CA2408315A1 - Aminoacyl trna synthetases - Google Patents

Aminoacyl trna synthetases Download PDF

Info

Publication number
CA2408315A1
CA2408315A1 CA002408315A CA2408315A CA2408315A1 CA 2408315 A1 CA2408315 A1 CA 2408315A1 CA 002408315 A CA002408315 A CA 002408315A CA 2408315 A CA2408315 A CA 2408315A CA 2408315 A1 CA2408315 A1 CA 2408315A1
Authority
CA
Canada
Prior art keywords
polypeptide
polynucleotide
atrs
seq
antibody
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
CA002408315A
Other languages
French (fr)
Inventor
Henry Yue
Tom Y. Tang
Chandra Patterson
Ameena R. Gandhi
Catherine M. Tribouley
Ernestine A. Lee
Monique G. Yao
Olga Bandman
Dyung Alina M. Lu
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.)
Incyte Corp
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2408315A1 publication Critical patent/CA2408315A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/93Ligases (6)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Abstract

The invention provides human aminoacyl tRNA synthetases (ATRS) and polynucleotides which identify and encode ATRS. The invention also provides expression vectors, host cells, antibodies, agonists, and antagonists. The invention also provides methods for diagnosing, treating, or preventing disorders associated with aberrant expression of ATRS.

Description

AMINOACYL tRNA SYNTHETASES
TECHNICAL FIELD
This invention relates to nucleic acid and amino acid sequences of aminoacyl tRNA
synthetases and to the use of these sequences in the diagnosis, treatment, and prevention of cell proliferative and autoimmune/inflammatory disorders, and in the assessment of the effects of exogenous compounds on the expression of nucleic acid and amino acid sequences of aminoacyl tRNA synthetases.
SACI~GROUND OF THE INVENTION
Correct translation of the genetic code depends upon each amino acid forming a linkage with the appropriate transfer RNA (tRNA). The aminoacyl-tRNA synthetases (aaRSs) are essential proteins found in all living organisms. The aaRSs are responsible for the activation and correct attachment of an amino acid with its cognate tRNA, as the first step in protein biosynthesis.
Prokaryotic organisms have at least twenty different types of aaRSs, one for each different amino acid, while eukaryotes usually have two aaRSs, a cytosolic form and a mitochondria) form, for each different amino acid. The 20 aaRS enzymes can be divided into two structural classes. Class I
enzymes add amino acids to the 2' hydroxyl at the 3' end of tRNAs while Class II enzymes add amino acids to the 3' hydroxyl at the 3' end of tRNAs. Each class is characterized by a distinctive topology of the catalytic domain. Class I enzymes contain a catalytic domain based on the nucleotide-binding 'Rossman fold'. In particular, a consensus tetrapeptide motif is highly conserved (Prosite Document PDOC00161, Aminoacyl-transfer RNA synthetases class-I signature). Class I
enzymes are specific for arginine, cysteine, glutamic acid, glutamine, isoleucine, leucine, methionine, tyrosine, tryptophan, and vafine. Class II enzymes contain a central catalytic domain, which consists of a seven-stranded antiparallel J3-sheet domain, as well as N- and C- terminal regulatory domains. Class II enzymes are separated into two groups based on the heterodimeric or homodimeric structure of the enzyme; the latter group is further subdivided by the structure of the N- and C-terminal regulatory domains (Ha~~tlein, M. and Cusack, S. (1995) J. Mol. Evol. 40:519-530). Class II
enzymes are specific for alanine, asparagine, aspartic acid, glycine, histidine, lysine, phenylalanine, proline, serine, and threonine.
Certain aaRSs also have editing functions. IleRS, for example, can misactivate valine to form Val-tRNAne, but this product is cleared by a hydrolytic activity that destroys the mischarged product.
This editing activity is located within a second catalytic site found in the connective polypeptide 1 region (CP1), a long insertion sequence within the Rossman fold domain of Class I enzymes (Schimmel, P. et al. (1998) FASEB J. 12:1599-1609). AaRSs also play a role in tRNA processing. It has been shown that mature tRNAs are charged with their respective amino acids in the nucleus before export to the cytoplasm, and charging may serve as a quality control mechanism to insure the tRNAs are functional (Martinis, S.A. et al. (1999) EMBO J. 18:4591-4596).
In addition to their function in protein synthesis, specific aminoacyl tRNA
synthetases also play roles in cellular fidelity, RNA splicing, RNA trafficking, apoptosis, and transcriptional and translational regulation. For example, human tyrosyl-tRNA synthetase can be proteolytically cleaved into two fragments with distinct cytokine activities. The carboxy-terminal domain exhibits monocyte and leukocyte chemotaxis activity as well as stimulating production of myeloperoxidase, tumor necrosis factor-cc, and tissue factor. The N-terminal domain binds to the interleukin-8 type A receptor and functions as an interleukin-8-like cytokine. Human tyrosyl-tRNA synthetase is secreted from apoptotic tumor cells and may accelerate apoptosis (Wakasugi, K., and Schimmel, P. (1999) Science 284:147-151). Mitochondria) Neurosuora crassa TyrRS and S. cerevisiae LeuRS
are essential factors for certain group I intron splicing activities, and human mitochondria) LeuRS
can substitute for the yeast LeuRS in a yeast null strain. Certain bacterial aaRSs are involved in regulating their own transcription or translation (Martinis, supra). Several aaRSs are able to synthesize diadenosine oligophosphates, a class of signalling molecules with roles in cell proliferation, differentiation, and apoptosis (Kisselev, L.L et al. (1998) FEBS Lett. 427:157-163; Vartanian, A.
et al. (1999) FEBS Lett.
456:175-180).
Autoantibodies against aminoacyl-tRNAs are generated by patients with autoimrnune diseases such as rheumatic arthritis, dermatomyositis and polymyositis, and correlate strongly with complicating interstitial lung disease (ILD) (Freist, W. et al. (1999) Biol. Chem. 380:623-646; Freist, W. et al.
(1996) Biol. Chem. Hoppe Seyler 377:343-356). These antibodies appear to be generated in response to viral infection, and coxsackie virus has been used to induce experimental viral myositis in animals.
Comparison of aaRS structures between humans and pathogens has been useful in the design of novel antibiotics (Schimmel, supra). Genetically engineered aaRSs have been utilized to allow site-specific incorporation of unnatural amino acids into proteins in vivo (Liu, D.R. et al. (1997) Proc. Nat).
Acad. Sci. USA 94:10092-10097).
The discovery of new aminoacyl tRNA synthetases and the polynucleotides encoding them satisfies a need in the art by providing new compositions which are useful in the diagnosis, prevention, and treatment of cell proliferative and autoimmunelinflammatory disorders, and in the assessment of the effects of exogenous compounds on the expression of nucleic acid and amino acid sequences of aminoacyl tRNA synthetases SUMMARY OF THE INVENTION
The invention features purified polypeptides, aminoacyl tRNA synthetases, referred to collectively as "ATRS" and individually as "ATRS-1," "ATRS-2," "ATRS-3," and "ATRS-4". In one aspect, the invention provides an isolated polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-4, b) a naturally occurring polypeptide comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID N0:1-4, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ
ID N0:1-4, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID N0:1-4. In one alternative, the invention provides an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:l-4.
The invention further provides an isolated polynucleotide encoding a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-4, b) a naturally occurring polypeptide comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-4, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID N0:1-4, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:l-4. In one alternative, the polynucleotide encodes a polypeptide selected from the group consisting of SEQ
ID N0:1-4. In another alternative, the polynucleotide is selected from the group consisting of SEQ ID NO:S-8.
Additionally, the invention provides a recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide encoding a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ )D NO:1-4, b) a naturally occurring polypeptide comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:l-4, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ )D NO:1-4, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ~ N0:1-4. In one alternative, the invention provides a cell transformed with the recombinant polynucleotide. In another alternative, the invention provides a transgenic organism comprising the recombinant polynucleotide.
The invention also provides a method for producing a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-4, b) a naturally occurring polypeptide comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID N0:1-4, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ 1D N0:1-4, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-4. The method comprises a) culturing a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide encoding the polypeptide, and b) recovering the polypeptide so expressed.
Additionally, the invention provides an isolated antibody which specifically binds to a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ 1D NO:1-4, b) a naturally occurring polypeptide comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:l-4, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID N0:1-4, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID N0:1-4.
The invention further provides an isolated polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ
ID NO:S-8, b) a naturally occurring polynucleotide comprising a polynucleotide sequence at least 90%
identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:S-8, c) a 2o polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). In one alternative, the polynucleotide comprises at least 60 contiguous nucleotides.
Additionally, the invention provides a method for detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ
>D NO:S-8, b) a naturally occurnng polynucleotide comprising a polynucleotide sequence at least 90%
identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:S-8, c) a polynucleotide complementary to the polyriucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). The method comprises a) hybridizing the sample with a probe comprising at least 20 contiguous nucleotides comprising a sequence complementary to said target polynucleotide in the sample, and which probe specifically hybridizes to said target polynucleotide, under conditions whereby a hybridization complex is formed between said probe and said target polynucleotide or fragments thereof, and b) detecting the presence or absence of said hybridization complex, and optionally, if present, the amount thereof. In one alternative, the probe comprises at least 60 contiguous nucleotides.
The invention further provides a method for detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ >D
N0:5-8, b) a naturally occurring polynucleotide comprising a polynucleotide sequence at least 90%
identical to a polynucleotide sequence selected from the group consisting of SEQ ID N0:5-8, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). The method comprises a) amplifying said target polynucleotide or fragment thereof using polymerase chain reaction amplification, and b) detecting the presence or absence of said amplified target polynucleotide or fragment thereof, and, optionally, if present, the amount thereof.
The invention further provides a composition comprising an effective amount of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID N0:1-4, b) a naturally occurring polypeptide comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ )D N0:1-4, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID N0:1-4, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:l-4, and a pharmaceutically acceptable excipient. In one embodiment, the composition comprises an amino acid sequence selected from the group consisting of SEQ >D NO:l-4. The invention additionally provides a method of treating a disease or condition associated with decreased expression of functional ATRS, comprising administering to a patient in need of such treatment the composition.
The invention also provides a method for screening a compound for effectiveness as an agonist of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ )D NO:1-4, b) a naturally occurring polypeptide comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ )D NO:1-4, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ~ N0:1-4, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID N0:1-4. The method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting agonist activity in the sample. In one alternative, the invention provides a composition comprising an agonist compound identified by the method and a pharmaceutically acceptable excipient. In another alternative, the invention provides a method of treating a disease or condition associated with decreased expression of functional ATRS, comprising administering to a patient in need of such treatment the composition.
Additionally, the invention provides a method for screening a compound for effectiveness as an antagonist of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ B7 NO:1-4, b) a naturally occurring polypeptide comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-4, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ m NO:1-4, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-4. The method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting antagonist activity in the sample.
In one alternative, the invention provides a composition comprising an antagonist compound identified by the method and a pharmaceutically acceptable excipient. In another alternative, the invention provides a method of treating a disease or condition associated with overexpression of functional ATRS, comprising administering to a patient in need of such treatment the composition.
The invention further provides a method of screening for a compound that specifically binds to a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ~ NO:1-4, b) a naturally occurring polypeptide comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ m NO:1-4, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ B? N0:1-4, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ
ID NO:1-4. The method comprises a) combining the polypeptide with at least one test compound under suitable conditions, and b) detecting binding of the polypeptide to the test compound, thereby identifying a compound that specifically binds to the polypeptide.
The invention further provides a method of screening for a compound that modulates the activity of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-4, b) a naturally occurring palypeptide comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ m N0:1-4, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID N0:1-4, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID N0:1-4. The method comprises a) combining the polypeptide with at least one test compound under conditions permissive for the activity of the polypeptide, b) assessing the activity of the polypeptide in the presence of the test compound, and c) comparing the activity of the polypeptide in the presence of the test compound with the activity of the polypeptide in the absence of the test compound, wherein a change in the activity of the polypeptide in the presence of the test compound is indicative of a compound that modulates the activity of the polypeptide.
The invention further provides a method for screening a compound for effectiveness in altering expression of a target polynucleotide, wherein said target polynucleotide comprises a sequence selected from the group consisting of SEQ )D N0:5-8, the method comprising a) exposing a sample comprising the target polynucleotide to a compound, and b) detecting altered expression of the target polynucleotide.
The invention further provides a method for assessing toxicity of a test compound, said method comprising a) treating a biological sample containing nucleic acids with the test compound; b) hybridizing the nucleic acids of the treated biological sample with a probe comprising at least 20 contiguous nucleotides of a polynucleotide selected from the group consisting of i) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ
ID N0:5-8, ii) a naturally occurring polynucleotide comprising a polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:S-8, iii) a polynucleotide having a sequence complementary to i)> iv) a polynucleotide complementary to the polynucleotide of ii), and v) an RNA equivalent of i)-iv). Hybridization occurs under conditions whereby a specific hybridization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide selected from the group consisting of i) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ )D NO:S-8, ii) a naturally occurring polynucleotide comprising a polynucleotide sequence at least 90%
identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:S-8, iii) a polynucleotide complementary to the polynucleotide of i), iv) a polynucleotide complementary to the polynucleotide of ii), and v) an RNA equivalent of i)-iv). Alternatively, the target polynucleotide comprises a fragment of a polynucleotide sequence selected from the group consisting of i)-v) above; c) quantifying the amount of hybridization complex; and d) comparing the amount of hybridization complex in the treated 3o biological sample with the amount of hybridization complex in an untreated biological sample, wherein a difference in the amount of hybridization complex in the treated biological sample is indicative of toxicity of the test compound.

BRIEF DESCRIPTION OF THE TABLES
Table 1 summarizes the nomenclature for the full length polynucleotide and polypeptide sequences of the present invention.
Table 2 shows the GenBank identification number and annotation of the nearest GenBank homolog for polypeptides of the invention. The probability score for the match between each polypeptide and its GenBank homolog is also shown.
Table 3 shows structural features of polypeptide sequences of the invention, including predicted motifs and domains, along with the methods, algorithms, and searchable databases used for analysis of the polypeptides.
Table 4 lists the cDNA and/or genomic DNA fragments which were used to assemble polynucleotide sequences of the invention, along with selected fragments of the polynucleotide sequences.
Table 5 shows the representative cDNA library for polynucleotides of the invention.
Table 6 provides an appendix which describes the tissues and vectors used for construction of the cDNA libraries shown in Table 5.
Table 7 shows the tools, programs, and algorithms used to analyze the polynucleotides and polypeptides of the invention, along with applicable descriptions, references, and threshold parameters.
DESCRIPTION OF THE INVENTION
Before the present proteins, nucleotide sequences, and methods are described, it is understood that this invention is not limited to the particular machines, materials and methods described, as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims.
It must be noted that as used herein and in the appended claims, the singular forms "a," "an,"
and "the" include plural reference unless the context clearly dictates otherwise. Thus, for example, a reference to "a host cell" includes a plurality of such host cells, and a reference to "an antibody" is a reference to one or more antibodies and equivalents thereof known to those skilled in the art, and so forth.
3o Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs.
Although any machines, materials, and methods similar or equivalent to those described herein can be used to practice or test the present invention, the preferred machines, materials and methods are now described. All publications mentioned herein are cited for the purpose of describing and disclosing the cell lines, protocols, reagents and vectors which are reported in the publications and which might be used in connection with the invention. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.
DEFINITIONS
"ATRS" refers to the amino acid sequences of substantially purified ATRS
obtained from any species, particularly a mammalian species, including bovine, ovine, porcine, marine, equine, and human, and from any source, whether natural, synthetic, semi-synthetic, or recombinant.
The term "agonist" refers to a molecule which intensifies or mimics the biological activity of ATRS. Agonists may include proteins, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of ATRS either by directly interacting with ATRS or by acting on components of the biological pathway in which ATRS
participates.
An "allelic variant" is an alternative form of the gene encoding ATRS. Allelic variants may result from at least one mutation in the nucleic acid sequence and may result in altered mRNAs or in polypeptides whose structure or function may or may not be altered. A gene may have none, one, or many allelic variants of its naturally occurring form. Common mutational changes which give rise to allelic variants are generally ascribed to natural deletions, additions, or substitutions of nucleotides.
Each of these types of changes may occur alone, or in combination with the others, one or more times in a given sequence.
"Altered" nucleic acid sequences encoding ATRS include those sequences with deletions, insertions, or substitutions of different nucleotides, resulting in a polypeptide the same as ATRS or a polypeptide with at least one functional characteristic of ATRS. Included within this definition are polymorphisms which may or may not be readily detectable using a particular oligonucleotide probe of the polynucleotide encoding ATRS, and improper or unexpected hybridization to allelic variants, with a locus other than the normal chromosomal locus for the polynucleotide sequence encoding ATRS. The encoded protein may also be "altered," and may contain deletions, insertions, or substitutions of amino acid residues which produce a silent change and result in a functionally equivalent ATRS. Deliberate amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues, as long as the biological or immunological activity of ATRS is retained. For example, negatively charged amino acids may include aspartic acid and glutamic acid, and positively charged amino acids may include lysine and arginine. Amino acids with uncharged polar side chains having similar hydrophilicity values may include: asparagine and glutamine; and serine and threonine. Amino acids with uncharged side chains having similar hydrophilicity values may include: leucine, isoleucine, and valine; glycine and alanine;
and phenylalanine and tyrosine.
