:1468-1474.
In one embodiment, gene targeting, which is a method of using homologous recombination to modify an animal's genome, can be used to introduce changes into cultured embryonic stem cells. By targeting a GLC 1 A gene of interest in ES
cells, these changes can be introduced into the germlines of animals to generate chimeras.
The gene targeting procedure is accomplished by introducing into tissue culture cells a DNA targeting construct that includes a segment homologous to a target GLC 1 A locus, and which also includes an intended sequence modification to the GLC 1 A genomic sequence (e.g., insertion, deletion, point mutation). The treated cells are then screened for accurate targeting to identify and isolate those which have been properly targeted.
Gene targeting in embryonic stem cells is in fact a scheme contemplated by the present invention as a means for disrupting a GLC 1 A gene function through the use of a targeting transgene construct designed to undergo homologous recombination with one or more GLC 1 A genomic sequences. The targeting construct can be arranged so that, upon recombination with an element of a GLC 1 A gene, a positive selection marker is inserted into (or replaces) coding sequences of the gene. The inserted sequence functionally disrupts 3o the GLC 1 A gene, while also providing a positive selection trait.
Exemplary GLC 1 A
targeting constructs are described in more detail below.

WO 98/20131 PCT/I1S97/20702 _ ' _78_ Generally, the embryonic stem cells (ES cells ) used to produce the knockout animals will be of the same species as the knockout animal to be generated.
Thus for example, mouse embryonic stem cells will usually be used for generation of knockout mice.
Embryonic stem cells are generated and maintained using methods well known to the skilled artisan such as those described by Doetschman et al. ( 1985) J.
Embryol. Exp. MoGLCIAhoI. 87:27-45). Any line of ES cells can be used, however, the line chosen is typically selected for the ability of the cells to integrate into and become part of the germ line of a developing embryo so as to create germ line transmission of the knockout l o construct. Thus, any ES cell line that is believed to have this capability is suitable for use herein. One mouse strain that is typically used for production of ES cells, is the 129J strain.
Another ES cell line is murine cell line D3 (American Type Culture Collection, catalog no.
CKL 1934) Still another preferred ES cell line is the WW6 cell line (Ioffe et al. (1995) PNAS 92:7357-7361 ). The cells are cultured and prepared for knockout construct insertion using methods well known to the skilled artisan, such as those set forth by Robertson in:
Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E.J.
Robertson, ed.
IRL Press, Washington, D.C. [ 1987]); by Bradley et al. ( 1986) Current Topics in Devel.
Biol. 20:357-371 ); and by Hogan et al. (Manipulating the Mouse Embryo: A
Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY [ 1986]) .
Insertion of the knockout construct into the ES cells can be accomplished using a variety of methods well known in the art including for example, electroporation, microinjection, and calcium phosphate treatment. A preferred method of insertion is electroporation .
Each knockout construct to be inserted into the cell must first be in the linear form. Therefore, if the knockout construct has been inserted into a vector (described infra), linearization is accomplished by digesting the DNA with a suitable restriction endonuclease selected to cut only within the vector sequence and not within the knockout construct sequence.
For insertion, the knockout construct is added to the ES cells under appropriate conditions for the insertion method chosen, as is known to the skilled artisan.

WO 98/20131 PCT/US97/20702 _ -79- .
Where more than one construct is to be introduced into the ES cell, each knockout construct can be introduced simultaneously or one at a time.
If the ES cells are to be electroporated, the ES cells and knockout construct DNA are exposed to an electric pulse using an electroporation machine and following the manufacturer's guidelines for use. After electroporation, the ES cells are typically allowed to recover under suitable incubation conditions. The cells are then screened for the presence of the knockout construct .
to Screening can be accomplished using a variety of methods. Where the marker gene is an antibiotic resistance gene, for example, the ES cells may be cultured in the presence of an otherwise lethal concentration of antibiotic. Those ES
cells that survive have presumably integrated the knockout construct. If the marker gene is other than an antibiotic resistance gene, a Southern blot of the ES cell genomic DNA can be probed with a sequence of DNA designed to hybridize only to the marker sequence Alternatively, PCR
can be used. Finally, if the marker gene is a gene that encodes an enzyme whose activity can be detected (e.g., b-galactosidase), the enzyme substrate can be added to the cells under suitable conditions, and the enzymatic activity can be analyzed. One skilled in the art will be familiar with other useful markers and the means for detecting their presence in a given 2o cell. All such markers are contemplated as being included within the scope of the teaching of this invention.
The knockout construct may integrate into several locations in the ES cell genome, and may integrate into a different location in each ES cell's genome due to the occurrence of random insertion events. The desired location of insertion is in a complementary position to the DNA sequence to be knocked out, e.g., the GLC 1 A coding sequence, transcriptional regulatory sequence, etc. Typically, less than about 1-5 % of the ES cells that take up the knockout construct will actually integrate the knockout construct in the desired location. To identify those ES cells with proper integration of the knockout 3 o construct, total DNA can be extracted from the ES cells using standard methods. The DNA
can then be probed on a Southern blot with a probe or probes designed to hybridize in a specific pattern to genomic DNA digested with particular restriction enzyme(s).
Alternatively, or additionally, the genomic DNA can be amplified by PCR with probes specifically designed to amplify DNA fragments of a particular size and sequence (i.e., only WO 98/20131 PCT/US97/20702 _ ' those cells containing the knockout construct in the proper position will generate DNA
fragments of the proper size).
After suitable ES cells containing the knockout construct in the proper location have been identified, the cells can be inserted into an embryo.
Insertion may be accomplished in a variety of ways known to the skilled artisan, however a preferred method is by microinjection. For microinjection, about 10-30 cells are collected into a micropipet and injected into embryos that are at the proper stage of development to permit integration of the foreign ES cell containing the knockout construct into the developing embryo. For instance, as the appended Examples describe, the transformed ES cells can be microinjected into blastocytes.
The suitable stage of development for the embryo used for insertion of ES
cells is very species dependent, however for mice it is about 3.5 days. The embryos are obtained by perfusing the uterus of pregnant females. Suitable methods for accomplishing this are known to the skilled artisan, and are set forth by, e.g., Bradley et al. (supra).
While any embryo of the right stage of development is suitable for use, preferred embryos are male. In mice, the preferred embryos also have genes coding for a coat color that is different from the coat color encoded by the ES cell genes.
In this way, the offspring can be screened easily for the presence of the knockout construct by looking for mosaic coat color (indicating that the ES cell was incorporated into the developing embryo).
Thus, for example, if the ES cell line carries the genes for white fur, the embryo selected will carry genes for black or brown fur.
After the ES cell has been introduced into the embryo, the embryo may be implanted into the uterus of a pseudopregnant foster mother for gestation.
While any foster mother may be used, the foster mother is typically selected for her ability to breed and reproduce well, and for her ability to care for the young. Such foster mothers are typically 3o prepared by mating with vasectomized males of the same species. The stage of the pseudopregnant foster mother is important for successful implantation, and it is species dependent. For mice, this stage is about 2-3 days pseudopregnant.