The terms "amino acid" and "amino acid sequence" refer to an oligopeptide, peptide, polypeptide, or protein sequence, or a fragment of any of these, and to naturally occurring or synthetic molecules. Where "amino acid sequence" is recited to refer to a sequence of a naturally occurring protein molecule, "amino acid sequence" and like terms are not meant to limit the amino acid sequence to the complete native amino acid sequence associated with the recited protein molecule.
"Amplification" relates to the production of additional copies of a nucleic acid sequence.
Amplification is generally carried out using polymerase chain reaction (PCR) technologies well known in the art.
The term "antagonist" refers to a molecule which inhibits or attenuates the biological activity of ATRS. Antagonists may include proteins such as antibodies, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of ATRS either by directly interacting with ATRS or by acting on components of the biological pathway in which ATRS
participates.
The term "antibody" refers to intact immunoglobulin molecules as well as to fragments thereof, such as Fab, F(ab')2, and Fv fragments, which are capable of binding an epitopic determinant.
Antibodies that bind ATRS polypeptides can be prepared using intact polypeptides or using fragments containing small peptides of interest as the immunizing antigen. The polypeptide or oJigopeptide used to immunize an animal (e.g., a mouse, a rat, or a rabbit) can be derived from the translation of RNA, or synthesized chemically, and can be conjugated to a carrier protein if desired. Commonly used carriers that are chemically coupled to peptides include bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin (KLI~. The coupled peptide is then used to immunize the animal.
The term "antigenic determinant" refers to that region of a molecule (i.e., an epitope) that makes contact with a particular antibody. When a protein or a fragment of a protein is used to immunize a host animal, numerous regions of the protein may induce the production of antibodies which bind specifically to antigenic determinants (particular regions or three-dimensional structures on the protein). An antigenic determinant may compete with the intact antigen (i.e., the immunogen used to elicit the immune response) for binding to an antibody.
The term "antasense" refers to any composition capable of base-pairing with the "sense"
(coding) strand of a specific nucleic acid sequence. Antisense compositions may include DNA; RNA;
peptide nucleic acid (PNA); oligonucleotides having modified backbone linkages such as phosphorothioates, methylphosphonates, or benzylphosphonates; oligonucleotides having modified sugar groups such as 2'-methoxyethyl sugars or 2'-methoxyethoxy sugars; or oligonucleotides having modified bases such as 5-methyl cytosine, 2'-deoxyuracil, or 7-deaza-2'-deoxyguanosine. Antisense molecules may be produced by any method including chemical synthesis or transcription. Once introduced into a cell, the complementary antisense molecule base-pairs with a naturally occurring nucleic acid sequence produced by tlae cell to form duplexes which block either transcription or translation. The designation "negative" or "minus" can refer to the antisense strand, and the designation "positive" or "plus" can refer to the sense strand of a reference DNA molecule.
The term "biologically active" refers to a protein having structural, regulatory, or biochemical functions of a naturally occurring molecule. Likewise, "immunologically active" or "immunogenic"
refers to the capability of the natural, recombinant, or synthetic ATRS, or of any oligopeptide thereof, to induce a specific immune response in appropriate animals or cells and to bind with specific antibodies.
"Complementary" describes the relationship between two single-stranded nucleic acid sequences that anneal by base-pairing. For example, 5'-AGT-3' pairs with its complement, 3'-TCA-5'.
A "composition comprising a given polynucleotide sequence" and a "composition comprising a given amino acid sequence" refer broadly to any composition containing the given polynucleotide or amino acid sequence. The composition may comprise a dry formulation or an aqueous solution.
Compositions comprising polynucleotide sequences encoding ATRS or fragments of ATRS may be employed as hybridization probes. The probes may be stored in freeze-dried form and may be associated with a stabilizing agent such as a carbohydrate. In hybridizations, the probe may be deployed in an aqueous solution containing salts (e.g., NaCl), detergents (e.g., sodium dodecyl sulfate;
SDS), and other components (e.g., Denhardt's solution, dry milk, salmon sperm DNA, etc.).
"Consensus sequence" refers to a nucleic acid sequence which has been subjected to repeated DNA sequence analysis to resolve uncalled bases, extended using the ~L-PCR kit (Applied Biosystems, Foster City CA) in the 5' and/or the 3' direction, and resequenced, or which has been assembled from one or more overlapping cDNA, EST, or genomic DNA fragments using a computer program for fragment assembly, such as the GELVIEW fragment assembly system (GCG, Madison W>] or Phrap (University of Washington, Seattle WA). Some sequences have been both extended and assembled to produce the consensus sequence.
"Conservative amino acid substitutions" are those substitutions that are predicted to least interfere with the properties of the original protein, i.e., the structure and especially the function of the protein is conserved and not significantly changed by such substitutions. The table below shows amino acids which may be substituted for an original amino acid in a protein and which are regarded as conservative amino acid substitutions.
Original Residue Conservative Substitution Ala Gly, Ser Arg His, Lys Asn Asp, Gln, His Asp Asn, Glu Cys Ala, Ser Gln Asn, Glu, His l0 Glu Asp, Gln, His Gly Ala His Asn, Arg, Gln, Glu Ile Leu, Val Leu Ile, Val Lys Arg, Gln, Glu Met Leu, Ile Phe His, Met, Leu, Trp, Tyr Ser Cys, Thr Thr Ser, Val 2o Trp Phe, Tyr Tyr His, Phe, Trp Val Ile, Leu, Thr Conservative amino acid substitutions generally maintain (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a beta sheet or alpha helical conformation, (b) the charge or hydrophobicity of the molecule at the site of the substitution, and/or (c) the bulk of the side chain.
A "deletion" refers to a change in the amino acid or nucleotide sequence that results in the absence of one or more amino acid residues or nucleotides.
The term "derivative" refers to a chemically modified polynucleotide or polypeptide.
Chemical modifications of a polynucleotide can include, for example, replacement of hydrogen by an alkyl, aryl, hydroxyl, or amino group. A derivative polynucleotide encodes a polypeptide which retains at least one biological or immunological function of the natural molecule. A
derivative polypeptide is one modified by glycosylation, pegylation, or any similar process that retains at least one biological or immunological function of the polypeptide from which it was derived.
A "detectable label" refers to a reporter molecule or enzyme that is capable of generating a measurable signal and is covalently or noncovalently joined to a polynucleotide or polypeptide.
"Differential expression" refers to increased or upregulated; or decreased, downregulated, or absent gene or protein expression, determined by comparing at least two different samples. Such comparisons may be carried out between, for example, a treated and an untreated sample, or a diseased and a normal sample.
A "fragment" is a unique portion of ATRS or the polynucleotide encoding ATRS
which is identical in sequence to but shorter in length than the parent sequence. A
fragment may comprise up to the entire length of the defined sequence, minus one nucleotide/amino acid residue. For example, a fragment may comprise from 5 to 1000 contiguous nucleotides or amino acid residues. A fragment used as a probe, primer, antigen, therapeutic molecule, or for other purposes, may be at least 5, 10, 15, 16, 20, 25, 30, 40, 50, 60, 75, 100, 150, 250 or at least 500 contiguous nucleotides or amino acid residues in length. Fragments may be preferentially selected from certain regions of a molecule. For example, a polypeptide fragment may comprise a certain length of contiguous amino acids selected from the first 250 or 500 amino acids (or first 25% or 50%) of a polypeptide as shown in a certain defined sequence. Clearly these lengths are exemplary, and any length that is supported by the specification, including the Sequence Listing, tables, and figures, may be encompassed by the present embodiments.
A fragment of SEQ ID N0:5-8 comprises a region of unique polynucleotade sequence that specifically identifies SEQ ID N0:5-8, for example, as distinct from any other sequence in the genome from which the fragment was obtained. A fragment of SEQ ID N0:5-8 is useful, for example, in hybridization and amplification technologies and in analogous methods that distinguish SEQ ID N0:5-8 from related polynucleotide sequences. The precise length of a fragment of SEQ
ID N0:5-8 and the region of SEQ ID N0:5-8 to which the fragment corresponds are routinely determinable by one of ordinary skill in the art based on the intended purpose for the fragment.
A fragment of SEQ ID NO:l-4 is encoded by a fragment of SEQ ID N0:5-8. A
fragment of SEQ ID NO:l-4 comprises a region of unique amino acid sequence that specifically identifies SEQ ID
N0:1-4. For example, a fragment of SEQ ID N0:1-4 is useful as an immunogenic peptide for the development of antibodies that specifically recognize SEQ ID N0:1-4. The precise length of a fragment of SEQ ID NO:l-4 and the region of SEQ ID N0:1-4 to which the fragment corresponds are routinely determinable by one of ordinary skill in the art based on the intended purpose for the fragment.
A "full length" polynucleotide sequence is one containing at least a translation initiation codon (e.g., methionine) followed by an open reading frame and a translation termination codon. A "full length" polynucleotide sequence encodes a "full length" polypeptide sequence.
"Homology' refers to sequence similarity or, interchangeably, sequence identity, between two or more polynucleotide sequences or two or more polypeptide sequences.
The terms "percent identity" and "% identity," as applied to polynucleotide sequences, refer to the percentage of residue matches between at least two polynucleotide sequences aligned using a standardized algorithm. Such an algorithm may insert, in a standardized and reproducible way, gaps in the sequences being compared in order to optimize alignment between two sequences, and therefore achieve a more meaningful comparison of the two sequences.
Percent identity between polynucleotide sequences may be determined using the default parameters of the CLUSTAL V algorithm as incorporated into the MEGALIGN
version 3.12e sequence alignment program. This program is part of the LASERGENE software package, a suite of molecular biological analysis programs (DNASTAR, Madison WI). CLUSTAL V is described in Higgins, D.G. and P.M. Sharp (1989) CABIOS 5:151-153 and in Higgins, D.G. et al. (1992) CABIOS
8:189-191. For pairwise alignments of polynucleotide sequences, the default parameters are set as follows: Ktuple=2, gap penalty=5, window=4, and "diagonals saved"=4. The "weighted" residue weight table is selected as the default. Percent identity is reported by CLUSTAL V as the "percent similarity" between aligned polynucleotide sequences.
Alternatively, a suite of commonly used and freely available sequence comparison algorithms is provided by the National Center for Biotechnology Information (NCB17 Basic Local Alignment Search Tool (BLAST) (Altschul, S.F. et al. (1990) J. Mol. Biol. 215:403-410), which is available from several sources, including the NCBI, Bethesda, MD, and on the Internet at http://www.ncbi.nlm.nih.gov/BLASTI. The BLAST software suite includes various sequence analysis programs including "blastn," that is used to align a known polynucleotide sequence with other polynucleotide sequences from a variety of databases. Also available is a tool called "BLAST 2 Sequences" that is used for direct pairwise comparison of two nucleotide sequences. "BLAST 2 Sequences" can be accessed and used interactively at http://www.ncbi.nlm.nih.gov/gorf/bl2.html. The "BLAST 2 Sequences" tool can be used for both blastn and blastp (discussed below). BLAST
programs are commonly used with gap and other parameters set to default settings. For example, to compare two nucleotide sequences, one may use blastn with the "BLAST 2 Sequences" tool Version 2Ø12 (April-21-2000) set at default parameters. Such default parameters may be, for example:
Matrix: BLOSUM62 Reyvard for match: 1 Penalty for mismatch: -2 Open Gap: 5 arad Exterzsiotz Gap: 2 pefaalties Gap x drop-off.' S0 Expect: 10 -Word Size: Il Filter: on.
Percent identity may be measured over the length of an entire defined sequence, for example, as defined by a particular SEQ 1D number, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined sequence, for instance, a fragment of at least 20, at least 30, at least 40, at least 50, at least 70, at least 100, or at least 200 contiguous nucleotides. Such lengths are exemplary only, and it is understood that any fragment length supported by the sequences shown herein, in the tables, figures, or Sequence Listing, may be used to describe a length over which percentage identity may be measured.
Nucleic acid sequences that do not show a high degree of identity may nevertheless encode similar amino acid sequences due to the degeneracy of the genetic code. It is understood that changes in a nucleic acid sequence can be made using this degeneracy to produce multiple nucleic acid sequences that all encode substantially the same protein.
The phrases "percent identity" and "% identity," as applied to polypeptide sequences, refer to the percentage of residue matches between at least two polypeptide sequences aligned using a standardized algorithm. Methods of polypeptide sequence alignment are well-known. Some alignment methods take into account conservative amino acid substitutions. Such conservative substitutions, explained in more detail above, generally preserve the charge and hydrophobicity at the site of substitution, thus preserving the structure (and therefore function) of the polypeptide.
Percent identity between polypeptide sequences may be determined using the default parameters of the CLUSTAL V algorithm as incorporated into the MEGALIGN
version 3.12e sequence alignment program (described and referenced above). For pairwise alignments of polypeptide sequences using CLUSTAL V, the default parameters are set as follows: Ktuple=1, gap penalty=3, window=5, and "diagonals saved"=5. The PAM250 matrix is selected as the default residue weight table. As with polynucleotide alignments, the percent identity is reported by CLUSTAL V as the "percent similarity" between aligned polypeptide sequence pairs.
Alternatively the NCBI BLAST software suite may be used. For example, for a pairwise~
comparison of two polypeptide sequences, one may use the "BLAST 2 Sequences"
tool Version 2Ø12 (April-21-2000) with blastp set at default parameters. Such default parameters may be, for example:
Matrix: BLOSUM62 Open Gap: 11 and Extensiofz Gap: 1 pefzaltr.'es Gap x drop-off SO
Expect: 10 Word Size: 3 Filter: on Percent identity may be measured over the length of an entire defined polypeptide sequence, for example, as defined by a particular SEQ ID number, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined polypeptide sequence, for instance, a fragment of at least 15, at least 20, at least 30, at least 40, at least 50, at least 70 or at least 150 contiguous residues. Such lengths are exemplary only, and it is understood that any fragment length supported by the sequences shown herein, in the tables, figures or Sequence Listing, may be used to describe a length over which percentage identity may be measured.
"Human artificial chromosomes" (HACs) are linear microchromosomes which may contain DNA sequences of about 6 kb to 10 Mb in size and which contain all of the elements required for chromosome replication, segregation and maintenance.
The term "humanized antibody" refers to an antibody molecule in which the amino acid sequence in the non-antigen binding regions has been altered so that the antibody more closely resembles a human antibody, and still retains its original binding ability.
"Hybridization" refers to the process by which a polynucleotide strand anneals with a complementary strand through base pairing under defined hybridization conditions. Specific hybridization is an indication that two nucleic acid sequences share a high degree of complementarity.
Specific hybridization complexes form under permissive annealing conditions and remain hybridized after the "washing" step(s). The washing steps) is particularly important in determining the stringency of the hybridization process, with more stringent conditions allowing less non-specific binding, i.e., binding between pairs of nucleic acid strands that are not perfectly matched. Permissive conditions for annealing of nucleic acid sequences are routinely determinable by one of ordinary skill in the art and may be consistent among hybridization experiments, whereas wash conditions may be varied among experiments to achieve the desired stringency, and therefore hybridization specificity.
Permissive annealing conditions occur, for example, at 68°C in the presence of about 6 x SSC, about 1 % (w/v) SDS, and about 100 ~g/ml sheared, denatured salmon sperm DNA.
Generally, stringency of hybridization is expressed, in part, with reference to the temperature under which the wash step is carried out. Such wash temperatures are typically selected to be about 5°C to 20°C lower than the thermal melting point (T~ for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength and pI-~ at which 50% of the target sequence hybridizes to a perfectly matched probe. An equation for calculating Tm and conditions for nucleic acid hybridization are well known and can be found in Sambrook, J. et al. (1989) Molecular Cloning: A Laborator'~ Manual, 2nd ed., vol. 1-3, Cold Spring Harbor Press, Plainview NY;
specifically see volume 2, chapter 9.
High stringency conditions for hybridization between polynucleotides of the present invention include wash conditions of 68°C in the presence of about 0.2 x SSC and about 0.1% SDS, for 1 hour.
Alternatively, temperatures of about 65°C, 60°C, 55°C, or 42°C may be used. SSC concentration may be varied from about 0.1 to 2 x SSC, with SDS being present at about 0.1 %.
Typically, blocking reagents are used to block non-specific hybridization. Such blocking reagents include, for instance, sheared and denatured salmon sperm DNA at about 100-200 ~glml. Organic solvent, such as formamide at a concentration of about 35-50% v/v, may also be used under particular circumstances, such as for RNA:DNA hybridizations. Useful variations on these wash conditions will be readily apparent to those of ordinary skill in the art. Hybridization, particularly under high stringency conditions, may be suggestive of evolutionary similarity between the nucleotides. Such similarity is strongly indicative of a similar role for the nucleotides and their encoded polypeptides.
The term "hybridization complex" refers to a complex formed between two nucleic acid sequences by virtue of the formation of hydrogen bonds between complementary bases. A
hybridization complex may be formed in solution (e.g., Cot or Rot analysis) or formed between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized on a solid support (e.g., paper, membranes, filters, chips, pins or glass slides, or any other appropriate substrate to which cells or their nucleic acids have been fixed).
The words "insertion" and "addition" refer to changes in an amino acid or nucleotide sequence resulting in the addition of one or more amino acid residues or nucleotides, respectively.
"Immune response" can refer to conditions associated with inflammation, trauma, immune disorders, or infectious or genetic disease, etc. These conditions can be characterized by expression of various factors, e.g., cytokines, chemokines, and other signaling molecules, which may affect cellular and systemic defense systems.
An "immunogenic fragment" is a polypeptide or oligopeptide fragment of ATRS
which is capable of eliciting an immune response when introduced into a living organism, for example, a mammal. The term "immunogenic fragment" also includes any polypeptide or oligopeptide fragment of ATRS which is useful in any of the antibody production methods disclosed herein or known in the art.
. The term "microarray" refers to an arrangement of a plurality of polynucleotides, polypeptides, or other chemical compounds on a substrate.
The terms "element" and "array element" refer to a polynucleotide, polypeptide, or other chemical compound having a unique and defined position on a microarray. ' The term "modulate" refers to a change in the activity of ATRS. For example, modulation may cause an increase or a decrease in protein activity, binding characteristics, or any other biological, functional, or immunological properties of ATRS.
The phrases "nucleic acid" and "nucleic acid sequence" refer to a nucleotide, oligonucleotide, polynucleotide, or any fragment thereof. These phrases also refer to DNA or RNA of genomic or synthetic origin which may be single-stranded or double-stranded and may represent the sense or the antisense strand, to peptide nucleic acid (PNA), or to any DNA-like or RNA-like material.
"Operably linked" refers to the situation in which a first nucleic acid sequence is placed in a functional relationship with a second nucleic acid sequence. For instance, a promoter is operably linked to a coding sequence if the promoter affects the transcription or expression of the coding sequence. Operably linked DNA sequences may be in close proximity or contiguous and, where necessary to join two protein coding regions, in the same reading frame.
"Peptide nucleic acid" (PNA) refers to an antisense molecule or anti-gene agent which comprises an oligonucleotide of at least about 5 nucleotides in length linked to a peptide backbone of amino acid residues ending in lysine. The terminal lysine confers solubility to the composition. PNAs preferentially bind complementary single stranded DNA or RNA and stop transcript elongation, and may be pegylated to extend their lifespan in the cell.
"Post-translational modification" of an ATRS may involve lipidation, glycosylation, phosphorylation, acetylation, racemization, proteolytic cleavage, and other modifications known in the art. These processes may occur synthetically or biochemically. Biochemical modifications will vary by cell type depending on the enzymatic milieu of ATRS.
"Probe" refers to nucleic acid sequences encoding ATRS, their complements, ~or fragments thereof, which are used to detect identical, allelic or related nucleic acid sequences. Probes are isolated oligonucleotides or polynucleotides attached to a detectable label or reporter molecule.
Typical labels include radioactive isotopes, ligands, chemiluminescent agents, and enzymes. "Primers"
are short nucleic acids, usually DNA oligonucleotides, which may be annealed to a target polynucleotide by complementary base-pairing. The primer may then be extended along the target DNA strand by a DNA polymerase enzyme. Primer pairs can be used for amplification (and identification) of a nucleic acid sequence, e.g., by the polymerase chain reaction (PCR).
Probes and primers as used in the present invention typically comprise at least 15 contiguous nucleotides of a known sequence. In order to enhance specificity, longer probes and primers may also be employed, such as probes and primers that comprise at least 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, or at least 150 consecutive nucleotides of the disclosed nucleic acid sequences. Probes and primers may be considerably longer than these examples, and it is understood that any length supported by the specification, including the tables, figures, and Sequence Listing, may be used.
Methods for preparing and using probes and primers are described in the references, for example Sambrook, J. et al. (1989) Molecular Cloning: A Laboratory Manual, 2nd ed., vol. 1-3,.Cold Spring Harbor Press, Plainview NY; Ausubel, F.M. et al. (1987) Current Protocols in Molecular Biolo~y, Greene Publ. Assoc. & Wiley-Intersciences, New York NY; Innis, M. et al. (1990) PCR
Protocols, A Guide to Methods and Applications, Academic Press, San Diego CA.
PCR primer pairs can be derived from a known sequence, fox example, by using computer programs intended for. that purpose such as Primer (Version 0.5, 1991, Whitehead Institute for Biomedical Research, Cambridge l0 MA).
Oligonucleotides for use as primers are selected using software known in the art for such purpose. For example, OLIGO 4.06 software is useful for the selection of PCR
primer pairs of up to 100 nucleotides each, and for the analysis of oligonucleotides and larger polynucleotides of up to 5,000 nucleotides from an input polynucleotide sequence of up to 32 kilobases.
Similar primer selection programs have incorporated additional features for expanded capabilities. Fox example, the PrimOU
primer selection program (available to the public from the Genome Center at University of Texas South West Medical Center, Dallas TX) is capable of choosing specific primers from megabase sequences and is thus useful for designing primers on a genome-wide scope. The Primer3 primer selection program (available to the public from the Whitehead InstitutelMIT
Center for Genome Research, Cambridge MA) allows the user to input a "mispriming library," in which sequences to avoid as primer binding sites are user-specified. Primer3 is useful, in particular, for the selection of oligonucleotides for microarrays. (The source code for the latter two primer selection programs may also be obtained from their respective sources and modified to meet the user's specific needs.) The PrimeGen program (available to the public from the UK Human Genome Mapping Project Resource Centre, Cambridge UK) designs primers based on multiple sequence alignments, thereby allowing selection of primers that hybridize to either the most conserved or least conserved regions of aligned nucleic acid sequences. Hence, this program is useful for identification of both unique and conserved oligonucleotides and polynucleotide fragments. The oligonucleotides and polynucleotide fragments identified by any of the above selection methods are useful in hybridization technologies, for example, as PCR or sequencing primers, microarray elements, or specific probes to identify fully or partially complementary polynucleotides in a sample of nucleic acids. Methods of oligonucleotide selection are not limited to those described above.
A "recombinant nucleic acid" is a sequence that is not naturally occurnng or has a sequence that is made by an artificial combination of two or more otherwise separated segments of sequence.
This artificial combination is often accomplished by chemical synthesis or, more commonly, by the artificial manipulation of isolated segments of nucleic acids, e.g., by genetic engineering techniques such as those described in Sambrook, supra. The term recombinant includes nucleic acids that have been altered solely by addition, substitution, or deletion of a portion of the nucleic acid. Frequently, a recombinant nucleic acid may include a nucleic acid sequence operably linked to a promoter sequence.
Such a recombinant nucleic acid may be part of a vector that is used, for example, to transform a cell.
Alternatively, such recombinant nucleic acids may be part of a viral vector, e.g., based on a vaccinia virus, that could be use to vaccinate a mammal wherein the recombinant nucleic acid is expressed, inducing a protective immunological response in the mammal.
A "regulatory element" refers to a nucleic acid sequence usually derived from untranslated regions of a gene and includes enhancers, promoters, introns, and 5' and 3' untranslated regions (UTRs). Regulatory elements interact with host or viral proteins which control transcription, translation, or RNA stability.
"Reporter molecules" are chemical or biochemical moieties used for labeling a nucleic acid, amino acid, or antibody. Reporter molecules include radionuclides; enzymes;
fluorescent, chemiluminescent, or chromogenic agents; substrates; cofactors; inhibitors;
magnetic particles; and other moieties known in the art.
An "RNA equivalent," in reference to a DNA sequence, is composed of the same linear sequence of nucleotides as the reference DNA sequence with the exception that all occurrences of the nitrogenous base thymine are replaced with uracil, and the sugar backbone is composed of ribose instead of deoxyribose.
The term "sample" is used in its broadest sense. A sample suspected of containing ATRS, nucleic acids encoding ATRS, or fragments thereof may comprise a bodily fluid;
an extract from a cell, chromosome, organelle, or membrane isolated from a cell; a cell; genomic DNA, RNA, or cDNA, in solution or bound to a substrate; a tissue; a tissue print; etc.
The terms "specific binding" and "specifically binding" refer to that interaction between a protein or peptide and an agonist, an antibody, an antagonist, a small molecule, or any natural or synthetic binding composition. The interaction is dependent upon the presence of a particular structure of the protein, e.g., the antigenic determinant or epitope, recognized by the binding molecule. For example, if an antibody is specific for epitope "A," the presence of a polypeptide comprising the epitope A, or the presence of free unlabeled A, in a reaction containing free labeled A and the antibody will reduce the amount of labeled A that binds to the antibody.

The term ''substantially purified" refers to nucleic acid or amino acid sequences that are removed from their natural environment and are isolated or separated, and are at least 60% free, preferably at least 75% free, and most preferably at least 90% free from other components with which they are naturally associated.
A "substitution" refers to the replacement of one or more amino acid residues or nucleotides by different amino acid'residues or nucleotides, respectively.
"Substrate" refers to any suitable rigid or semi-rigid support including membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, tubing, plates, polymers, microparticles and capillaries. The substrate can have a variety of surface forms, such as wells, trenches, pins, channels and pores, to which polynucleotides or polypeptides are bound.
A "transcript image" refers to the collective pattern of gene expression by a particular cell type or tissue under given conditions at a given time.
"Transformation" describes a process by which exogenous DNA is introduced into a recipient cell. Transformation may occur under natural or artificial conditions according to various methods well known in the art, and may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method for transformation is selected based on the type of host cell being transformed and may include, but is not limited to, bacteriophage or viral infection, electroporation, heat shock, lipofection, and particle bombardment.
The term "transformed cells" includes stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome, as well as transiently transformed cells which express the inserted DNA or RNA for limited periods of time.
A "transgenic organism," as used herein, is any organism, including but not limited to animals and plants, in which one or more of the cells of the organism contains 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 rather is directed to the introduction of a recombinant DNA
molecule. The transgenic organisms contemplated in accordance with the present invention include bacteria, cyanobacteria, fungi, plants and animals. The isolated DNA of the present invention can be introduced into the host by methods known in the art, for example infection, transfection, transformation or transconjugation.
Techniques for transfernng the DNA of the present invention into such organisms are widely known and provided in references such as Sambrook et al. (1989), su ra.

A "variant'' of a particular nucleic acid sequence is defined as a nucleic acid sequence having at least 40% sequence identity to the particular nucleic acid sequence over a certain length of one of the nucleic acid sequences using blastn with the "BLAST 2 Sequences" tool Version 2Ø9 (May-07-1999) set at default parameters. Such a pair of nucleic acids may show, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% or greater sequence identity over a certain defined length. A variant may be described as, for example, an "allelic" (as defined above), "splice," "species," or "polymorphic" variant. A
splice variant may have significant identity to a reference molecule, but will generally have a greater or lesser number of polynucleotides due to alternative splicing of exons during mRNA processing.
The corresponding polypeptide may possess additional functional domains or lack domains that are present in the reference molecule. Species variants are polynucleotide sequences that vary from one species to another. The resulting polypeptides will generally have significant amino acid identity relative to each other. A polymorphic variant is a variation in the polynucleotide sequence of a particular gene between individuals of a given species. Polymorphic variants also may encompass "single nucleotide polymorphisms" (SNPs) in which the polynucleotide sequence varies by one nucleotide base. The presence of SNPs may be indicative of, for example, a certain population, a disease state, or a propensity for a disease state.
A "variant" of a particular polypeptide sequence is defined as a polypeptide sequence having at least 40% sequence identity to the particular polypeptide sequence over a certain length of one of the polypeptide sequences using blastp with the "BLAST 2 Sequences" tool Version 2Ø9 (May-07-1999) set at default parameters. Such a pair of polypeptides may show, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%
or greater sequence identity over a certain defined length of one of the polypeptides.
THE INVENTION
The invention is based on the discovery of new human aminoacyl tRNA
synthetases (ATRS), the polynucleotides encoding ATRS, and the use of these compositions for the diagnosis, treatment, or prevention of cell proliferative and autoimmune/inflammatory disorders.
Table 1 summarizes the nomenclature for the full length polynucleotide and polypeptide sequences of the invention. Each polynucleotide and its corresponding polypeptide are correlated to a single Incyte project identification number (Incyte Project ID). Each polypeptide sequence is denoted by both a polypeptide sequence identification number (Polypeptide SEQ ID NO:) and an Incyte polypeptide sequence number (Incyte Polypeptide ID) as shown. Each polynucleotide sequence is denoted by both a polynucleotide sequence identification number (Polynucleotide SEQ ID NO:) and an Incyte polynucleotide consensus sequence number (Incyte Polynucleotide ID) as shown.
Table 2 shows sequences with homology to the polypeptides of the invention as identified by BLAST analysis against the GenBank protein (genpept) database. Columns 1 and 2 show the polypeptide sequence identification number (Polypeptide SEQ ID NO:) and the corresponding Incyte polypeptide sequence number (Incyte Polypeptide ID) for polypeptides of the invention. Column 3 shows the GenBank identification number (Genbank ID NO:) of the nearest GenBank homolog.
Column 4 shows the probability score for the match between each polypeptide and its GenBank homolog. Column 5 shows the annotation of the GenB ank homolog along with relevant citations where applicable, all of which are expressly incorporated by reference herein.
Table 3 shows various structural features of the polypeptides of the invention. Columns 1 and 2 show the polypeptide sequence identification number (SEQ ID NO:) and the corresponding Incyte polypeptide sequence number (Incyte Polypeptide ID) for each polypeptide of the invention. Column 3 shows the number of amino acid residues in each polypeptide. Column 4 shows potential phosphorylation sites, and column 5 shows potential glycosylation sites, as determined by the MOTIFS
program of the GCG sequence analysis software package (Genetics Computer Group, Madison WI).
Column 6 shows amino acid residues comprising signature sequences, domains, and motifs. Column 7 shows analytical methods for protein sti-ucture/function analysis and in some cases, searchable databases to which the analytical methods were applied.
Together, Tables 2 and 3 summarize the properties of polypeptides of the invention, and these properties establish that the claimed polypeptides are aminoacyl tRNA
synthetases. For example, SEQ ID N0:1 is 41 % identical from amino acid residues 51 to 503 to Pyrococcus abyssi cysteinyl-tRNA synthetase (GenBank ID 85458823) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 1.4e-81, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ >D NO:l also contains a tRNA synthetase class I (C) domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) Data from BLIMPS analyses provide further corroborative evidence that SEQ ID N0:1 is a cysteinyl-tRNA synthetase. In an alternate example, SEQ ID NO:2 is 46%
identical to Synechocystis s~ asparaginyl-tRNA synthetase (GenBank ID 81001357) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 7.8e-104, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance.
SEQ ID N0:2 also contains a tRNA synthetase class II (D, K and N) domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM
database of conserved protein family domains. (See Table 3.) Data from BLIMPS, MOTIFS, and PROFILESCAN analyses provide further corroborative evidence that SEQ ID N0:2 is an asparaginyl-tRNA synthetase. In a further example, SEQ ID N0:4 is 39%
identical to Bacillus caldotenax tyrosyl-tRNA synthetase (GenBank ID 8143793) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 2.3e-72, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ
ID N0:4 also contains an tyrosyl-tRNA synthetase domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) Data from BLIMPS, MOTIFS, and PROFILESCAN
analyses provide further corroborative evidence that SEQ ID N0:4 is a tRNA synthetase.
SEQ ID N0:3 was analyzed and annotated in a similar manner. The algorithms and parameters for the analysis of SEQ
ID NO:1-4 are described in Table 7.
As shown in Table 4, the full length polynucleotide sequences of the present invention were assembled using cDNA sequences or coding (exon) sequences derived from genomic DNA, or any combination of these two types of sequences. Columns 1 and 2 list the polynucleotide sequence identification number (Polynucleotide SEQ ID NO:) and the corresponding Incyte polynucleotide consensus sequence number (Incyte Polynucleotide ID) for each polynucleotide of the invention.
Column 3 shows the length of each polynucleotide sequence in basepairs. Column 4 lists fragments of the polynucleotide sequences which are useful, for example, in hybridization or amplification technologies that identify SEQ ID N0:5-8 or that distinguish between SEQ )D
N0:5-8 and related polynucleotide sequences. Column 5 shows identification numbers corresponding to cDNA
sequences, coding sequences (exons) predicted from genomic DNA, and/or sequence assemblages comprised of both cDNA and genomic DNA. These sequences were used to assemble the full length polynucleotide sequences of the invention. Columns 6 and 7 of Table 4 show the nucleotide start (5') and stop (3') positions of the cDNA and/or genomic sequences in column 5 relative to their respective full length sequences.