WO 98/20131 PCT/US97/20702 _ ' Offspring that are born to the foster mother may be screened initially for mosaic coat color where the coat color selection strategy (as described above, and in the appended examples) has been employed. In addition, or as an alternative, DNA
from tail tissue of the offspring may be screened for the presence of the knockout construct using Southern blots and/or PCR as described above. Offspring that appear to be mosaics may then be crossed to each other, if they are believed to carry the knockout construct in their germ line, in order to generate homozygous knockout animals. Homozygotes may be identified by Southern blotting of equivalent amounts of genomic DNA from mice that are the product of this cross, as well as mice that are known heterozygotes and wild type mice.
to Other means of identifying and characterizing the knockout offspring are available. For example, Northern blots can be used to probe the mRNA for the presence or absence of transcripts encoding either the gene knocked out, the marker gene, or both. In addition, Western blots can be used to assess the level of expression of the GLC 1 A gene l 5 knocked out in various tissues of the offspring by probing the Western blot with an antibody against the particular GLC 1 A protein, or an antibody against the marker gene product, where this gene is expressed. Finally, in situ analysis (such as fixing the cells and labeling with antibody) and/or FRCS (fluorescence activated cell sorting) analysis of various cells from the offspring can be conducted using suitable antibodies to look for the presence or 2o absence of the knockout construct gene product.
Yet other methods of making knock-out or disruption transgenic animals are also generally known. See, for example, Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986). Recombinase dependent 25 knockouts can also be generated, e.g. by homologous recombination to insert target sequences, such that tissue specific and/or temporal control of inactivation of a GLClA-gene can be controlled by recombinase sequences (described infra).
Animals containing more than one knockout construct and/or more than one 3o transgene expression construct are prepared in any of several ways. The preferred manner of preparation is to generate a series of mammals, each containing one of the desired transgenic phenotypes. Such animals are bred together through a series of crosses, backcrosses and selections, to ultimately generate a single animal containing all desired knockout constructs and/or expression constructs, where the animal is otherwise congenic WO 98/20131 PCT/US97/20702 _ ' _g7_ (genetically identical) to the wild type except for the presence of the knockout constructs) and/or transgene(s) .
The present invention is further illustrated by the following examples which should not be construed as limiting in any way. The contents of all cited references (including literature references, issued patents, published patent applications as cited throughout this application are hereby expressly incorporated by reference.The practice of the present invention will employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant 1 o DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. See, for example. Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press:
1989); DNA
Cloning, Volumes I and II (D. N. Glover ed., 1985); Oligonucleotide Synthesis (M. J. Gait ed., 1984); Mullis et al. U.S. Patent No: 4,683,195; Nucleic Acid Hybridization (B. D.
i s Hames & S. J. Higgins eds. 1984); Transcription And Translation (B. D.
Hames & S. J.
Higgins eds. 1984); Culture Of Animal Cells (R. I. Freshney, Alan R. Liss, Inc., 1987);
Immobilized Cells And Enzymes (IRL Press, 1986); B. Perbal, A Practical Guide To Molecular Cloning ( 1984); the treatise, Methods In Enzymology (Academic Press, Inc., N.Y.); Gene Transfer Vectors For Mammalian Cells (J. H. Miller and M. P. Calos eds., 20 1987, Cold Spring Harbor Laboratory); Methods In Enzymology, Vols. 154 and 155 (Wu et al. eds.), Immunochemical Methods In Cell And Molecular Biology (Mayer and Walker, eds., Academic Press, London, 1987); Handbook Of Experimental Immunology, Volumes I-IV (D. M. Weir and C. C. Blackwell, eds., 1986); Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986).
The present invention is further illustrated by the following examples which should not be construed as limiting in any way. The contents of all cited references (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby 3o expressly incorporated by reference.