The identification numbers in Column 5 of Table 4 may refer specifically, for example, to Incyte cDNAs along with their corresponding cDNA libraries. For example, 2700694F6 is the identification number of an Incyte cDNA sequence, and OVARTUT10 is the cDNA
library from which it is derived. Alternatively, the identification numbers in column 5 may refer to GenBank cDNAs or ESTs (e.g., 86451182) which contributed to the assembly of the full length polynucleotide sequences. Incyte cDNAs for which cDNA libraries are not indicated were derived from pooled cDNA libraries (e.8., 70997854V1). Alternatively, the identification numbers in column 5 may refer to coding regions predicted by Genscan analysis of genomic DNA. The Genscan-predicted coding sequences may have been edited prior to assembly. (See Example IV.) Alternatively, the identification numbers in column 5 may refer to assemblages of both cDNA and Genscan-predicted exons brought together by an "exon stitching" algorithm. (See Example V.) Alternatively, the identification numbers in column 5 may refer to assemblages of both cDNA and Genscan-predicted exons brought together by an "exon-stretching" algorithm. (See Example V.) In some cases, Incyte cDNA coverage redundant with the sequence coverage shown in column 5 was obtained to confirm the final consensus polynucleotide sequence, but the relevant Incyte cDNA
identification numbers are not shown.
Table 5 shows the representative cDNA libraries for those full length polynucleotide sequences which were assembled using Incyte cDNA sequences. The representative cDNA library is the Incyte cDNA library which is most frequently represented by the Incyte cDNA sequences which were used to assemble and confirm the above polynucleotide sequences.
The tissues and vectors which were used to construct the cDNA libraries shown in Table 5 are describedin Table 6.
The invention also encompasses ATRS variants. A preferred ATRS variant is one which has at least about 80%, or alternatively at least about 90%, or even at least about 95% amino acid sequence identity to the ATRS amino acid sequence, and which contains at least one functional or structural characteristic of ATRS.
The invention also encompasses polynucleotides which encode ATRS. In a particular embodiment, the invention encompasses a polynucleotide sequence comprising a sequence selected from the group consisting of SEQ D? N0:5-8, which encodes ATRS. The polynucleotide sequences of SEQ ID NO:S-8, as presented in the Sequence Listing, embrace the equivalent RNA sequences, wherein occurrences of the nitrogenous base thymine are replaced with uracil, and the sugar backbone is composed of ribose instead of deoxyribose.
The invention also encompasses a variant of a polynucleotide sequence encoding ATRS. In particular, such a variant polynucleotide sequence will have at least about 80%, or alternatively at least about 90%, or even at least about 95% polynucleotide sequence identity to the polynucleotide sequence encoding ATRS. A particular aspect of the invention encompasses a variant of a polynucleotide sequence comprising a sequence selected from the group consisting of SEQ ID NO:S-8 which has at least about 70%, or alternatively at least about 85%, or even at least about 95%

polynucleotide sequence identity to a nucleic acid sequence selected from the group consisting of SEQ
ID N0:5-8. Any one of the polynucleotide variants described above can encode an amino acid sequence which contains at least one functional or structural characteristic of ATRS.
It will be appreciated by those skilled in the art that as a result of the degeneracy of the genetic code, a multitude of polynucleotide sequences encoding ATRS, some bearing minimal similarity to the polynucleotide sequences of any known and naturally occurring gene, may be produced. Thus, the invention contemplates each and every possible variation of polynucleotide sequence that could be made by selecting combinations based on possible codon choices. These combinations are made in accordance with the standard triplet genetic code as applied to the polynucleotide sequence of naturally occurring ATRS, and all such variations are to be considered as being specifically disclosed.
Although nucleotide sequences which encode ATRS and its variants are generally capable of hybridizing to the nucleotide sequence of the naturally occurring ATRS under appropriately selected conditions of stringency, it may be advantageous to produce nucleotide sequences encoding ATRS or its derivatives possessing a substantially different codon usage, e.g., inclusion of non-naturally occurring codons. Codons may be selected to increase the rate at which expression of the peptide occurs in a particular prokaryotic or eukaryotic host in accordance with the frequency with which particular codons are utilized by the host. Other reasons for substantially altering the nucleotide sequence encoding ATRS and its derivatives without altering the encoded amino acid sequences include the production of RNA transcripts having more desirable properties, such as a greater half life, than transcripts produced from the naturally occurring sequence.
The invention also encompasses production of DNA sequences which encode ATRS
and ATRS derivatives, or fragments thereof, entirely by synthetic chemistry. After production, the synthetic sequence may be inserted into any of the many available expression vectors and cell systems using reagents well known in the art. Moreover, synthetic chemistry may be used to introduce mutations into a sequence encoding ATRS or any fragment thereof.
Also encompassed by the invention are polynucleotide sequences that are capable of hybridizing to the claimed polynucleotide sequences, and, in particular, to those shown in SEQ 1D
N0:5-8 and fragments thereof under various conditions of stringency. (See, e.g., Wahl, G.M. and S.L. Berger (1987) Methods Enzymol. 152:399-407; Kimmel, A.R. (1987) Methods Enzymol. 152:507 511.) Hybridization conditions, including annealing and wash conditions, are described in "Definitions."
Methods for DNA sequencing are well known in the art and may be used to practice any of the embodiments of the invention. The methods may employ such enzymes as the Klenow fragment of DNA polymerase I, SEQUENASE (US Biochemical, Cleveland OH), Taq polymerase (Applied Biosystems)> thermostable T7 polymerase (Amersham Pharmacia Biotech, Piscataway NJ), or combinations of polymerases and proofreading exonucleases such as those found in the ELONGASE
amplification system (Life Technologies, Gaithersburg MD). Preferably, sequence preparation is automated with machines such as the MICROLAB 2200 liquid transfer system (Hamilton, Reno N~, PTC200 thermal cycler (MJ Research, Watertown MA) and ABI CATALYST 800 thermal cycler (Applied Biosystems). Sequencing is then carried out using either the ABI 373 or 377 DNA
sequencing system (Applied Biosystems), the MEGABACE 1000 DNA sequencing system (Molecular Dynamics, Sunnyvale CA), or other systems known in the art. The resulting sequences are analyzed using a variety of algorithms which are well known in the an.
(See, e.g., Ausubel, F.M.
(1997) Short Protocols in Molecular Biology, John Wiley & Sons, New York NY, unit 7.7; Meyers, R.A. (1995) Molecular Biolo~y and Biotechnology, Wiley VCH, New York NY, pp.
856-853.) The nucleic acid sequences encoding ATRS may be extended utilizing a partial nucleotide sequence and employing various PCR-based methods known in the art to detect upstream sequences, such as promoters and regulatory elements. For example, one method which may be employed, restriction-site PCR, uses universal and nested primers to amplify unknown sequence from genomic DNA within a cloning vector. (See, e.g., Sarkar, G. (1993) PCR Methods Applic.
2:318-322.) Another method, inverse PCR, uses primers that extend in divergent directions to amplify unknown sequence from a circularized template. The template is derived from restriction fragments comprising a known genomic locus and surrounding sequences. (See, e.g., Triglia, T. et al. (1988) Nucleic Acids Res. 16:8186.) A third method, capture PCR, involves PCR amplification of DNA
fragments adjacent to known sequences in human and yeast artificial chromosome DNA. (See, e.g., Lagerstrom, M. et al. (1991) PCR Methods Applic. 1:111-119.) In this method, multiple restriction enzyme digestions and ligations may be used to insert an engineered double-stranded sequence into a region of unknown sequence before performing PCR. Other methods which may be used to retrieve unknown sequences are known in the art. (See, e.g., Parker, J.D. et al. (1991) Nucleic Acids Res. 19:3055-3060).
Additionally, one may use PCR, nested primers, and PROMOTERFINDER libraries (Clontech, Palo Alto CA) to walk genomic DNA. This procedure avoids the need to screen libraries and is useful in finding intronlexon junctions. For all PCR-based methods, primers rnay be designed using commercially available softwaxe, such as OLIGO 4.06 primer analysis software (National Biosciences, Plymouth MN) or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the template at temperatures of about 68°C to 72°C.

When screening for full length cDNAs, it is preferable to use libraries that have been size-selected to include larger cDNAs. In addition, random-primed libraries, which often include sequences containing the 5' regions of genes, are preferable for situations in which an oligo d(T) library does not yield a full-length cDNA. Genomic libraries may be useful for extension of sequence into 5' non-transcribed regulatory regions.
Capillary electrophoresis systems which are commercially available may be used to analyze the size or confirm the nucleotide sequence of sequencing or PCR products. In particular, capillary sequencing may employ flowable polymers for electrophoretic separation, four different nucleotide-specific, laser-stimulated fluorescent dyes, and a charge coupled device camera for detection of the emitted wavelengths. Outputllight intensity may be converted to electrical signal using appropriate software (e.g., GENOTYPER and SEQUENCE NAVIGATOR, Applied Biosystems), and the entire process from loading of samples to computer analysis and electronic data display may be computer controlled. Capillary electrophoresis is especially preferable for sequencing small DNA fragments which may be present in limited amounts in a particular sample.
In another embodiment of the invention, polynucleotide sequences or fragments thereof which encode ATRS may be cloned in recombinant DNA molecules that direct expression of ATRS, or fragments or functional equivalents thereof, in appropriate host cells. Due to the inherent.degeneracy of the genetic code, other DNA sequences which encode substantially the same or a functionally equivalent amino acid sequence may be produced and used to express ATRS.
The nucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter ATRS-encoding sequences for a variety of purposes including, but not limited to, modification of the cloning, processing, and/or expression of the gene product. DNA
shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides may be used to engineer the nucleotide sequences. For example, oligonucleotide mediated site-directed mutagenesis may be used to introduce mutations that create new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, and so forth.
The nucleotides of the present invention may be subjected to DNA shuffling techniques such as MOLECULARBREEDING (Maxygen Inc., Santa Clara CA; described in U.S. Patent Number 5,837,458; Chang, C.-C. et al. (1999) Nat. Biotechnol. 17:793-797; Christians, F.C. et al. (1999) Nat.
Biotechnol. 17:259-264; and Crameri, A. et al. (1996) Nat. Biotechnol. 14:315-319) to alter or improve the biological properties of ATRS, such as its biological or enzymatic activity or its ability to bind to other molecules or compounds. DNA shuffling is a process by which a library of gene variants is produced using PCR-mediated recombination of gene fragments. The library is then subjected to selection or screening procedures that identify those gene variants with the desired properties. These preferred variants may then be pooled and further subjected to recursive rounds of DNA shuffling and selection/screening. Thus, genetic diversity is created through "artificial"
breeding and rapid molecular evolution. For example, fragments of a single gene containing random point mutations may be recombined, screened, and then reshuffled until the desired properties are optimized. Alternatively, fragments of a given gene may be recombined with fragments of homologous genes in the same gene family, either from the same or different species, thereby maximizing the genetic diversity of multiple naturally occurnng genes in a directed and controllable manner.
In another embodiment, sequences encoding ATRS may be synthesized, in whole or in part, using chemical methods well known in the art. (See, e.g., Caruthers, M.H. et al. (1980) Nucleic Acids Symp. Ser. 7:215-223; and Horn, T. et al. (1980) Nucleic Acids Symp. Ser.
7:225-232.) Alternatively, ATRS itself or a fragment thereof may be synthesized using chemical methods.
For example, peptide synthesis can be performed using various solution-phase or solid-phase techniques. (See, e.g., Creighton, T. (1984) Proteins, Structures and Molecular Properties, WH
Freeman, New York NY, pp.
55-60; and Roberge, J.Y. et al. (1995) Science 269:202-204.) Automated synthesis may be achieved using the ABI 431A peptide synthesizer (Applied Biosystems). Additionally, the amino acid sequence of ATRS, or any part thereof, may be altered during direct synthesis andlor combined with sequences from other proteins, or any part thereof, to produce a variant polypeptide or a polypeptide having a sequence of a naturally occurring polypeptide.
The peptide may be substantially purified by preparative high performance liquid chromatography. (See, e.g., Chiez, R.M. and F.Z. Regnier (1990) Methods Enzymol. 182:392-421.) The composition of the synthetic peptides may be confirmed by amino acid analysis or by sequencing.
(See, e.g., Creighton, supra, pp. 28-53.) In order to express a biologically active ATRS, the nucleotide sequences encoding ATRS or derivatives thereof may be inserted into an appropriate expression vector, i.e., a vector which contains the necessary elements for transcriptional and translational control of the inserted coding sequence in a suitable host. These elements include regulatory sequences, such as enhancers, constitutive and ~inducible promoters, and 5' and 3' untranslated regions in the vector and in polynucleotide sequences encoding ATRS. Such elements may vary in their strength and specificity.
Specific initiation signals may also be used to achieve more e~cient translation of sequences encoding ATRS. Such signals include the ATG initiation codon and adjacent sequences, e.g. the I~ozak sequence. In cases where sequences encoding ATRS and its initiation codon and upstream regulatory sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a fragment thereof, is inserted, exogenous translational control signals including an in-frame ATG initiation codon should be provided by the vector. Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers appropriate for the particular host cell system used. (See, e.g., Scharf, D.
et al. (1994) Results Probl..
Cell Differ. 20:125-162.) Methods which are well known to those skilled in the art may be used to construct expression vectors containing sequences encoding ATRS and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. (See, e.g., Sambrook, J. et al. (1989) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Plainview NY, ch. 4, 8, and 16-17; Ausubel, F.M. et al. (1995) Current Protocols in Molecular Biolo~y, John Wiley & Sons, New York NY, ch. 9, 13, and 16.) A variety of expression vector/host systems may be utilized to contain and express sequences encoding ATRS. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors;
yeast transformed with yeast expression vectors; insect cell systems infected with viral expression vectors (e.g., baculovirus);
plant cell systems transformed with viral expression vectors (e.g., cauliflower mosaic virus, CaMV, or tobacco mosaic virus, TMV) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids); or animal cell systems. (See, e.g., Sambrook, supra; Ausubel, supra; Van Heeke, G. and S.M. Schuster (1989) J. Biol. Chem. 264:5503-5509; Engelhard, E.I~. et al. (1994) Proc.
Natl. Acad. Sci. USA
91:3224-3227; Sandig, V. et al. (1996) Hum. Gene Ther. 7:1937-1945; Takamatsu, N. (1987) EMBO
J. 6:307-31 l; The McGraw Hill Yearbook of Science and Technolo~y (1992) McGraw Hill, New York NY, pp. 191-196; Logan, J. and T. Shenk (1984) Proc. Natl. Acad. Sci. USA
81:3655-3659; and Harrington, J.J. et al. (1997) Nat. Genet. 15:345-355.) Expression vectors derived from retroviruses, adenoviruses, or herpes or vaccinia viruses, or from various bacterial plasmids, may be used for delivery of nucleotide sequences to the targeted organ, tissue, or cell population. (See, e.g., Di Nicola, M. et al. (1998) Cancer Gen. Ther. 5(6):350-356; Yu, M. et al. (1993) Proc.
Natl. Acad. Sci. USA
90(13):6340-6344; Buller, R.M. et al. (1985) Nature 317(6040):813-815;
McGregor, D.P. et al. (1994) Mol. Immunol. 31(3):219-226; and Verma, LM. and N. Somia (1997) Nature 389:239-242.) The invention is not limited by the host cell employed.
Iri bacterial systems, a number of cloning and expression vectors may be selected depending upon the use intended for polynucleotide sequences encoding ATRS. For example, routine cloning, subcloning, and propagation of polynucleotide sequences encoding ATRS can be achieved using a multifunctional E. coli vector such as PBI,UESCRIPT (Stratagene, La Jolla CA) or PSPORT1 plasmid (Life Technologies). Ligation of sequences encoding ATRS into the vector's multiple cloning site disrupts the IacZ gene, allowing a colorimetric screening procedure for identification of transformed bacteria containing recombinant molecules. In addition, these vectors may be useful for in vitro transcription, dideoxy sequencing, single strand rescue with helper phage, and creation of nested deletions in the cloned sequence. (See, e.g., Van Heeke, G. and S.M.
Schuster (1989) J. Biol.
Chem. 264:5503-5509.) When large quantities of ATRS are needed, e.g. for the production of antibodies, vectors which direct high level expression of ATRS maybe used. For example, vectors containing the strong, inducible SP6 or T7 bacteriophage promoter may be used.
Yeast expression systems may be used for production of ATRS. A number of vectors containing constitutive or inducible promoters, such as alpha factor, alcohol oxidase, and PGH
promoters, may be used in the yeast Saccharomvces cerevisiae or Pichia pastoris. In addition, such vectors direct either the secretion or intracellular retention of expressed proteins and enable integration of foreign sequences into the host genome for stable propagation. (See, e.g., Ausubel, 1995, sera;
Bitter, G.A. et al. (1987) Methods Enzymol. 153:516-544; and Scorer, C.A. et al. (1994) Bio/Technology 12:181-184.) Plant systems may also be used for expression of ATRS. Transcription of sequences encoding ATRS may be driven by viral promoters, e.g., the 35S and 19S
promoters of CaMV used alone or in combination with the omega leader sequence from TMV (Takamatsu, N.
(1987) EMBO J.
6:307-311). Alternatively, plant promoters such as the small subunit of RUBISCO or heat shock promoters may be used. (See, e.g., Coruzzi, G. et al. (1984) EMBO J. 3:1671-1680; Brogue, R. et al.
(1984) Science 224:838-843; and Winter, J. et al. (1991) Results Probl. Cell Differ. 17:85-105.) These constructs can be introduced into plant Bells by direct DNA transformation or pathogen-mediated transfection. (See, e.g., The McGraw Hill Yearbook of Science and Technolo~y (1992) McGraw Hill, New York NY, pp. 191-196.) In mammalian cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, sequences encoding ATRS
may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential E1 or E3 region of the viral genome may be used to obtain infective virus which expresses ATRS in host cells. (See, e.g., Logan, J. and T. Shenk (1984) Proc.
Natl. Acad. Sci. USA 81:3655-3659.) In addition, transcription enhancers, such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells. SV40 or EBV-based vectors may also be used for high-level protein expression.

Human artificial chromosomes (HACs) may also be employed to deliver larger fragments of DNA than can be contained in and expressed from a plasmid. HACs of about 6 kb to 10 Mb are constructed and delivered via conventional delivery methods (liposomes, polycationic amino polymers, or vesicles) for therapeutic purposes. (See, e.g., Harrington, J.J. et al.
(1997) Nat. Genet. 15:345-355.) For long term production of recombinant proteins in mammalian systems, stable expression of ATRS in cell lines is preferred. For example, sequences encoding ATRS can be transformed into cell lines using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the l0 introduction of the vector, cells may be allowed to grow for about 1 to 2 days in enriched media before being switched to selective media. The purpose of the selectable marker is to confer resistance to a selective agent, and its presence allows growth and recovery of cells which successfully express the introduced sequences. Resistant clones of stably transformed cells may be propagated using tissue culture techniques appropriate to the cell type.
Any number of selection systems may be used to recover transformed cell lines.
These include, but are not limited to, the herpes simplex virus thymidine kinase and adenine phosphoribosyltransferase genes, for use in tl~ and apr cells, respectively.
(See, e.g., Wigler, M. et al. (1977) Cell 11:223-232; Lowy, I. et al. (1980) Cell 22:817-823.) Also, antimetabolite, antibiotic, or herbicide resistance can be used as the basis for selection. For example, dhfr confers resistance to methotrexate; fzeo confers resistance to the aminoglycosides neomycin and G-418; and als and pat confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively. (See, e.g., Wigler, M. et al. (1980) Proc. Natl. Acad. Sci. USA 77:3567-3570; Colbere-Garapin, F. et al. (1981) J. Mol. Biol. 150:1-14.) Additional selectable genes have been described, e.g., trpB and hisD, which alter cellular requirements for metabolites. (See, e.g.; Hartman, S.C. and R.C. Mulligan (1988) Proc.
Natl. Acad. Sci. USA 85:8047-8051.) Visible markers, e.g., anthocyanins, green fluorescent proteins (GFP; Clontech),13 glucuronidase and its substrate 13-glucuronide, or luciferase and its substrate luciferin may be used. These markers can be used not only to identify transformants, but also to quantify the amount of transient or stable protein expression attributable to a specific vector system.
(See, e.g., Rhodes, C.A. (1995) Methods Mol. Biol. 55:121-131.) Although the presence/absence of marker gene expression suggests that the gene of interest is also present, the presence and expression of the gene may need to be confirmed. For example, if the sequence encoding ATRS is inserted within a marker gene sequence, transformed cells containing sequences encoding ATRS can be identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a sequence encoding ATRS under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well.
In general, host cells that contain the nucleic acid sequence encoding ATRS
and that express ATRS may be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations, PCR
amplification, and protein bioassay or immunoassay techniques which include membrane, solution, or chip based technologies for the detection and/or quantification of nucleic acid or protein sequences.
Immunological methods for detecting and measuring the expression of ATRS using either l0 specific polyclonal or monoclonal antibodies are known in the art. Examples of such techniques include enzyme-linked immunosorbent assays (ELISAs), radioimmunoassays (RIAs), and fluorescence activated cell sorting (FACS). A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on ATRS is preferred, but a competitive binding assay may be employed. These and other assays are well known in the art. (See, e.g., Hampton, R. et al. (1990) Serological Methods, a Laboratom Manual, APS
Press, St. Paul MN, Sect. IV; Coligan, J.E. et al. (1997) Current Protocols in Immunolo~y, Greene Pub. Associates and Wiley-Interscience, New York NY; and Pound, J.D. (1998) Immunochemical Protocols, Humana Press, Totowa NJ.) A wide variety of labels and conjugation techniques are known by those skilled in the art and may be used in various nucleic acid and amino acid assays. Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides encoding ATRS
include oligolabeling, nick translation, end-labeling, or PCR amplification using a labeled nucleotide.
Alternatively, the sequences encoding ATRS, or any fragments thereof, may be cloned into a vector for the production of an mRNA probe. Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by addition of an appropriate RNA polymerise such as T7, T3, or SP6 and labeled nucleotides. These procedures may be conducted using a variety of commercially available kits, such as those provided by Amersham Pharmacia Biotech, Promega (Madison WI), and US Biochemical. Suitable reporter molecules or labels which may be used for ease of detection include radionuclides, enzymes, fluorescent, chemiluminescent, or chromogenic agents, as well as substrates, cofactors, inhibitors, magnetic particles, and the like.
Host cells transformed with nucleotide sequences encoding ATRS may be cultured under conditions suitable for the expression and recovery of the protein from cell culture. The protein produced by a transformed cell may be secreted or retained intracellularly depending on the sequence and/or the vector used. As will be understood by those of skill in the art, expression vectors containing polynucleotides which encode ATRS may be designed to contain signal sequences which direct secretion of ATRS through a prokaryotic or eukaryotic cell membrane.
In addition, a host cell strain may be chosen for its ability to modulate expression of the inserted sequences or to process the expressed protein in the desired fashion.
Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphoiylation, lipidation, and acylation. Post-translational processing which cleaves a "prepro" or "pro" form of the protein may also be used to specify protein targeting, folding, and/or activity. Different host cells which have specific cellular machinery and characteristic mechanisms for post-translational activities l0 (e.g., CHO, HeLa, MDCK, HEK293, and WI3~) are available from the American Type Culture Collection (ATCC, Manassas VA) and may be chosen to ensure the correct modification and processing. of the foreign protein.
In another embodiment of the invention, natural, modified, or recombinant nucleic acid sequences encoding ATRS may be ligated to a heterologous sequence resulting in translation of a fusion protein in any of the aforementioned host systems. For example, a chimeric ATRS protein containing a heterologous moiety that can be recognized by a commercially available antibody may facilitate the screening of peptide libraries for inhibitors of ATRS activity.
Heterologous protein and peptide moieties may also facilitate purification of fusion proteins using commercially available affinity matrices. Such moieties include, but are not limited to, glutathione S-transferase (GST), maltose binding protein (MBP), thioredoxin (Trx), calmodulin binding peptide (CBP), 6-His, FLAG, c-myc, and hemagglutmin (HA). GST, MBP, Trx, CBP, and 6-His enable purification of their cognate fusion proteins on immobilized glutathione, maltose, phenylarsine oxide, calmodulin, and metal-chelate resins, respectively. FLAG, c-myc, and hemagglutinin (HA) enable immunoaffinity purification of fusion proteins using commercially available monoclonal and polyclonal antibodies that specifically recognize these epitope tags. A fusion protein may also be engineered to contain a proteolytic cleavage site located between the ATRS encoding sequence and the heterologous protein sequence, so that ATRS
may be cleaved away from the heterologous moiety following purification.
Methods for fusion protein expression and purification are discussed in Ausubel (1995, su ra, ch. 10). A
variety of commercially available kits may also be used to facilitate expression and purification of fusion proteins.
In a further embodiment of the invention, synthesis of radiolabeled ATRS may be achieved in vitro using the TNT rabbit reticulocyte lysate or wheat germ extract system (Promega). These systems couple transcription and translation of protein-coding sequences operably associated with the T7, T3, or SP6 promoters. Translation takes place in the presence of a radiolabeled anuno acid precursor, for example, 35S-methionine.
ATRS of the present invention or fragments thereof may be used to screen for compounds that specifically bind to ATRS. At least one and up to a plurality of test compounds may be screened for specific binding to ATRS. Examples of test compounds include antibodies, oligonucleotides, proteins (e.g., receptors), or small molecules.
In one embodiment, the compound thus identified is closely related to the natural ligand of ATRS, e.g., a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a natural binding partner. (See, e.g., Coligan, J.E. et al. (1991) Current Protocols in Immunolo~y 1(2):
Chapter 5.) Similarly, the compound can be closely related to the natural receptor to which ATRS
binds, or to at least a fragment of the receptor, e.g., the ligand binding site. In either case, the compound can be rationally designed using known techniques. In one embodiment, screening for these compounds involves producing appropriate cells which express ATRS, either as a secreted protein or on the cell membrane. Preferred cells include cells from mammals, yeast, Drosophila, or E.
coli. Cells expressing ATRS or cell membrane fractions which contain ATRS are then contacted with a test compound and binding, stimulation, or inhibition of activity of either ATRS or the compound is analyzed.
An assay may simply test binding of a test compound to the polypeptide, wherein binding is detected by a fluorophore, radioisotope, enzyme conjugate, or other detectable label. For example, the assay may comprise the steps of combining at least one test compound with ATRS, either in solution or affixed to a solid support, and detecting the binding of ATRS to the compound. Alternatively, the assay may detect or measure binding of a test compound in the presence of a labeled competitor.
Additionally, the assay may be carried out using cell-free preparations, chemical libraries, or natural product mixtures, and the test compounds) may be free in solution or affixed to a solid support.
ATRS of the present invention or fragments thereof may be used to screen for compounds that modulate the activity of ATRS. Such compounds may include agonists, antagonists, or partial or inverse agonists. In one embodiment, an assay is performed under conditions permissive for ATRS
activity, wherein ATRS is combined with at least one test compound, and the activity of ATRS in the presence of a test compound is compared with the activity of ATRS in the absence of the test compound. A change in the activity of ATRS in the presence of the test compound is indicative of a compound that modulates the activity of ATRS. Alternatively, a test compound is combined with an in vitro or cell-free system comprising ATRS under conditions suitable for ATRS
activity, and the assay is performed. In either of these assays, a test compound which modulates the activity of ATRS may do so indirectly and need not come in direct contact with the test compound.
At least one and up to a plurality of test compounds may be screened.
In another embodiment, polynucleotides encoding ATRS or their mammalian homologs may be "knocked out" in an animal model system using homologous recombination in embryonic stem (ES) cells. Such techniques are well known in the art and are useful for the generation of animal models of human disease. (See, e.g., U.S. Patent Number 5,175,383 and U.S. Patent Number 5,767,337.) For example, mouse ES cells, such as the mouse 129/SvJ cell line, are derived from the early mouse embryo and grown in culture. The ES cells are transformed with a vector containing the gene of interest disrupted by a marker gene, e.g., the neomycin phosphotransferase gene (neo; Capecchi, M.R. (1989) Science 244:1288-1292). The vector integrates into the corresponding region of the host genome by homologous recombination. Alternatively, homologous recombination takes place using the Cre-loxP system to knockout a gene of interest in a tissue- or developmental stage-specific manner (Marth, J.D. (1996) Clin. Invest. 97:1999-2002; Wagner, K.U. et al. (1997) Nucleic Acids Res.
25:4323-4330). Transformed ES cells are identified and microinjected into mouse cell blastocysts such as those from the C57BL/6 mouse strain. The blastocysts are surgically transferred to pseudopregnant dams, and the resulting chimeric progeny are genotyped and bred to produce heterozygous or homozygous strains. Transgenic animals thus generated may be tested with potential therapeutic or toxic agents.
Polynucleotides encoding ATRS may also be manipulated in vitro in ES cells derived from human blastocysts. Human ES cells have the potential to differentiate into at least eight separate cell lineages including endoderm, mesoderm, and ectodermal cell types. These cell lineages differentiate into, for example, neural cells, hematopoietic lineages, and cardiomyocytes (Thomson, J.A. et al.
(1998) Science 282:1145-1147).
Polynucleotides encoding ATRS can also be used to create "knockin" humanized animals (pigs) or transgenic animals (mice or rats) to model human disease. With knockin technology, a region of a polynucleotide encoding ATRS is injected into animal ES cells, and the injected sequence integrates into the animal cell genome. Transformed cells are injected into blastulae, and the blastulae are implanted as described above. Transgenic progeny or inbred lines are studied and treated with potential pharmaceutical agents to obtain information on treatment of a human disease. Alternatively, a mammal inbred to overexpress ATRS, e.g., by secreting ATRS in its milk, may also serve as a convenient source of that protein (Janne, J. et al. (1998) Biotechnol. Annu.
Rev. 4:55-74).
THERAPEUTICS
Chemical and structural similarity, e.g., in the context of sequences and motifs, exists between regions of ATRS and aminoacyl tRNA synthetases. In addition, the expression of ATRS is closely associated with lung tumor tissue, with neonatal keratinocytes and lymph node tissue, and with disease states of the colon and prostate. Therefore, ATRS appears to play a role in cell proliferative and autoimmune/inflammatory disorders. In the treatment of disorders associated with increased ATRS
expression or activity, it is desirable to decrease the expression or activity of ATRS. In the treatment of disorders associated with decreased ATRS expression or activity, it is desirable to increase the expression or activity of ATRS.
Therefore, in one embodiment, ATRS or a fragment or derivative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of ATRS. Examples of such disorders include, but are not limited to, a cell proliferative disorder such as actinic keratosis, arteriosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCTD), myelofibrosis, paroxysmal nocturnal hemoglobinuria, polycythemia vera, psoriasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; and an autoimmune/inflammatory disorder such as acquired immunodeficiency syndrome (AIDS), Addison's disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), bronchitis, cholecystitis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes mellitus, emphysema, episodic lymphopenia with lymphocytotoxins, erythroblastosis fetalis, erythema nodosum, atrophic gastritis, glomerulonephritis, Goodpasture's syndrome, gout, Graves' disease, Hashimoto's thyroiditis, hypereosinophilia, irritable bowel syndrome, multiple sclerosis, myasthenia gravis, myocardial or pericardial inflammation, osteoarthritis, osteoporosis, pancreatitis, polymyositis, psoriasis, Reiter's syndrome, rheumatoid arthritis, scleroderma, Sjogren's syndrome, systemic anaphylaxis, systemic lupus erythematosus, systemic sclerosis, thrombocytopenic purpura, ulcerative colitis, uveitis, Werner syndrome, complications of cancer, hemodialysis, and extracorporeal circulation, viral, bacterial, fungal, parasitic, protozoal, and helininthic infections, and trauma.
In another embodiment, a vector capable of expressing ATRS or a fragment or derivative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of ATRS including, but not limited to, those described above.
In a further embodiment, a composition comprising a substantially purified ATRS in conjunction with a suitable pharmaceutical carrier may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of ATRS including, but not limited to, those provided above.
In still another embodiment, an agonist which modulates the activity of ATRS
may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of ATRS including, but not limited to, those listed above.
In a further embodiment, an antagonist of ATRS may be administered to a subject to treat or prevent a disorder associated with increased expression or activity of ATRS.
Examples of such disorders include, but are not limited to, those cell proliferative and autoimmune/inflammatory disorders described above. In one aspect, an antibody which specifically binds ATRS may be used directly as an antagonist or indirectly as a targeting or delivery mechanism for bringing a pharmaceutical agent to cells or tissues which express ATRS.
In an additional embodiment, a vector expressing the complement of the polynucleotide encoding ATRS may be administered to a subject to treat or prevent a disorder associated with.
increased expression or activity of ATRS including, but not limited to, those described above.
In other embodiments, any of the proteins, antagonists, antibodies, agonists, complementary sequences, or vectors of the invention may be administered in combination with other appropriate therapeutic agents. Selection of the appropriate agents for use in combination therapy may be made by one of ordinary skill in the art, according to conventional pharmaceutical principles. The combination of therapeutic agents may act synergistically to effect the treatment or prevention of the various disorders described above. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects.
An antagonist of ATRS may be produced using methods which are generally known in the art. In particular, purified ATRS may be used to produce antibodies or to screen libraries of pharmaceutical agents to identify those which specifically bind ATRS.
Antibodies to ATRS may also be generated using methods that are well known in the art. Such antibodies may include, but are not limited to, polyclonal, monoclonal, chimeric, and single chain antibodies, Fab fragments, and fragments produced by a Fab expression library. Neutralizing antibodies (i.e., those which inhibit dimer formation) are generally preferred for therapeutic use.
For the production of antibodies, various hosts including goats, rabbits, rats, mice, humans, and others may be immunized by injection with ATRS or with any fragment or oligopeptide thereof which has immunogenic properties. Depending on the host species, various adjuvants may be used to increase immunological response. Such adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, KLH, and dinitrophenol. Among adjuvants used in humans, BCG
(bacilli Calmette-Guerin) and Corynebacterium parvum are especially preferable.
It is preferred that the oligopeptides, peptides, or fragments used to induce antibodies to ATRS
have an amino acid sequence consisting of at least about 5 amino acids, and generally will consist of at least about 10 amino acids. It is also preferable that these oligopeptides, peptides, or fragments are identical to a portion of the amino acid sequence of the natural protein.