WO 98/20131 PCT/US97/20702 ' 5. 1 Genetic Linkaee of Familial Open Angle Glaucoma to Chromosome 1 X21-a31 Materials and Methods Pedigree A family in which five consecutive generations have been affected with juvenile-onset, open-angle glaucoma without iridocorneal angle abnormalities was identified. The family comprised descendants of a woman who emigrated from t o Germany to the midwestern United States in the late 1800s. The disease state in affected family members included onset during the first 3 decades of life, normal anterior chamber angles, high intraocular pressures, lack of systemic or other ocular abnormalities, and need for surgery to control the glaucoma in affected individuals. A
total of 35 family members at 50% risk for glaucoma had complete eye examinations including visual acuity with refraction, slit-lamp biomicroscopy, applanation tomometry, gonioscopy, stereo disc photography and Humphrey, Goldmann or Octopus perimetry.
Two other affected patients were ascertained by reviewing records of other ophthalmologists. Patients were considered to be affected for linkage if they had documented pressures greater than 30 mm Hg and evidence of optic nerve or visual field 2o damage; or, if they had intraocular pressures greater than 22 mm Hg and an obviously affected child. Affected family members are characterized by an early age of diagnosis, a normal appearing trabecular meshwork, very high intraocular pressures (often above 50 mm Hg), and relatively pressure-resistant optic nerves.
DNA typing Blood samples were obtained from all living affected family members as well as six spouses of affected patients with children. 1 Oml blood were obtained from each patient in EDTA-containing glass tubes. DNA was prepared from the blood using a non-organic extraction procedure (Grimberg, J, et al. Nucl. Acids Res 17, 8390 (1989)).
3o Short tandem repeat polymorphisms (STRPs) distributed across the entire autosomal genome were selected from the literature or from those kindly provided by J.L.
Weber.
The majority were [dC-dAJ-[dG-dTJ dinucleotide repeats. Oligonucleotide primers flanking each STRP were synthesized using standard phosphoramidite chemistry (Applied Biosystems model 391 DNA synthesizer). Amplification of each STRP was WO 98/20131 PCT/US97/20702 _ _84_ performed with 50 ng. of each patient's DNA in a 8.35 1 PCR containing each of the following: 1.25 1 10 X buffer ( 1 OOmM Tris-HCl pH 8.8, 500 mM KC1, 15 mM
MgCl2, 0.01 % w/v gelatin), 300 M each of dCTP, dGTP and dTTP, 37M dATP, SOpmoles each primer, 0.25 1 -35 S-dATP (Amersham,> 1000 Ci mmol-1 ), and 0.25 U Taq polymerase (Perkin-Elmer/Cetus). Samples were incubated in a DNA thermocycler (Perkin-Elmer/Cetus) for 35 cycles under the following conditions: 94C for 30 s, SSC
for 30 s, and 72C for 30 s. Following amplification, 51 of stop solution (95% formamide, IOmM
NaOH, 0.05% Bromophenol Blue, 0.05% Xylene Cyanol) was added to each sample.
Following denaturation for 3 min at 95C, 5 I of each sample was immediately loaded onto prewarmed polyacrylamide gels (6% polyacrylamide, 7 M urea) and electrophoresed for 3-4h. Gels were then placed on Whatman, 3mm paper and dried in a slab gel dryer. Autoradiographs were created by exposing Kodak Xomat AR film to the dried gels for 24-36h.
Linkage analysis Genotypic data from the autoradiographs were entered into a Macintosh computer. A Hypercard-based program (Nichols, BE et al., Am J Hum Genet 51 (1992)) was used to store and retrieve marker data as well as to export it to a DOS-compatible machine for analysis with the computer program LINKAGE (version 5.1 ) (Lathrop, GM and LaLouel, JM 3 59, 794-801 ( 1992)). Allele frequencies were assumed to be equal for each marker. The MLINK routine was used for pairwise analysis.
The relative odds of all possible orders of the disease and two markers (D 1 S 191 and D 1 S 194) was performed under the ILINK program. Significance of linkage was evaluated using the standard criterion (Zmax>3.0).
Results clinical findings All of the 37 family members studied were at 50% risk of having the disease because of a known affected parent or sibling. Nineteen of these patients had 3o elevated intraocular pressures and visual field defects consistent with the diagnosis of primary open angle glaucoma. Three more patients had moderately elevated intraocular pressures and obviously affected children.
linkage analysis WO 98/20131 PCT/US97/20702 _ _85_ , _ Over 90 short tandem repeat polymorphisms were typed in the family before linkage was detected with markers that map to the long arm of chromosome 1.
Two-point maximum likelihood calculations using all available family members and 33 chromosome 1 markers revealed significant linkage to eight of them (Table 2).

was fully informative for all affected members of the family, and pairwise linkage analysis produced a lod score of 6.5 ( = 0). Multipoint linkage analysis did not add to the peak lod score. The glaucoma locus was therefore determined to be located in a region of about 20 centiMorgans (cM) in size between D 1 S 19 I and D 1 S 194.
Both of these markers demonstrated multiple recombinants (two and three, respectively) in 1 o affected individuals in the family. The order D 1 S 191-glaucoma-D 1 S 194 was more than 1,000 times more likely than the other two possible orders.
Table Pairwise linkage data Recombination Fraction 0.15 0.20 0.25 0.30 0.40 Zma Locu g x s D1S212 6.0 5.4 4.8 4.2 3.6 2.9 1.4 6.5 0.00 1 D1S215 5.1 4.6 4.0 3.5 2.9 2.3 1.0 5.6 0.00 1 D1S218 4.7 4.3 3.8 3.3 2.7 2.2 1.0 5.2 0.00 1 D1S238 4.4 4.2 3.9 3.4 2.9 2.4 1.2 4.4 0.04 I

2o DIS117 3.8 3.6 3.3 2.8 2.3 1.8 0.7 3.8 0.04 lq D1S104 3.2 2.9 2.6 2.3 2.0 1.6 0.7 3.4 0.00 1q21-q23 DIS191 3.0 3.2 3.0 2.7 2.4 1.9 0.9 3.2 0.09 I

DIS196 2.9 2.6 2.3 2.0 1.6 1.3 0.5 3.1 0.00 1 Table 2 Pairwise linkage data Recombination ftaction 0.05 0.19 WO 98/20131 PCT/US97/20702 _ ' 5.2 Genetic Fine Manning of the Juvenile Primar~pen Angle Glaucoma Locus and Identification and Characterization of a Glaucoma Gene Once primary linkage has been identified, the next step in identifying any disease gene by positional cloning is the narrowing of the candidate locus to the smallest possible genetic region. The initial study described in Example 5.1 demonstrated that a primary open angle glaucoma gene lies within an approximately 20 cM region flanked by markers D 1 S 194 and D 1 S 191 on chromosome 1 q. Additional markers and families were obtained and used to refine the genetic locus to a 2.5 cM region using two of these t 0 families. The third family should allow the interval to be further narrowed.
In addition to the family resources, polymorphic DNA markers and genetic maps were used to refine the 1 q glaucoma locus. Using STRPs, the genotype of each family member was determined. Amplification of each STRP was performed using ~ 5 the following protocol:
1 ) Dilute genomic DNA (about 1 g/1) 1 /50 i.e. 201 "stock" DNA and 980 dd H20.
2) Use 2.51 of "dilute" DNA as template for PCR
20 3) Prepare PCR reaction mix as follows:
1.251 10 X Buffer (Stratagene) 0.121 of each primer (50pmoles each primer) 0.51 dNTPs (5mM C,T,&G and 0.625 mM A "cold") 3.51 dd H20 25 0.251 3 5 S-dATP
0.11 Taq polymerise oil (one drop) 4) Perform PCR at optimal conditions for given primers (usually 94 30 s, 30 55 30s and 72 30 s) and run for 35 cycles.
5) Add 51 stop solution (95% formamide, IOmM NaOH, 0.05%
bromophenol blue, 0.05% xylene cyanol) to each tube.

WO 98/20131 PCT/US97/20702 _ ' _87_ , 6) Denature samples at 95C for 3 minutes and load immediately onto a prewarmed polyacrylamide gel.
7) Dry gels on Whatmann paper and expose autoradiography film for 1-2 s days.
Where possible, multiple loadings of different STRPs on gels were performed. Up to 6 markers per gel have been successfully loaded. In addition, the PCR
1 o amplification (up to three markers) have been successfully multiplexed.
The juvenile glaucoma gene is believed to lie between markers AFM238 and AT3 (an 8 centimorgan interval) based on observed recombinations within the families studied.
Haplotypic analysis between families has further narrowed this interval to the 2 centimorgan interval between D 15210 and AT3.
is Since the genetic interval has been narrowed significantly physical mapping strategies can be used. The closest flanking markers to screen total human genomic yeast artificial chromosome (YAC) libraries to identify YACs mapping to the region of interest. The CEPH and CEPH mega-YAC libraries can be used for this 2o purpose (available from the Centre d'Etude du Polymorphisme Humain (CEPH) Paris, France). Forty-four percent of the clones in the CEPH mega-YAC library have an average size of 560 kb, an additional 21% have an average size of 800 kb, and 35% have an average size of 120 kb. This library is available in a gridded micro-titer plate format such that only 50-200 PCR reactions need to be performed using a specific sequence 25 tagged site (STS) to identify a unique YAC containing the STS. The YAC
contigs identified by CEPH have been used to begin constructing a contig across the 1 q candidate region (see Figure 3). YAC contigs using YAC ends can be constructed to identify additional YACs. YAC ends can be rescued using anchored PCR (Riley, J. et al ( 1990) Nucleic Acids Res 18:2887-2890), the ends can then be sequenced and the 3o sequence can be used to develop a sequence tagged site (STS). The STS can be used to rescreen the YAC library to obtain an overlapping adjacent YAC.
Because some YACs have been shown to be chimeric or to contain deletions or rearrangements, particularly those from the mega YAC library, the correctness of each YAC contig should be verified by constructing a pulse field map of the region. In addition, chimeric YACs are minimized by ensuring that the YAC
maps to a single chromosome by fluorescent in situ hybridization (FISH) or that the two YAC
ends map to the same chromosome using monochromosomal somatic cell hybrids (NIGMs Panel 2). In addition, the YAC chimera problem can be minimized by not relying on any single YAC to span a given chromosome segment, but rather by obtaining at least two overlapping independent YACs to ensure coverage of a given region.
Once a YAC contig spanning the candidate region has been isolated, this reagent can be used to generate additional genetic markers for potentially finer genetic mapping. In addition, the YACs can be used to make higher resolution physical mapping reagents such as region specific lambda and cosmid clones. Lambda and cosmid clones can be used for isolation of candidate genes. A modification of "exon trapping" (Duyk, G.M. ( I 990) Proc Natl Acad Sci USA 87:8995-8999) known as exon amplification (Buckler, A.J. ( I 991 ) Proc Natl Acad Sci USA 88:4005-4009) can be used to identify exons from genes within the region. Exons trapped from the candidate region can be used as probes to screen eye cDNA libraries to isolate eDNAs. Where necessary, other strategies can be utilized to identify genes in genomic DNA including screening 2o cDNA libraries with YAC fragments subcloned into cosmids, zoo blot analysis, coincidence cloning strategies such as direct selection of cDNAs with biotin-streptavidin tagged cosmid clones (Morgan, J.G. et al (1992) Nucleic Acid Res 20 (19):5173-5179), and HTF island analysis (Bird, A.P. ( 1987) Trends Genet 3:342-247). Promising genes will be further evaluated by searching for mutations using GC-clamped denaturing gradient gel electrophoresis (Sheffield, V.C. et al ( 1989) Genomics 16:325-332), single strand conformational gel polymorphism (SSCP) analysis (Orita, M. et al (1989) Proc Natl Acad Sci USA 86:2766-2770) and direct DNA sequencing.
5.3 Primer Pairs for Use In Identifying Subjects Having a Predisposition to 3o Glaucoma Two primer pairs that can be used in conjunction with the polymerase chain reaction to amplify a 190 base pair sequence from human genomic DNA that harbors mutations causing glaucoma (primers 1 and 2 in Table 3) have been identified.

WO 98/20131 PCT/US97/20702 _ ' _89_ .

Primer 1 forward - ATACTGCCTAGGCCACTGGA (SEQ ID
N0.4) reverse - CAATGTCCGTGTAGCCACC (SEQ ID
NO.S) Primer 2 forward - GAACTCGAACAAACCTGGGA (SEQ ID
N0.6) reverse - CATGCTGCTGTACTTATAGCGG (SEQ ID
N0.7) These primers which hybridize to a portion of the Trabecular Meshwork Induced Glucocorticoid (TIGR) gene (International Publication No. 96/14411 to Nguyen et al.) were used to screen 410 patients with glaucoma and 81 normal individuals. Four amino acid altering sequence changes were detected in a total of 12 glaucoma patients (2.9%). No amino acid altering sequence changes were observed in the normal individuals.
2o The specific mutations observed are shown in Figure 4 below the normal sequence. The prevalence of mutations in the segment of DNA amplified by these primer pairs suggest that use of these primers in conjunction with an appropriate detection method can be used to identify a predisposition to glaucoma in approximately 100 thousand patients in the United States alone.