Short stretches of ATRS
amino acids may be fused with those of another protein, such as KLH, and antibodies to the chimeric molecule may be produced.
Monoclonal antibodies to ATRS may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique. (See, e.g., Kohler, G. et al. (1975) Nature 256:495-497; Kozbor, D.
et al. (1985) J.
Immunol. Methods 81:31-42; Cote, R.J. et al. (1983) Proc. Natl. Acad. Sci. USA
80:2026-2030; and Cole, S.P. et al. (1984) Mol. Cell Biol. 62:109-120.) In addition, techniques developed for the production of "chimeric antibodies,"
such as the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used. (See, e.g., Morrison, S.L. et al. (1984) Proc.
Natl. Acad. Sci. USA 81:6851-6855; Neuberger, M.S. et al. (1984) Nature 312:604-608; and Takeda, S. et al. (1985) Nature 314:452-454.) Alternatively, techniques described for the production of single chain antibodies may be adapted, using methods known in the art, to produce ATRS-specific single chain antibodies. Antibodies with related specificity, but of distinct idiotypic composition, may be generated by chain shuffling from random combinatorial immunoglobulin libraries. (See, e.g., Burton, D.R. (1991) Proc. Natl. Acad. Sci. USA 88:10134-10137.) Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature. (See, e.g., Orlandi, R. et al. (1989) Proc. Natl. Acad. Sci.
USA 86:3833-3837; Winter, G. et al. (1991) Nature 349:293-299.) Antibody fragments which contain specific binding sites for ATRS may also be generated.
For example, such fragments include, but are not limited to, F(ab')2 fragments produced by pepsin digestion of the antibody molecule and Fab fragments generated by reducing the disulfide bridges of the F(ab')2 fragments. Alternatively, Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity.
(See, e.g., Huse, W.D.

et al. (1989) Science 246:1275-1281.) Various immunoassays may be used for screening to identify antibodies having the desired specificity. Numerous protocols for competitive binding or immunoradiometric assays using either polyclonal or monoclonal antibodies with established specificities are well known in the art. Such immunoassays typically involve the measurement of complex formation between ATRS and its specific antibody. A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering ATRS epitopes is generally used, but a competitive binding assay may also be employed (Pound, su ra).
Various methods such as Scatchard analysis in conjunction with radioimmunoassay techniques may be used to assess the affinity of antibodies for ATRS. Affinity is expressed as an association constant, I~, which is defined as the molar concentration of ATRS-antibody complex divided by the molar concentrations of free antigen and free antibody under equilibrium conditions. The K.
determined for a preparation of polyclonal antibodies, which are heterogeneous in their afFnities for multiple ATRS epitopes, represents the average affinity, or avidity, of the antibodies for ATRS. The I~ determined for a preparation of monoclonal antibodies, which are monospecific for a particular ATRS epitope, represents a true measure of affinity. High-affinity antibody preparations with I~
ranging from about 109 to 1012 L/mole are preferred for use in immunoassays in which the ATRS-antibody complex must withstand rigorous manipulations. Low-affinity antibody preparations with I
ranging from about 106 to 10' L/mole are preferred for use in immunopurification and similar procedures which ultimately require dissociation of ATRS, preferably in active form, from the antibody (Catty, D. (1988) Antibodies, Volume I: A Practical Approach, IRL Press, Washington DC; Liddell, J.E. and A. Cryer (1991) A Practical Guide to Monoclonal Antibodies, John Wiley & Sons, New York NY).
The titer and avidity of polyclonal antibody preparations may be further evaluated to determine the quality and suitability of such preparations for certain downstream applications. For example, a polyclonal antibody preparation containing at least 1-2 mg specific antibody/ml, preferably 5-10 mg specific antibody/ml, is generally employed in procedures requiring precipitation of ATRS-antibody complexes. Procedures for evaluating antibody specificity, titer, and avidity, and guidelines for antibody quality and usage in various applications, are generally available.
(See, e.g., Catty, supra, and Coligan et al. supra.) In another embodiment of the invention, the polynucleotides encoding ATRS, or any fragment or complement thereof, may be used for therapeutic purposes. In one aspect, modifications of gene expression can be achieved by designing complementary sequences or antisense molecules (DNA, RNA, PNA, or modified oligonucleotides) to the coding or regulatory regions of the gene encoding ATRS. Such technology is well known in the art, and antisense oligonucleotides or larger fragments can be designed from various locations along the coding or control xegions of sequences encoding ATRS. (See, e.g., Agrawal, S., ed. (1996) Antisense Therapeutics, Humana Press Inc., Totawa NJ.) In therapeutic use, any gene delivery system suitable for introduction of the antisense sequences into appropriate target cells can be used. Antisense sequences can be delivered intracellulaxly in the form of an expression plasmid which, upon transcription, produces a sequence complementary to at least a portion of the cellular sequence encoding the target protein. (See, e.g., Slater, J.E, et al. (1998) J. Allergy Cli. Immunol. 102(3):469-475; and Scanlon, K.J. et al. (1995) 9(13):1288-1296.) Antisense sequences can also be introduced intracellularly through the use of viral vectors, such as retrovirus and adeno-associated virus vectors. (See, e.g., Miller, A.D. (1990) Blood 76:271; Ausubel, supra; Uckert, W. and W. Walther (1994) Pharmacol. Ther.
63(3):323-347.) Other gene delivery mechanisms include liposome-derived systems, artificial viral envelopes, and other systems known in the art. (See, e.g., Rossi, J.J. (1995) Br. Med. Bull.
51(1):217-225; Boado, R.J. et al. (1998) J. Pharm. Sci. 87(11):1308-1315; and Morris, M.C. et al. (1997) Nucleic Acids Res.
25(14):2730-2736.) In another embodiment of the invention, polynucleotides encoding ATRS may be used for somatic or germline gene therapy. Gene therapy may be performed to (i) correct a genetic deficiency (e.g., in the cases of severe combined immunodeficiency (SCID)-X1 disease characterized by X-linked inheritance (Cavazzana-Calvo, M. et al. (2000) Science 288:669-672), severe combined immunodeficiency syndrome associated with an inherited adenosine deaminase (ADA) deficiency (Blaese, R.M. et al. (1995) Science 270:475-480; Bordignon, C. et al. (1995) Science 270:470-475), cystic fibrosis (Zabner, J. et al. (1993) Cell 75:207-216; Crystal, R.G. et al. (1995) Hum. Gene Therapy 6:643-666; Crystal, R.G. et al. (1995) Hum. Gene Therapy 6:667-703), thalassamias, familial hypercholesterolemia, and hemophilia resulting from Factor VIII or Factor IX
deficiencies (Crystal, R.G. (1995) Science 270:404-410; Verma, LM. and N. Somia (1997) Nature 389:239-242)), (ii) express a conditionally lethal gene product (e.g., in the case of cancers which result from unregulated cell proliferation), or (iii) express a protein which affords protection against intracellular parasites (e.g., against human retroviruses, such as human immunodeficiency virus (HIV) (Baltimore, D. (1988) Nature 335:395-396; Poeschla, E. et al. (1996) Proc. Natl. Acad. Sci. USA.
93:11395-11399), hepatitis B or C virus (HBV, HCV); fungal parasites, such as Candida albicans and Paracoccidioides brasiliensis; and protozoan parasites such as Plasmodium falciparum and Trypanosoma cruzi). In the case where a genetic deficiency in ATRS expression or regulation causes disease, the expression of ATRS from an appropriate population of transduced cells may alleviate the clinical manifestations caused by the genetic deficiency.
In a further embodiment of the invention, diseases or disorders caused by deficiencies in ATRS are treated by constructing mammalian expression vectors encoding ATRS
and introducing these vectors by mechanical means into ATRS-deficient cells. Mechanical transfer technologies for use with cells in vivo or ex vitro include (i) direct DNA microinjection into individual cells, (ii) ballistic gold particle delivery, (iii) liposome-mediated transfection, (iv) receptor-mediated gene transfer, and (v) the use of DNA transposons (Morgan, R.A. and W.F. Anderson (1993) Annu.
Rev. Biochem.
62:191-217; Ivies, Z. (1997) Cell 91:501-510; Boulay, J-L. and H. Recipon (1998) Curr. Opin.
Biotechno1.9:445-450).
Expression vectors that may be effective for the expression of ATRS include, but are not limited to, the PCDNA 3.1, EPITAG, PRCCMV2, PREP, PVAX vectors (Invitrogen, Carlsbad CA), PCMV-SCRIPT, PCMV-TAG, PEGSH/PERV (Stratagene, La Jolla CA), and PTET-OFF, PTET-ON, PTRE2, PTRE2-LUC, PTK-HYG (Clontech, Palo Alto CA). ATRS may be expressed using (i) a constitutively active promoter, (e.g., from cytomegalovirus (CMV), Rous sarcoma virus (RSV), SV40 virus, thymidine kinase (TK), or ~3-actin genes), (ii) an inducible promoter (e.g., the tetracycline-regulated promoter (Gossen, M. and H. Bujard (1992) Proc. Natl.
Acad. Sci. USA
89:5547-5551; Gossen, M. et al. (1995) Science 268:1766-1769; Rossi, F.M.V.
and H.M. Blau (1998) C~rr. Opin. Biotechnol. 9:451-456), commercially available in the T-REX
plasmid (Invitrogen)); the ecdysone-inducible promoter (available in the plasmids PVGRXR and PIND;
Invitrogen); the FK506/rapamycin inducible promoter; or the RU486/mifepristone inducible promoter (Rossi, F.M.V.
and Blau, H.M. supra)), or (iii) a tissue-specific promoter or the native promoter of the endogenous gene encoding ATRS from a normal individual.
Commercially available liposome transformation kits (e.g., the PERFECT LIPID
TRANSFECTION KIT, available from Invitrogen) allow one with ordinary skill in the art to deliver polynucleotides to target cells in culture and require minimal effort to optimize experimental parameters. In the alternative, transformation is performed using the calcium phosphate method (Graham, F.L. and A.J. Eb (1973) Virology 52:456-467), or by electroporation (Neumann, E. et al.
(1982) EMBO J. 1:841-845). The introduction of DNA to primary cells requires modification of these standardized mammalian transfection protocols.
In another embodiment of the invention, diseases or disorders caused by genetic defects with respect to ATRS expression are treated by constructing a retrovirus vector consisting of (i) the polynucleotide encoding ATRS under the control of an independent promoter or the retrovirus long terminal repeat (LTR) promoter, (ii) appropriate RNA packaging signals, and (iii a Rev-responsive element (RRE) along with additional retrovirus cis-acting RNA sequences and coding sequences required for efFicient vector propagation. Retrovirus vectors (e.g., PFB and PFBNEO) are commercially available (Stratagene) and are based on published data (Riviere, I. et al. (1995) Proc.
Natl. Acad. Sci. USA 92:6733-6737), incorporated by reference herein. The vector is propagated in an appropriate vector producing cell line (VPCL) that expresses an envelope gene with a tropism for receptors on the target cells or a promiscuous envelope protein such as VSVg (Armentano, D. et al.
(1987) J. Virol. 61:1647-1650; Bender, M.A. et al. (1987) J. Virol. 61:1639-1646; Adam, M.A. and A.D. Miller (1988) J. Virol. 62:3802-3806; Dull, T. et al. (1998) J. Virol.
72:8463-8471; Zufferey, R. et al. (1998) J. Virol. 72:9873-9880). U.S. Patent Number 5,910,434 to Rigg ("Method for obtaining retrovims packaging cell lines producing high transducing efficiency retroviral supernatant") discloses a method for obtaining retrovirus packaging cell lines and is hereby incorporated by reference.
Propagation of retrovirus vectors, transduction of a population of cells (e.g., CD4+ T-cells), and the return of transduced cells to a patient are procedures well known to persons skilled in the art of gene therapy and have been well documented (Ranga, U. et al. (1997) J. Virol.
71:7020-7029; Bauer, G. et al. (1997) Blood 89:2259-2267; Bonyhadi, M.L. (1997) J. Virol. 71:4707-4716;
Ranga, U. et al. (1998) Proc. Natl. Acad. Sci. USA 95:1201-1206; Su, L. (1997) Blood 89:2283-2290).
In the alternative, an adenovirus-based gene therapy delivery system is used to deliver polynucleotides encoding ATRS to cells which have one or more genetic abnormalities with respect to 2o the expression of ATRS. The construction and packaging of adenovirus-based vectors are well known to those with ordinary skill in the art. Replication defective adenovirus vectors have proven to be versatile for importing genes encoding immunoregulatory proteins into intact islets in the pancreas (Csete, M.E. et al. (1995) Transplantation 27:263-268). Potentially useful adenoviral vectors are described in U.S. Patent Number 5,707,618 to Armentano ("Adenovirus vectors for gene therapy"), hereby incorporated by reference. For adenoviral vectors, see also Antinozzi, P.A. et al. (1999) Annu. Rev. Nutr. 19:511-544 and Verma, LM. and N. Somia (1997) Nature 18:389:239-242, both incorporated by reference herein.
In another alternative, a herpes-based, gene therapy delivery system is used to deliver polynucleotides encoding ATRS to target cells which have one or more genetic abnormalities with respect to the expression of ATRS. The use of herpes simplex virus (HSV)-based vectors may be especially valuable for introducing ATRS to cells of the central nervous system, for which HSV has a tropism. The construction and packaging of herpes-based vectors are well known to those with ordinary skill in the art. A replication-competent herpes simplex virus (HSV) type 1-based vector has been used to deliver a reporter gene to the eyes of primates (Liu, X. et al.
(1999) Exp. Eye Res.
169:385-395). The construction of a HSV-1 virus vector has also been disclosed in detail in U.S.
Patent Number 5,804,413 to DeLuca ("Herpes simplex virus strains for gene transfer"), which is hereby incorporated by reference. U.S. Patent Number 5,804,413 teaches the use of recombinant HSV d92 which consists of a genome containing at least one exogenous gene to be transferred to a cell under the control of the appropriate promoter for purposes including human gene therapy. Also taught by this patent are the construction and use of recombinant HSV strains deleted for ICP4, ICP27 and ICP22. For HSV vectors, see also Goins, W.F. et al. (1999) J. Virol.
73:519-532 and Xu, H. et al. (1994) Dev. Biol. 163:152-161, hereby incorporated by reference. The manipulation of cloned herpesvirus sequences, the generation of recombinant virus following the transfection of multiple plasmids containing different segments of the large herpesvirus genomes, the growth and propagation of herpesvirus, and the infection of cells with herpesvirus are techniques well known to those of ordinary skill in the art.
In another alternative, an alphavirus (positive, single-stranded RNA virus) vector is used to deliver polynucleotides encoding ATRS to target cells. The biology of the prototypic alphavirus, Semliki Forest Virus (SFV), has been studied extensively and gene transfer vectors have been based on the SFV genome (Garoff, H. and K.-J. Li (1998) Curr. Opin. Biotechnol.
9:464-469). During alphavirus RNA replication, a subgenomic RNA is generated that normally encodes the viral capsid proteins. This subgenomic RNA replicates to higher levels than the full length genomic RNA, resulting in the overproduction of capsid proteins relative to the viral proteins with enzymatic activity (e.g., protease and polymerase). Similarly, inserting the coding sequence for ATRS into the alphavirus genome in place of the capsid-coding region results in the production of a large number of ATRS-coding RNAs and the synthesis of high levels of ATRS in vector transduced cells. While alphavirus infection is typically associated with cell lysis within a few days, the ability to establish a persistent infection in hamster normal kidney cells (BHK-21) with a variant of Sindbis virus (SIN) indicates that the lytic replication of alphaviruses can be altered to suit the needs of the gene therapy application (Dryga, S.A. et al. (1997) Virology 228:74-83). The wide host range of alphaviruses will allow the introduction of ATRS into a variety of cell types. The specific transduction of a subset of cells in a population may require the sorting of cells prior to transduction. The methods of manipulating infectious cDNA clones of alphaviruses, performing alphavirus cDNA and RNA
transfections, and performing alphavirus infections, are well known to those with ordinary skill in the art.
Oligonucleotides derived from the transcription initiation site, e.g., between about positions -10 and +10 from the start site, may also be employed to inhibit gene expression.
Similarly, inhibition can be achieved using triple helix base-pairing methodology. Triple helix pairing is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or regulatory molecules. Recent therapeutic advances using triplex DNA have been described in the literature. (See, e.g., Gee, J.E. et al. (1994) in Huber, B.E. and B.I. Carr, Molecular and Immunolo~ic Approaches, Futura Publishing, Mt. Kisco NY, pp. 163-177.) A
complementary sequence or antisense molecule may also be designed to block translation of mRNA
by preventing the transcript from binding to ribosomes.
Ribozymes, enzymatic RNA molecules, may also be used to catalyze the specific cleavage of RNA. The mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
For example, engineered hammerhead motif ribozyme molecules may specifically and efficiently catalyze endonucleolytic cleavage of sequences encoding ATRS.
Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, including 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 secondary structural features which may render the oligonucleotide inoperable.
The suitability of candidate targets may also be evaluated by testing accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays.