WO 98/20131 _9~_ PCT/US97/20702 _ ' SEQUENCE LISTING
' (1) GENERAL INFORMATION:
(i) APPLICANT:
(A) NAME: University of Iowa Research Foundation tB) STREET: 214 Technology Innovation Center, Oakdale Research Campus, 20STIC, I00 Oakdale Campus (C) CITY: Iowa City (D) STATE: Iowa (E) COUNTRY: USA
(F) ZIP: 52242-5000 (ii) TITLE OF INVENTION: GLAUCOMA COMPOSITIONS AND THERAPEUTIC
AND DIAGNOSTIC USES THEREFOR
(iii) NUMBER OF SEQUENCES: 18 (iv) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk (B) COMPUTER: IBM PC compatible (C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Release #1.0, Version #1.30 (v) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: PCT Unassigned (B) FILING DATE: Concurrently Herewith (vi} PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/822,999 (B) FILING DATE: 21-MAR-1997 (vi) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/791,347 (B) FILING DATE: 30-JAN-1997 (vi) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/748,479 (B) FILING DATE: 08-NOV-1996 (vi) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/239,218 (B) FILING DATE: 28-APR-1994 (2) INFORMATION FOR SEQ ID N0:1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 2166 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:

, AGAAGGGG~C AGTGTCCCCAGACCCGAGACACTGCTCGGGCTGTGCCACCAGGCTCCAGA780 WO 98/20131 PCT/US97/20702 _ ' AATTGTAGTC TGAGGGCGTA GACAATTTCA TATAATAAAT ATCCTTTATC TTCTGTCAGC ~ 1920 (2) INFORMATION FOR SEQ ID N0:2:
(i) SEQUENCE CHARACTERISTICS:
(A} LENGTH: 1512 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1512 (xi)SEQUENCE SEQID
DESCRIPTION: N0:2:

MetArg PhePheCys AlaArg CysCysSer PheGlyProGlu MetPro AlaVal GlnLeuLeu LeuLeu AlaCysLeu ValTrpAspVal GlyAla ArgThr AlaGlnLeu ArgLys AlaAsnAsp GlnSerGlyArg CysGln TyrThr PheSerVal AlaSer ProAsnGlu SerSerCysPro GluGln SerGln AlaMetSer ValIle HisAsnLeu GlnArgAspSer SerThr GlnArg LeuAspLeu GluAla ThrLysAla ArgLeuSerSer LeuGlu SerLeu LeuHisGln LeuThr LeuAspGln AlaAlaArgPro GlnGlu Thr Gln Glu Gly Leu Gln Arg Glu Leu Gly Thr Leu Arg Arg Glu Arg ACC ACC

AspGln LeuGluThr GlnThrArgGluLeu GluThrAlaTyr SerAsn LeuLeu ArgAspLys SerValLeuGluGlu GluLysLysArg LeuArg GlnGlu AsnGluAsn LeuAlaArgArgLeu GluSerSerSer GlnGlu ValXaa ArgLeuArg ArgGlyGlnCysPro GlnThrArgAsp ThrAla ArgAla ValProPro GlySerArgGluVal SerThrTrpAsn LeuAsp ThrLeu AlaPheGln GluLeuLysSerGlu LeuThrGluVal ProAla TCCCGA ATTTTGAAG GAGAGCCCATCTGGC TATCTCAGGAGT GGAGAG 720.

SerArg IleLeuLys GluSerProSerGly TyrLeuArgSer GlyGlu GlyAsp ThrGlyCys GlyGluLeuValTrp ValGlyGluPro LeuThr LeuArg ThrAlaGlu ThrIleThrGlyLys TyrGlyValTrp MetArg AspPro LysProThr TyrProTyrThrGln GluThrThrTrp ArgIle AspThr ValGlyThr AspValArgGlnVal PheGluTyrAsp LeuIle SerGln PheMetGln GlyTyrProSerLys ValHisIleLeu ProArg ProLeu GluSerThr GlyAlaValValTyr SerGlySerLeu TyrPhe GlnGly AlaGluSer ArgThrValIleArg TyrGluLeuAsn ThrGlu _94_ Thr Val Lys Ala Glu Lys Glu Ile Pro Gly Ala Gly Tyr His Gly Gln PheProTyrSer TrpGlyGlyTyr ThrAspIleAspLeu AlaValAsp GluAlaGlyLeu TrpValIleTyr SerThrAspGluAla LysGlyAla IleValLeuSer LysLeuAsnPro GluAsnLeuGluLeu GluGlnThr TrpGluThrAsn IleArgLysGln SerValAlaAsnAla PheIleIle CysGlyThrLeu TyrThrValSer SerTyrThrSerAla AspAlaThr GTCAACTTTGCT TATGACACAGGC ACAGGTATCAGCAAG rCCCTGACC 1392 ValAsnPheAla TyrAspThrGly ThrGlyIleSerLys hr LeuThr IleProPheLys AsnArgTyrLys TyrSerSerMetIle 'rspTyrAsn ProLeuGluLys LysLeuPheAla TrpAspAsnLeuAsn MetValThr TyrAspIIeLys LeuSerLysMet (2) INFORMATION FOR SEQ ID N0:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 504 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:3:
Met Arg Phe Phe Cys Ala Arg Cys Cys Ser Phe Gly Pro Glu Met Pro 1 5 10 ~15 Ala Val Gln Leu Leu Leu Leu Ala Cys Leu Val Trp Asp Val Gly Ala Arg Thr Ala Gln Leu Arg Lys Ala Asn Asp Gln Ser Gly Arg Cys Gln Tyr Thr Phe Ser Val Ala Ser Pro Asn Glu Ser Ser Cys Pro Glu Gln Ser Gln Ala Met Ser Val Ile His Asn Leu Gln Arg Asp Ser Ser Thr Gln Arg Leu Asp Leu Glu Ala Thr Lys Ala Arg Leu Ser Ser Leu Glu Ser Leu Leu His Gln Leu Thr Leu Asp Gln Ala Ala Arg Pro Gln Glu Thr Gln Glu Gly Leu Gln Arg Glu Leu Gly Thr Leu Arg Arg Glu Arg Asp Gln Leu Glu Thr Gln Thr Arg Glu Leu Glu Thr Ala Tyr Ser Asn Leu Leu Arg Asp Lys Ser Val Leu Glu Glu Glu Lys Lys Arg Leu Arg Gln Glu Asn Glu Asn Leu Ala Arg Arg Leu Glu Ser Ser Ser Gln Glu Val Xaa Arg Leu Arg Arg Gly Gln Cys ?ro Gln Thr Arg Asp Thr Ala Arg Ala Val Pro Pro Gly Ser Arg Glu Val Ser Thr Trp Asn Leu Asp Thr Leu Ala Phe Gln Glu Leu Lys Ser Glu Leu Thr Glu Val Pro Ala Ser Arg Ile Leu Lys Glu Ser Pro Ser Gly Tyr Leu Arg Ser Gly Glu Gly Asp Thr Gly Cys Gly Glu Leu Val Trp Val Gly Glu Pro Leu Thr Leu Arg Thr Ala Glu Thr Ile Thr Gly Lys Tyr Gly Val Trp Met Arg Asp Pro Lys Pro Thr Tyr Pro Tyr Thr Gln Glu Thr Thr Trp Arg Ile Asp Thr Val Gly Thr Asp Val Arg Gln Val Phe Glu Tyr Asp Leu Ile Ser Gln Phe Met Gln Gly Tyr Pro Ser Lys Val His Ile Leu Pro Arg Pro Leu Glu Ser Thr Gly Ala Val Val Tyr Ser Gly Ser Leu Tyr Phe Gln Gly Ala Glu Ser Arg Thr Val Ile Arg Tyr Glu Leu Asn Thr Glu Thr Val Lys Ala Glu Lys Glu Ile Pro Gly Ala Gly Tyr His Gly Gln -96- .
_ Phe Pro Tyr Ser Trp Gly Gly Tyr Thr Asp Ile Asp Leu Ala Val Asp Glu Ala Gly Leu Trp Val Ile Tyr Ser Thr Asp Glu Ala Lys Gly Ala Ile Val Leu Ser Lys Leu Asn Pro Glu Asn Leu Glu Leu Glu Gln Thr Trp Glu Thr Asn Ile Arg Lys Gln Ser Val Ala Asn Ala Phe Ile Ile Cys Gly Thr Leu Tyr Thr Val Ser Ser Tyr Thr Ser Ala Asp Ala Thr Val Asn Phe Ala Tyr Asp Thr Gly Thr Gly Ile Ser Lys Thr Leu Thr Ile Pro Phe Lys Asn Arg Tyr Lys Tyr Ser Ser Met Ile Asp Tyr Asn Pro Leu Glu Lys Lys Leu Phe Ala Trp Asp Asn Leu Asn Met Val Thr Tyr Asp Ile Lys Leu Ser Lys Met (2) INFORMATION FOR SEQ ID N0:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:4:

(2) INFORMATION FOR SEQ ID N0:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:5:

-97- , -(2) INFORMATION FOR SEQ ID N0:6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:6:

(2) INFORMATION FOR SEQ ID N0:7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 22 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "primer"
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:7:

(2) INFORMATION FOR SEQ ID N0:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 29 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:8:

(2) INFORMATION FOR SEQ ID N0:9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 642 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single WO 98/20131 PCT/US97120702' _98_ (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE
DESCRIPTION:
SEQ ID
N0:9:

CAGTCCATAGCA_~1AAGGAGA AATAAAAGGA TGTGTCTGCA TATGAGCAGT300 CCGGGTGAGA

(2) INFORMATION
FOR SEQ
ID N0:10:

(i) SEQUENCE
CHARACTERISTICS:

(A) LENGTH: 685 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE
TYPE:
DNA (genomic) (xi) SEQUENCE
DESCRIPTION:
SEQ ID
NO:10:

_99_ ACATGCGATA ACTGAGGCGT AGAGAGCTAA GTAACTTACT GAAAGTCACA CAGCCAGCGG~ -590 (2) INFORMATION FOR SEQ ID NO:11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 195 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:11:

(2) INFORMATION FOR SEQ ID N0:12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 195 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:12:

(2) INFORMATION FOR SEQ ID N0:13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 190 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single WO 98/20131 _ ~ ~~_ PCT/US97/20702 -_ (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:16:

(2) INFORMATION FOR SEQ ID N0:17:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 11 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:17:

(2) INFORMATION FOR SEQ ID N0:18:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 11 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:18:

Claims (61)

What is claimed is:
1. An isolated nucleic acid molecule comprising a member selected from the group consisting of:
a) a nucleic acid set forth in any of SEQ. ID. Nos. 1 or 2 or the complement of a nucleic acid set forth in any of SEQ. ID.
Nos. 1 or 2; and b) a nucleic acid capable of hybridizing under an appropriate stringency to a nucleic acid molecule of a).
2. An isolated nucleic acid molecule of claim 1, wherein the appropriate stringency is mildly stringent.
3. An isolated nucleic acid molecule of claim 1, wherein the appropriate stringency is highly stringent.
4. An isolated nucleic acid molecule of claim 1, which further comprises a label.
5. An isolated nucleic acid molecule of claim 1, wherein the nucleic acid encodes a functional GLC1A bioactivity.
6. An isolated nucleic acid molecule of claim 5, wherein the nucleic acid encodes a protein, which is capable of treating or preventing glaucoma.
7. A vector comprising a nucleic acid molecule of claim 1.
8. A vector of claim 7, which is capable of replicating in a cell.
9. A host cell comprising a vector of claim 7.
10. A transgenic animal containing a vector of claim 7.
11. A method for treating glaucoma in a human subject, comprising administering to the subject, a therapeutically effective amount of a compound that modulates the expression of a GLC1A gene or the activity of a GLC1A gene product.
12. A method of claim 11, wherein the compound is selected from the group consisting of: a protein, peptide, peptidomimetic, small molecule or nucleic acid.
13. A method of claim 12, wherein the nucleic acid is selected from the group consisting of: a gene, antisense, ribozyme and triplex nucleic acid.
14. A method of claim 11, wherein the compound is an agonist of a normal GLCA1 gene or gene product.
15. A method of claim 14, wherein the compound is a gene therapeutic.
16. A method of claim 14. wherein the compound is a protein therapeutic.
17. A method of claim 14, wherein the compound is an agent that upregulates the expression of a subject's endogenous GLC1A gene.
18. A method of claim 17, wherein the compound is a strong promoter.
19. A method of claim 11, wherein the compound is comprised of an antagonist of a mutant GLC1A gene or gene product.
20. A method of claim 19, wherein the compound is an antisense, ribozyme or triple helix molecule.
21. A method of claim 20, wherein the compound is a small molecule, peptide, or peptidomimetic.
22. A method of claim 21, wherein the compound is an antibody.
23. A method for determining whether a human subject has or is at risk for developing glaucoma, comprising:

a) obtaining a sample comprising a GLC1A protein from a subject;
and b) determnning whether the GLC1A protein is normal or altered.
24. The method of claim 23, wherein the determining step b) comprises contacting the protein with an antibody that is capable of binding tow a wild-type GLC1A protein but not an altered GLC1A protein or an antibody, which is capable of binding to an altered GLC1A protein, but not a normal GLC1A protein.
25. A method for screening fox a GLC1A agonist or antagonist comprising the steps of:
a) combining a GLC1A polypeptide or bioactive fragment thereof, a GLC1A binding partner and a test compound under conditions wherein, but for the test compound, the GLC1A protein and GLC1A
binding partner are able to interact; and b) detecting the extent to which, in the presence of the test compound, a GLC1A protein/GLC1A binding partner complex is formed, wherein an increase or decrease in the amount of complex formed in the presence of the compound relative to in the absence of the compound indicates that the compound interacts with a GLC1A
polypeptide or GLC1A binding partner.
26. A method of claim 25, wherein th,e GLC1A polypeptide is a human GLC1A polypeptide.
27. A method of claim 25, wherein the GLC1A polypeptide is encoded by a wildrype GLC1A gene.
28. A method of claim 25, wherein the GLC1A polypeptide is encoded by a mutant GLC1A gene.
29. A method of claim 25, wherein the agonist or antagonist is selected from the group consisting of: a protein, peptide, peptidomimetic, small molecule or nucleic acid.
30. A method of claim 29, wherein the nucleic acid is selected from the group consisting of: a GLC1A gene, an antisense, ribozyme and triplex nucleic acid.
31. A method of claim 25, wherein the compound is a steroid.
32. A method of claim 25, wherein the compound blocks the steroid induced activation of GLC1A.
33. A method of claim 25, which additionally comprises the step of preparing a pharmaccutical composition from the compound.
34. A method for identifying a compound that modulates a GLC1A
bioactivity, comprising the steps of:
(a) contacting an appropriate amount of the compound with a cell or cellular extract, which expresses a GLC1A gene; and (b) determining the resulting GLC1A bioactivity, wherein an increase or decrease in the GLC1A bioactivity in the presence of the compound as compared to the bioactivity in the absence of the compound indicates that the compound is a modulator of a GLC1A
bioactivity.
35. A method of claim 34, wherein the GLC1A gene is a human GLC1A
gene.
36. A method of claim 34, wherein the GLC1A gene is a wildtype gene.
31. A method of claim 34, wherein the GLC1A gene is a mutant gene.
38. A method of claim 34, wherein the modulator is an agonist of a GLC1A
bioactivity.
39. A method of claim 34, wherein the modulator is an antagonist of a GLC1A bioactivity.
40. A method of claim 34, wherein in step (b), the GLC1A bioactivity is determined by determining the expression level of a GLC1A gene.
41. A method of claim 40, wherein the expression level is determined by detecting the amount of mRNA transcribed from a GLC1A gene.
42. A method of claim 40, wherein the expression level is determined by detecting the amount of GLC1A gene product produced.
43. A method of claim 42, wherein the expression level is determined using an anti-GLC1A antibody in an immunodetection assay.
44. A method of claim 34, which additionally comprises the step of preparing a pharmaceutical composition from the compound.
45. A method of claim 34, wherein said cell is contained in an animal.
46. A method of claim 45, wherein the animal is transgenic.
47. A method of claim 46, wherein the transgenic animal contains a human GLC1A gene.
48. A method for identifying a compound that modulates steroid induction of GLC1A expression, comprising the steps of:
(a) contacting an appropriate amount of the compound with a cell or cellular extract, which is steroid induced to express a GLC1A gene;
and (b) determining the expression level of the GLC1A gene, wherein an increase or decrease in the expression level in the presence of the compound as compared to the expression level in the absence of the compound indicates that the compound modulates steroid induction of GLC1A expression.
49. A method of claim. 48, wherein the GLC1A gene is a human GLC1A
gene.
50. A method of claim 48, wherein the GLC1A gene is a wildtype gene.
51. A method cf claim 48, wherein the GLC1A gene is a mutant gene.
52. A method of claim 48, wherein the compound is an agonist of a GLC1A
bioactivity.
53. A method of claim 48, wherein the compound is an antagonist of a GLC1A bioactivity.
54. A method of claim 48, wherein the expression level is determined by detecting the amount of mRNA transcribed from a GLC1A gene.
55. A method of claim 48, wherein the expression level is determined by detecting the amount of GLC1A gene product produced.
56. A method of claim 48, wherein the expression level is determined using an anti-GLC1A antibody in an immunodetection assay.
57. A method of claim 48, which additionally comprises the step of preparing a pharmaceutical composition from the compound.
58. A method for identifying whether a test compound is a GLC1A binding partner or measuring the strength of an interaction between a GLC1A
polypeptide and said GLC1A binding partner comprising:

(a) allowing (i) a first molecule comprising a GLC1A polypeptide operably linked to a heterologous DNA binding domain to interact with (ii) a second molecule comprising a test compound operably linked to a polypeptide transcriptional activation domain and (iii) a hybrid reporter gene comprising a nucleic acid encoding a reporter operably linked to a DNA sequence comprising a binding site for said heterologous DNA binding domain; and (b) detecting or measuring the expression of the hybrid reporter gene as an indication of the existence or strength of an interaction between the first molecule and the second molecule wherein high levels of hybrid reporter expression indicate a strong interaction between GLC1A and said test molecule thereby identifying a test compound which is a GLC1A binding partner.
59. A method of claim 58, wherein said second molecule is encoded by a nucleic acid and comprises a test polypeptide operably linked to a polypeptide transcriptional activation domain, and which further comprises the step of isolating the nucleic acid encoding said second molecule from a cell expressing the hybrid reporter gene.
60. A method for identifying a molecule which is a downstream or an upstream component of a GLC1A biochemical pathway or for measuring the strength of the interaction between a GLC1A biochemical pathway component and a GLG1A binding partner comprising:
(a) allowing (i) a first molecule comprising a GLC1A binding partner polypeptide operably linked to a heterologous DNA binding domain to interact with (ii) a second molecule comprising a test molecule operably linked to a polypeptide transcriptional activation domain and (iii) a hybrid reporter gene comprising a nucleic acid encoding a reporter operably linked to a DNA sequence comprising a binding site for said heterologous DNA binding domain; and (b) detecting or measuring the expression of the hybrid reporter gene as an indication of the existence or strength of an interaction between the first molecule and the second molecule wherein high levels of hybrid reporter expression indicate a strong interaction between a GLClA binding partner and said test molecule thereby identifying a test molecule which is a downstream or an upstream component of the GLClA biochemical pathway.
61. A method of claim 60, wherein said second molecule is encoded by a nucleic acid and comprises a test polypeptide operably linked to a polypeptide transcriptional activation domain, and which further comprises the step of isolating the nucleic acid encoding said second molecule from a cell expressing the hybrid reporter gene.
CA002271235A 1996-11-08 1997-11-07 Glaucoma-associated protein and corresponding nucleic acid and their therapeutic and diagnostic uses Abandoned CA2271235A1 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US08/748,479 US5925748A (en) 1994-04-28 1996-11-08 DNA diagnostics for glaucoma
US08/791,347 US5885776A (en) 1997-01-30 1997-01-30 Glaucoma compositions and therapeutic and diagnositic uses therefor
US08/822,999 US6271026B1 (en) 1997-03-21 1997-03-21 Glaucoma compositions
US08/822,999 1997-03-21
US08/748,479 1997-03-21
US08/791,347 1997-03-21
PCT/US1997/020702 WO1998020131A1 (en) 1996-11-08 1997-11-07 Glaucoma-associated protein and corresponding nucleic acid and their therapeutic and diagnostic uses

Publications (1)

Publication Number Publication Date
CA2271235A1 true CA2271235A1 (en) 1998-05-14

Family

ID=27419366

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002271235A Abandoned CA2271235A1 (en) 1996-11-08 1997-11-07 Glaucoma-associated protein and corresponding nucleic acid and their therapeutic and diagnostic uses

Country Status (5)

Country Link
EP (1) EP0942975A1 (en)
JP (1) JP2001503631A (en)
AU (1) AU5436498A (en)
CA (1) CA2271235A1 (en)
WO (1) WO1998020131A1 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6956103B2 (en) 1994-04-28 2005-10-18 The University Of Iowa Research Foundation Glaucoma therapeutics and diagnostics
US6403307B1 (en) 1997-03-21 2002-06-11 University Of Iowa Research Foundation Glaucoma therapeutics and diagnostics
US5789169A (en) * 1994-11-03 1998-08-04 Regents Of The University Of California Methods for the diagnosis of glaucoma
US7138511B1 (en) 1997-01-28 2006-11-21 The Regents Of The University Of California Nucleic acids, kits and methods for the diagnosis, prognosis and treatment of glaucoma and related disorders
US6475724B1 (en) 1997-01-28 2002-11-05 The Regents Of The University Of California Nucleic acids, kits, and methods for the diagnosis, prognosis and treatment of glaucoma and related disorders
US6171788B1 (en) 1997-01-28 2001-01-09 The Regents Of The University Of California Methods for the diagnosis, prognosis and treatment of glaucoma and related disorders
CA2386177A1 (en) * 1999-10-08 2001-04-19 Toshihiko Matsuo Oculomedin and glaucoma
JP4255382B2 (en) * 2001-10-31 2009-04-15 アルコン,インコーポレイテッド Bone morphogenetic protein (BMP), BMP receptor and BMP binding protein and their use in the diagnosis and treatment of glaucoma
US7071211B2 (en) * 2002-09-27 2006-07-04 Bausch & Lomb Inc. Small organic molecules that increase the activity of gelatinase a in ocular cells

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5606043A (en) * 1994-11-03 1997-02-25 The Regents Of The University Of California Methods for the diagnosis of glaucoma

Also Published As

Publication number Publication date
AU5436498A (en) 1998-05-29
EP0942975A1 (en) 1999-09-22
WO1998020131A1 (en) 1998-05-14
JP2001503631A (en) 2001-03-21

Similar Documents

Publication Publication Date Title
US5800998A (en) Assays for diagnosing type II diabetes in a subject
US5885776A (en) Glaucoma compositions and therapeutic and diagnositic uses therefor
AU6952498A (en) Novel human delta3 compositions and therapeutic and diagnostic uses therefor
US6031076A (en) Conservin compositions
US6143491A (en) Therapeutic compositions and methods and diagnostic assays for type II diabetes involving HNF-1
US6399760B1 (en) RP compositions and therapeutic and diagnostic uses therefor
US6833239B1 (en) Methods to identify modulators of FKHL7 DNA-binding activity
US6593104B1 (en) Macular degeneration diagnostics and therapeutics
US6403307B1 (en) Glaucoma therapeutics and diagnostics
CA2271235A1 (en) Glaucoma-associated protein and corresponding nucleic acid and their therapeutic and diagnostic uses
US6518411B1 (en) RGS compositions and therapeutic and diagnostic uses therefor
WO1998046748A1 (en) Therapeutic compositions and diagnostic assays for diseases involving trbp
AU766890B2 (en) Glaucoma therapeutics and diagnostics based on a novel human transcription factor
US6271026B1 (en) Glaucoma compositions
US6956103B2 (en) Glaucoma therapeutics and diagnostics
US6008014A (en) Method of making lipid metabolic pathway compositions
WO1998021363A1 (en) Compositions and methods for treating type ii diabetes involving hnf-4
AU3556402A (en) Glaucoma-associated protein and corresponding nucleic acid and their therapeutic and diagnostic uses
US6306586B1 (en) Methods and compositions for the diagnosis and treatment of cataracts
US20050123960A1 (en) Glaucoma therapeutics and diagnostics
WO1998040495A1 (en) Doublin, a gene involved in neuronal development and uses therefor

Legal Events

Date Code Title Description
EEER Examination request
FZDE Dead