Complementary ribonucleic acid molecules and ribozymes of the invention may be prepared by any method known in the art for the synthesis of nucleic acid molecules.
These include techniques for chemically synthesizing oligonucleotides such as solid phase phosphoramidite chemical synthesis.
Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA
sequences encoding ATRS. Such DNA sequences may be incorporated into a wide variety of vectors with suitable RNA polymerase promoters such as T7 or SP6.
Alternatively, these cDNA
constructs that synthesize complementary RNA, constitutively or inducibly, can be introduced into cell lines, cells, or tissues.
RNA molecules may be modified to increase intracellular stability and half life. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5' and/or 3' ends of the molecule, or the use of phosphorothioate or 2' O-methyl rather than phosphodiesterase linkages within the backbone of the molecule. This concept is inherent in the production of PNAs and can be extended in all of these molecules by the inclusion of nontraditional bases such as inosine, queosine, and wybutosine, as well as acetyl-, methyl-, thio-, and similarly modified forms of adenine, cytidine, guanine, thymine, and uridine which are not as easily recognized by endogenous endonucleases.
An additional embodiment of the invention encompasses a method for screening for a compound which is effective in altering expression of a polynucleotide encoding ATRS. Compounds which may be effective in altering expression of a specific polynucleotide may include, but are not limited to, oligonucleotides, antisense oligonucleotides, triple helix-forming oligonucleotides, transcription factors and other polypeptide transcriptional regulators, and non-macromolecular chemical entities which are capable of interacting with specific polynucleotide sequences. Effective compounds may alter polynucleotide expression by acting as either inhibitors or promoters of polynucleotide expression. Thus, in the treatment of disorders associated with increased ATRS
expression or activity, a compound which specifically inhibits expression.of the polynucleotide encoding ATRS may be therapeutically useful, and in the treatment of disorders associated with decreased ATRS expression or activity, a compound which specifically promotes expression of the polynucleotide encoding ATRS may be therapeutically useful.
At least one, and up to a plurality, of test compounds may be screened for effectiveness in altering expression of a specific polynucleotide. A test compound may be obtained by any method commonly known in the art, including chemical modification of a compound known to be effective in altering polynucleotide expression; selection from an existing, commercially-available or proprietary library of naturally-occurring or non-natural chemical compounds; rational design of a compound based on chemical and/or structural properties of the target polynucleotide;
and selection from a library of chemical compounds created combinatorially or randomly. A sample comprising a polynucleotide encoding ATRS is exposed to at least one test compound thus obtained. The sample may comprise, for example, an intact or permeabilized cell, or an in vitro cell-free or reconstituted biochemical system. Alterations in the expression of a polynucleotide encoding ATRS are assayed by any method commonly known in the art. Typically, the expression of a specific nucleotide is detected by hybridization with a probe having a nucleotide sequence complementary to the sequence of the polynucleotide encoding ATRS. The amount of hybridization may be quantified, thus forming the basis for a comparison of the expression of the polynucleotide both with and without exposure to one or more test compounds. Detection of a change in the expression of a polynucleotide exposed to a test compound indicates that the test compound is effective in altering the expression of the polynucleotide. A screen for a compound effective in altering expression of a specific polynucleotide can be carried out, for example, using a Schizosaccharom~ces pombe gene expression system (Atkins, D. et al. (1999) U.S. Patent No. 5,932,435; Arndt, G.M. et al. (2000) Nucleic Acids Res. 28:E15) or a human cell line such as HeLa cell (Clarke, M.L. et al. (2000) Biochem.
Biophys. Res. Common.

268:8-13). A particular embodiment of the present invention involves screening a combinatorial library of oligonucleotides (such as deoxyribonucleotides, ribonucleotides, peptide nucleic acids, and modified oligonucleotides) for antisense activity against a specific polynucleotide sequence (Bruice, T.W. et al.
(1997) U.S. Patent No. 5,686,242; Bruice, T.W. et al. (2000) U.S. Patent No.
6,022,691).
Many methods for introducing vectors into cells or tissues are available and equally suitable for use in vivo, in vitro, and ex vivo. For ex vivo therapy, vectors may be introduced into stem cells taken from the patient and clonally propagated for autologous transplant back into that same patient.
Delivery by transfection, by liposome injections, or by polycationic amino polymers may be achieved using methods which are well known in the art. (See, e.g., Goldman, C.K. et al. (1997) Nat.
Biotechno1.15:462-466.) Any of the therapeutic methods described above may be applied to any subject in need of such therapy, including, for example, mammals such as humans, dogs, cats, cows, horses, rabbits, and monkeys.
An additional embodiment of the invention relates to the administration of a composition which generally comprises an active ingredient formulated with a pharmaceutically acceptable excipient.
Excipients may include, for example, sugars, starches, celluloses, gums, and proteins. Various formulations are commonly known and are thoroughly discussed in the latest edition of Remin~ton's Pharmaceutical Sciences (Maack Publishing, Easton PA). Such compositions may consist of ATRS, antibodies to ATRS, and mimetics, agonists, antagonists, or inhibitors of ATRS.
The compositions utilized in this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, infra-arterial, intramedullary, intrathecal, intraventricular, pulmonary, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, or rectal means.
Compositions for pulmonary administration may be prepared in liquid or dry powder form.
These compositions are generally aerosolized immediately prior to inhalation by the patient. In the case of small molecules (e.g. traditional low molecular weight organic drugs), aerosol delivery of fast-acting formulations is well-known in the art. In the case of macromolecules (e.g. larger peptides and proteins), recent developments in the field of pulmonary delivery via the alveolar region of the lung have enabled the practical delivery of drugs such as insulin to blood circulation (see, e.g., Patton, J.S.
et al., U.S. Patent No. 5,997,848). Pulmonary delivery has the advantage of administration without needle injection, and obviates the need for potentially toxic penetration enhancers.
Compositions suitable for use in the invention include compositions wherein the active ingredients are contained in an effective amount to achieve the intended purpose. The determination of an effective dose is well within the capability of those skilled in the art.
Specialized forms of compositions may be prepared for direct intracellular delivery of macromolecules comprising ATRS or fragments thereof. For example, liposome preparations containing a cell-impermeable macromolecule may promote cell fusion and intracellular delivery of the macromolecule. Alternatively, ATRS or a fragment thereof may be joined to a short cationic N-terminal portion from the HIV Tat-1 protein. Fusion proteins thus generated have been found to transduce into the cells of all tissues, including the brain, in a mouse model system (Schwarze, S.R. et al. (1999) Science 285:1569-1572).
For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays, e.g., of neoplastic cells, or in animal models such as mice, rats, rabbits, dogs, monkeys, or pigs. An animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
A therapeutically effective dose refers to that amount of active ingredient, for example ATRS
or fragments thereof, antibodies of ATRS, and agonists, antagonists or inhibitors of ATRS, which ameliorates the symptoms or condition. Therapeutic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or with experimental animals, such as by calculating the EDSO (the dose therapeutically effective in 50% of the population) or LDSO (the dose lethal to 50% of the population) statistics. The dose ratio of toxic to therapeutic effects is the therapeutic index, which can be expressed as the LDSOBDso ratio. Compositions which exhibit large therapeutic indices are preferred. The data obtained from cell culture assays and animal studies are used to formulate a range of dosage for human use. The dosage contained in such compositions is preferably within a range of circulating concentrations that includes the EDSO
with little or no toxicity.
The dosage varies within this range depending upon the dosage form employed, the sensitivity of the patient, and the route of administration.
The exact dosage will be determined by the practitioner, in light of factors related to the subject requiring treatment. Dosage and administration are adjusted to provide sufficient levels of the active moiety or to maintain the desired effect. Factors which may be taken into account include the severity of the disease state, the general health of the subject, the age, weight, and gender of the subject, time and frequency of administration, drug combination(s), reaction sensitivities, and response to therapy. Long-acting compositions may be administered every 3 to 4 days, every week, or biweekly depending on the half life and clearance rate of the particular formulation.
Normal dosage amounts may vary from about 0.1 ~cg to 100,000 ,ug, up to a total dose of about 1 gram, depending upon the route of administration. Guidance as to particular dosages and methods of delivery is provided in the literature and generally available to practitioners in the art.
Those skilled in the art will employ different formulations for nucleotides than for proteins or their inhibitors. Similarly, delivery of polynucleotides or polypeptides will be specific to particular cells, conditions, locations, etc.
DIAGNOSTICS
In another embodiment, antibodies which specifically bind ATRS may be used for the diagnosis of disorders characterized by expression of ATRS, or in assays to monitor patients being treated with ATRS or agonists, antagonists, or inhibitors of ATRS. Antibodies useful for diagnostic purposes may be prepared in the same manner as described above for therapeutics. Diagnostic assays for ATRS include methods which utilize the antibody and a label to detect ATRS in human body fluids or in extracts of cells or tissues. The antibodies may be used with or without modification, and may be labeled by covalent or non-covalent attachment of a reporter molecule. A wide variety of reporter molecules, several of which are described above, are known in the art and may be used.
A variety of protocols for measuring ATRS, including ELISAs, RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of ATRS expression. Normal or standard values for ATRS expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, for example, human subjects, with antibodies to ATRS under conditions suitable for complex formation. The amount of standard complex formation may be quantitated by various methods, such as photometric means. Quantities of ATRS
expressed in subject, control, and disease samples from biopsied tissues are compared with the standaxd values.
Deviation between standard and subject values establishes the parameters for diagnosing disease.
In another embodiment of the invention, the polynucleotides encoding ATRS may be used for diagnostic purposes. The polynucleotides which may be used include oligonucleotide sequences, complementary RNA and DNA molecules, and PNAs. The polynucleotides may be used to detect and quantify gene expression in biopsied tissues in which expression of ATRS
may be correlated with disease. The diagnostic assay may be used to determine absence, presence, and excess expression of ATRS, and to monitor regulation of ATRS levels during therapeutic intervention.
In one aspect, hybridization with PCR probes which axe capable of detecting pulynucleutide sequences, including genomic sequences, encoding ATRS or closely related molecules may be used to identify nucleic acid sequences which encode ATRS. The specificity of the probe, whether it is made from a highly specific region, e.g., the 5'regulatory region, or from a less specific region, e.g., a conserved motif, and the stringency of the hybridization or amplification will determine whether the probe identifies only naturally occurnng sequences encoding ATRS, allelic variants, or related sequences.
Probes may also be used for the detection of related sequences, and may have at least 50%
sequence identity to any of the ATRS encoding sequences. The hybridization probes of the subject invention may be DNA or RNA and may be derived from the sequence of SEQ ID
NO:S-8 or from genomic sequences including promoters, enhancers, and introns of the ATRS
gene.
Means for producing specific hybridization probes for DNAs encoding ATRS
include the cloning of polynucleotide sequences encoding ATRS or ATRS derivatives into vectors for the production of mRNA probes. Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by means of the addition of the appropriate RNA
polymerases and the appropriate labeled nucleotides. Hybridization probes may be labeled by a variety of reporter groups, fox example, by radionuclides such as 32P or 355, or by enzymatic labels, such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems, and the like.
Polynucleotide sequences encoding ATRS may be used for the diagnosis of disorders associated with expression of ATRS. Examples of such disorders include, but are not limited to, a cell proliferative disorder such as actinic keratosis, arteriosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCTD), myelofibrosis, paroxysmal nocturnal hemoglobinuria, polycythemia vera, psoriasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus;
and an autoimmune/inflammatory disorder such as acquired immunodeficiency syndrome (AIDS), Addison's disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), bronchitis, cholecystitis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes mellitus, emphysema, episodic lymphopenia with lymphocytotoxins, erythroblastosis fetalis, erythema nodosum, atrophic gastritis, glomerulonephritis, Goodpasture's syndrome, gout, Graves' disease, Hashimoto's thyroiditis, hypereosinophilia, irritable bowel syndrome, multiple sclerosis, myasthenia gravis, myocardial or pericardial inflammation, osteoarthritis, osteoporosis, pancreatitis, polymyositis, psoriasis, Reiter's syndrome, rheumatoid arthritis, scleroderma, Sjogren's syndrome, systemic anaphylaxis, systemic lupus erythematosus, systemic sclerosis, thrombocytopenic purpura, ulcerative colitis, uveitis, Werner syndrome, complications of cancer, hemodialysis, and extracorporeal circulation, viral, bacterial, fungal, parasitic, protozoal, and helminthic infections, and trauma. The polynucleotide sequences encoding ATRS may be used in Southern or northern analysis, dot blot, or other membrane-based technologies; in PCR technologies; in dipstick, pin, and multiformat ELISA-like assays; and in microarrays utilizing fluids or tissues from patients to detect altered ATRS
expression. Such qualitative or quantitative methods are well known in the art.
In a particular aspect, the nucleotide sequences encoding ATRS may be useful in assays that detect the presence of associated disorders, particularly those mentioned above. The nucleotide sequences encoding ATRS may be labeled by standard methods and added to a fluid or tissue sample from a patient under conditions suitable for the formation of hybridization complexes. After a suitable incubation period, the sample is washed and the signal is quantified and compared with a standard ~ value. If the amount of signal in the patient sample is significantly altered in comparison to a control sample then the presence of altered levels of nucleotide sequences encoding ATRS in the sample indicates the presence of the associated disorder. Such assays may also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies, in clinical trials, or to monitor the treatment of an individual patient.
In order to provide a basis for the diagnosis of a disorder associated with expression of ATRS, a normal or standard profile for expression is established. This may be accomplished by combining body fluids or cell extracts taken from normal subjects, either animal or human, with a sequence, or a fragment thereof, encoding ATRS, under conditions suitable for hybridization or amplification.
Standard hybridization may be quantified by comparing the values obtained from normal subjects with values from an experiment in which a known amount of a substantially purified polynucleotide is used.
Standard values obtained in this manner may be compared with values obtained from samples from patients who are symptomatic for a disorder. Deviation from standard values is used to establish the presence of a disorder.
Once the presence of a disorder is established and a treatment protocol is initiated, hybridization assays may be repeated on a regular basis to deterrrune if the level of expression in the patient begins to approximate that which is observed in the normal subject.
The results obtained from successive assays may be used to show the efficacy of treatment over a period ranging from several days to months.
With respect to cancer, the presence of an abnormal amount of transcript (either under- or overexpressed) in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer.
Additional diagnostic uses for oligonucleotides designed from the sequences encoding ATRS
may involve the use of PCR. These oligomers may be chemically synthesized, generated enzymatically, or produced in vitro. Oligomers will preferably contain a fragment of a polynucleotide encoding ATRS, or a fragment of a polynucleotide complementary to the polynucleotide encoding ATRS, and will be employed under optimized conditions for identification of a specific gene or condition. Oligomers may also be employed under less stringent conditions for detection or l0 quantification of closely related DNA or RNA sequences.
In a particular aspect, oligonucleotide primers derived from the polynucleotide sequences encoding ATRS may be used to detect single nucleotide polymorphisms (SNPs).
SNPs are substitutions, insertions and deletions that are a frequent cause of inherited or acquired genetic disease in humans. Methods of SNP detection include, but are not limited to, single-stranded conformation i5 polymorphism (SSCP) and fluorescent SSCP (fSSCP) methods. In SSCP, oligonucleotide primers derived from the polynucleotide sequences encoding ATRS are used to amplify DNA using the polymerase chain reaction (PCR). The DNA may be derived, for example, from diseased or normal tissue, biopsy samples, bodily fluids, and the like. SNPs in the DNA cause differences in the secondary and tertiary structures of PCR products in single-stranded form, and these differences are 20 detectable using gel electrophoresis in non-denaturing gels. In fSCCP, the oligonucleotide primers are fluorescently labeled, which allows detection of the amplimers in high-throughput equipment such as DNA sequencing machines. Additionally, sequence database analysis methods, termed in silico SNP
(isSNP), are capable of identifying polymorphisms by comparing the sequence of individual overlapping DNA fragments which assemble into a common consensus sequence.
These computer-25 based methods filter out sequence variations due to laboratory preparation of DNA and sequencing errors using statistical models and automated analyses of DNA sequence chromatograms. In the alternative, SNPs may be detected and characterized by mass spectrometry using, for example, the high throughput MASSARRAY system (Sequenom, Inc., San Diego CA).
Methods which may also be used to quantify the expression of ATRS include radiolabeling or 30 biotinylating nucleotides, coamplification of a control nucleic acid, and interpolating results from standard curves. (See, e.g., Melby, P.C. et al. (1993) J. Immunol. Methods 159:235-244; Duplaa, C.
et al. (1993) Anal. Biochem. 212:229-236.) The speed of quantitation of multiple samples may be accelerated by running the assay in a high-throughput format where the oligomer or polynucleotide of interest is presented in various dilutions and a spectrophotometric or colorimetric response gives rapid quantitation.
In further embodiments, oligonucleotides or longer fragments derived from any of the polynucleotide sequences described herein may be used as elements on a microarray. The microarray can be used in transcript imaging techniques which monitor the relative expression levels of large numbers of genes simultaneously as described below. The microarray may also be used to identify genetic variants, mutations, and polymorphisms. This information may be used to determine gene function, to understand the genetic basis of a disorder, to diagnose a disorder, to monitor progression/regression of disease as a function of gene expression, and to develop and monitor the activities of therapeutic agents in the treatment of disease. In particular, this information may be used to develop a pharmacogenomic profile of a patient in order to select the most appropriate and effective treatment regimen for that patient. For example, therapeutic agents which are highly effective and display the fewest side effects may be selected for a patient based on his/her pharmacogenomic profile.
In another embodiment, ATRS, fragments of ATRS, or antibodies specific for ATRS may be used as elements on a microarray. The microarray may be used to monitor or measure protein-protein interactions, drug-target interactions, and gene expression profiles, as described above.
A particular embodiment relates to the use of the polynucleotides of the present invention to generate a transcript image of a tissue or cell type. A transcript image represents the global pattern of gene expression by a particular tissue or cell type. Global gene expression patterns are analyzed by quantifying the number of expressed genes and their relative abundance under given conditions and at a given time. (See Seilhamer et al., "Comparative Gene Transcript Analysis,"
U.S. Patent Number 5,840,484, expressly incorporated by reference herein.) Thus a transcript image may be generated by hybridizing the polynucleotides of the present invention or their complements to the totality of transcripts or reverse transcripts of a particular tissue or cell type. In one embodiment, the hybridization takes place in high-throughput format, wherein the polynucleotides of the present invention or their complements comprise a subset of a plurality of elements on a microarray. The resultant transcript image would provide a profile of gene activity.
Transcript images may be generated using transcripts isolated from tissues, cell lines, biopsies, or other biological samples. The transcript image may thus reflect gene expression in vivo, as in the case of a tissue or biopsy sample, or in vitro, as in the case of a cell line.
Transcript images which profile the expression of the polynucleotides of the present invention may also be used in conjunction with in vitro model systems and preclinical evaluation of pharmaceuticals, as well as toxicological testing of industrial and naturally-occurring environmental compounds. All compounds induce characteristic gene expression patterns, frequently termed molecular fingerprints or toxicant signatures, which are indicative of mechanisms of action and toxicity (Nuwaysir, E.F. et al. (1999) Mol. Carcinog. 24:153-159; Steiner, S. and N.L.
Anderson (2000) Toxicol. Lett. 112-113:467-471, expressly incorporated by reference herein).
If a test compound has a signature similar to that of a compound with known toxicity, it is likely to share those toxic properties.
These fingerprints or signatures are most useful and refined when they contain expression information from a large number of genes and gene families. Ideally, a genome-wide measurement of expression provides the highest quality signature. Even genes whose expression is not altered by any tested compounds are important as well, as the levels of expression of these genes are used to normalize the rest of the expression data. The normalization procedure is useful for comparison of expression data after treatment with different compounds. While the assignment of gene function to elements of a toxicant signature aids in interpretation of toxicity mechanisms, knowledge of gene function is not necessary for the statistical matching of signatures which leads to prediction of toxicity. (See, for example, Press Release 00-02 from the National Institute of Environmental Health Sciences, released February 29, 2000, available at http://www.niehs.nih.gov/oc/news/toxchip.htm.) Therefore, it is important and desirable in toxicological screening using toxicant signatures to include all expressed gene sequences.
In one embodiment, the toxicity of a test compound is assessed by treating a biological sample containing nucleic acids with the test compound. Nucleic acids that are expressed in the treated biological sample are hybridized with one or more probes specific to the polynucleotides of the present invention, so that transcript levels corresponding to the polynucleotides of the present invention may be quantified. The transcript levels in the treated biological sample are compared with levels in an untreated biological sample. Differences in the transcript levels between the two samples are indicative of a toxic response caused by the test compound in the treated sample.
Another particular embodiment relates to the use of the polypeptide sequences of the present .
invention to analyze the proteome of a tissue or cell type. The term proteome refers to the global pattern of protein expression in a particular tissue or cell type. Each protein component of a proteome can be subjected individually to further analysis. Proteome expression patterns, or profiles, are analyzed by quantifying the number of expressed proteins and their relative abundance under given conditions and at a given time. A profile of a cell's proteome may thus be generated by separating and analyzing the polypeptides of a particular tissue or cell type. In one embodiment, the separation is achieved using two-dimensional gel electrophoresis, in which proteins from a sample are separated by isoelectric focusing in the first dimension, and then according to molecular weight by sodium dodecyl sulfate slab gel electrophoresis in the second dimension (Steiner and Anderson, supra). The proteins are visualized in the gel as discrete and uniquely positioned spots, typically by staining the gel with an agent such as Coomassie Blue or silver or fluorescent stains. The optical density of each protein spot is generally proportional to the level of the protein in the sample. The optical densities of equivalently positioned protein spots from different samples, for example, from biological samples either treated or untreated with a test compound or therapeutic agent, are compared to identify any changes in protein spot density related to the treatment. The proteins in the spots are partially sequenced using, for example, standard methods employing chemical or enzymatic cleavage followed by mass spectrometry. The identity of the protein in a spot may be determined by comparing its partial sequence, preferably of at least 5 contiguous amino acid residues, to the polypeptide sequences of the present invention. In some cases, further sequence data may be obtained for definitive protein identification.
A proteomic profile may also be generated using antibodies specific for ATRS
to quantify the levels of ATRS expression. In one embodiment, the antibodies are used as elements on a microarray, and protein expression levels are quantified by exposing the microarray to the sample and detecting the levels of protein bound to each array element (Lueking, A. et al. (1999) Anal. Biochem. 270:103-111; Mendoze, L.G. et al. (1999) Biotechniques 27:778-788). Detection may be performed by a variety of methods known in the art, for example, by reacting the proteins in the sample with a thiol- or amino-reactive fluorescent compound and detecting the amount of fluorescence bound at each array element.
Toxicant signatures at the proteome level are also useful for toxicological screening, and should be analyzed in parallel with toxicant signatures at the transcript level. There is a poor correlation between transcript and protein abundances for some proteins in some tissues (Anderson, N.L. and J. Seilhamer (1997) Electrophoresis 18:533-537), so proteome toxicant signatures may be useful in the analysis of compounds which do not~significantly affect the transcript image, but which alter the proteomic profile. In addition, the analysis of transcripts in body fluids is difficult, due to rapid degradation of mRNA, so proteomic profiling may be more reliable and informative in such cases.
In another embodiment, the toxicity of a test compound is assessed by treating a biological sample containing proteins with the test compound. Proteins that are expressed in the treated biological sample are separated so that the amount of each protein can be quantified. The amount of each protein is compared to the amount of the corresponding protein in an untreated biological sample.
A difference in the amount of protein between the two samples is indicative of a toxic response to the test compound in the treated sample. Individual proteins are identified by sequencing the amino acid residues of the individual proteins and comparing these partial sequences to the polypeptides of the present invention.
In another embodiment, the toxicity of a test compound is assessed by treating a biological sample containing proteins with the test compound. Proteins from the biological sample are incubated with antibodies specific to the polypeptides of the present invention. The amount of protein recognized by the antibodies is quantified. The amount of protein in the treated biological sample is compared with the amount in an untreated biological sample. A difference in the amount of protein between the two samples is indicative of a toxic response to the test compound in the treated sample.
Microarrays may be prepared, used, and analyzed using methods known in the art. (See, e.g., Brennan, T.M. et al. (1995) U.S. Patent No. 5,474,796; Schena, M. et al.
(1996) Proc. Natl. Acad.
Sci. USA 93:10614-10619; Baldeschweiler et al. (1995) PCT application W0951251116; Shalon, D. et al. (1995) PCT application W095135505; Heller, R.A. et al. (1997) Proc. Natl.
Acad. Sci. USA
94:2150-2155; and Heller, M.J. et al. (1997) U.S. Patent No. 5,605,662.) Various types of microarrays are well known and thoroughly described in DNA Microarrays: A
Practical Approach, M. Schena, ed. (1999) Oxford University Press, London, hereby expressly incorporated by reference.
In another embodiment of the invention, nucleic acid sequences encoding ATRS
may be.used to generate hybridization probes useful in mapping the naturally occurring genomic sequence. Either coding or noncoding sequences may be used, and in some instances, noncoding sequences may be preferable over coding sequences. For example, conservation of a coding sequence among members of a mufti-gene family may potentially cause undesired cross hybridization during chromosomal mapping. The sequences may be mapped to a particular chromosome, to a specific region of a chromosome, or to artificial chromosome constructions, e.g., human artificial chromosomes (HACs), yeast artificial chromosomes (YACs), bacterial artificial chromosomes (BACs), bacterial Pl constructions, or single chromosome cDNA libraries. (See, e.g., Harrington, J.J. et al. (1997) Nat.
Genet. 15:345-355; Price, C.M. (1993) Blood Rev. 7:127-134; and Trask, B.J.
(1991) Trends Genet.
7:149-154.) Once mapped, the nucleic acid sequences of the invention may be used to develop genetic linkage maps, for example, which correlate the inheritance of a disease state with the inheritance of a particular chromosome region or restriction fragment length polymorphism (RFLP).
(See, for example, Lander, E.S. and D. Botstein (1986) Proc. Natl. Acad. Sci.
USA 83:7353-7357.) Fluorescent in situ hybridization (FISH) may be correlated with other physical and genetic map data. (See, e.g., Heinz-Ulrich, et al. (1995) in Meyers, supra, pp. 965-968.) Examples of genetic map data can be found in various scientific journals or at the Online Mendelian Inheritance in Man (OMIM) World Wide Web site. Correlation between the location of the gene encoding ATRS on a physical map and a specific disorder, or a predisposition to a specific disorder, may help define the region of DNA associated with that disorder and thus may further positional cloning efforts.
In situ hybridization of chromosomal preparations and physical mapping techniques, such as linkage analysis using established chromosomal markers, may be used for extending genetic maps.
Often the placement of a gene on the chromosome of another mammalian species, such as mouse, may reveal associated markers even if the exact chromosomal locus is not known. This information is valuable to investigators searching for disease genes using positional cloning or other gene discovery techniques. Once the gene or genes responsible for a disease or syndrome have been crudely localized by genetic linkage to a particular genomic region, e.g., ataxia-telangiectasia to 11q22-23, any sequences mapping to that area may represent associated or regulatory genes for further investigation.
(See, e.g., Gatti, R.A. et al. (1988) Nature 336:577-580.) The nucleotide sequence of the instant invention may also be used to detect differences in the chromosomal location due to translocation, inversion, etc., among normal, carrier, or affected individuals.
In another embodiment of the invention, ATRS, its catalytic or immunogenic fragments, or oligopeptides thereof can be used for screening libraries of compounds in any of a variety of drug screening techniques. The fragment employed in such screening may be free in solution, affixed to a solid support, borne on a cell surface, or located intracellularly. The formation of binding complexes between ATRS and the agent being tested may be measured.
Another technique for drug screening provides for high throughput screening of compounds having suitable binding affinity to the protein of interest. (See, e.g., Geysen, et al. (1984) PCT
application W084/03564.) In this method, large numbers of different small test compounds are synthesized on a solid substrate. The test compounds are reacted with ATRS, or fragments thereof, and washed. Bound ATRS is then detected by methods well known in the art.
Purified ATRS can also be coated directly onto plates for use in the aforementioned drug screening techniques.
Alternatively, non-neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support.
In another embodiment, one may use competitive drug screening assays in which neutralizing antibodies capable of binding ATRS specifically compete with a test compound for binding ATRS. In this manner, antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants with ATRS.
In additional embodiments, the nucleotide sequences which encode ATRS may be used in any molecular biology techniques that have yet to be developed, provided the new techniques rely on properties of nucleotide sequences that are currently known, including, but not limited to, such properties as the triplet genetic code and specific base pair interactions.
Without flurther elaboration, it is believed that one skilled in the art can, using the preceding description, utilize the present invention to its fullest extent. The following embodiments are, therefore, to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever.
The disclosures of all patents, applications and publications, mentioned above and below, including U.S. Ser. No. 60/207,248, U.S. Ser. No. 60/208,791, and U.S. Ser.
No. 60/210,585, are expressly incorporated by reference herein.
EXAMPLES
I. Construction of cDNA Libraries Incyte cDNAs were derived from cDNA libraries described in the LIFESEQ GOLD
database (Incyte Genomics, Palo Alto CA) and shown in Table 4, column 5. Some tissues were homogenized and lysed in guanidinium isothiocyanate, while others were homogenized and lysed in phenol or in a suitable mixture of denaturants, such as TRIZOL (Life Technologies), a monophasic solution of phenol and guanidine isothiocyanate. The resulting lysates were centrifuged over CsCI
cushions or extracted with chloroform. RNA was precipitated from the lysates with either isopropanol or sodium acetate and ethanol, or by other routine methods.
Phenol extraction and precipitation of RNA were repeated as necessary to increase RNA
purity. In some cases, RNA was treated with DNase. For most libraries, poly(A)+ RNA was isolated using oligo d(T)-coupled paramagnetic particles (Promega), OLIGOTEX
latex particles (QIAGEN, Chatsworth CA), or an OLIGOTEX mRNA purification kit (QIAGEN).
Alternatively, RNA was isolated directly from tissue lysates using other RNA isolation kits, e.g., the POLY(A)PURE mRNA purification kit (Ambion, Austin TX).
In some cases, Stratagene was provided with RNA and constructed the corresponding cDNA
libraries. Otherwise, cDNA was synthesized and cDNA libraries were constructed with the .
UNIZAP vector system (Stratagene) or SUPERSCRIPT plasmid system (Life Technologies), using the recommended procedures or similar methods known in the art. (See, e.g., Ausubel, 1997, supra, units 5.1-6.6.) Reverse transcription was initiated using oligo d(T) or random primers. Synthetic oligonucleotide adapters were ligated to double stranded cDNA, and the eDNA
was digested with the appropriate restriction enzyme or enzymes. For most libraries, the cDNA was size-selected (300-1000 bp) using SEPHACRYL 51000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (Amersham Pharmacia Biotech) or preparative agarose gel electrophoresis. cDNAs were ligated into compatible restriction enzyme sites of the polylinker of a suitable plasmid, e.g., PBLUESCRIPT plasmid (Stratagene), PSPORT1 plasmid (Life Technologies), PCDNA2.1 plasmid (Invitrogen, Carlsbad CA), PBK-CMV plasmid (Stratagene)> or pINCY (Incyte Genomics, Palo Alto CA), or derivatives thereof. Recombinant plasmids were transformed into competent E. coli cells including XLl-Blue, XLl-BIueMRF, or SOLR from Stratagene or DHSa, DH10B, or ElectroMAX
DH10B from Life Technologies.
II. Isolation of cDNA Clones Plasmids obtained as described in Example I were recovered from host cells by in vivo excision using the UNIZAP vector system (Stratagene) or by cell lysis.
Plasmids were purified using at least one of the following: a Magic or WIZARD Minipreps DNA purification system (Promega); an AGTC Miniprep purification kit (Edge Biosystems, Gaithersburg MD); and QIAWELL
8 Plasmid, QIAWELL 8 Plus Plasmid, QIAWELL 8 Ultra Plasmid purification systems or the R.E.A.L. PREP
96 plasmid purification kit from QIAGEN. Following precipitation, plasmids were resuspended in 0.1 ml of distilled water and stored, with or without lyophilization, at 4 ° C.
Alternatively, plasmid DNA was amplified from host cell lysates using direct link PCR in a high-throughput format (Rao, V.B. (1994) Anal. Biochem. 216:1-14). Host cell lysis and thermal cycling steps were carried out in a single reaction mixture. Samples were processed and stored in 384-well plates, and the concentration of amplified plasmid DNA was quantified fluorometrically using PICOGREEN dye (Molecular Probes, Eugene OR) and a FLUOROSKAN II fluorescence scanner (Labsystems Oy, Helsinki, Finland).
III. Sequencing and Analysis Incyte cDNA recovered in plasmids as described in Example II were sequenced as follows.
Sequencing reactions were processed using standard methods or high-throughput instrumentation such as the ABI CATALYST 800 (Applied Biosystems) thermal cycler or the PTC-200 thermal cycler (MJ Research) in conjunction with the HYDRA microdispenser (Robbins Scientific) or the MICROLAB 2200 (Hamilton) liquid transfer system. cDNA sequencing reactions were prepared using reagents provided by Amersham Pharmacia Biotech or supplied in ABI
sequencing kits such as the ABI PRISM BIGDYE Terminator cycle sequencing ready reaction kit (Applied Biosystems).
Electrophoretic separation of cDNA sequencing reactions and detection of labeled polynucleotides were carried out using the MEGABACE 1000 DNA sequencing system (Molecular Dynamics); the ABI PRISM 373 or 377 sequencing system (Applied Biosystems) in conjunction with standard ABI
protocols and base calling software; or other sequence analysis systems known in the art. Reading frames within the cDNA sequences were identified using standard methods (reviewed in Ausubel, 1997, supra, unit 7.7). Some of the cDNA sequences were selected for extension using the techniques disclosed in Example VIII.
The polynucleotide sequences derived from Incyte cDNAs were validated by removing vector, linker, and poly(A) sequences and by masking ambiguous bases, using algorithms and programs based on BLAST, dynamic programming, and dinucleotide nearest neighbor analysis. The Incyte cDNA sequences or translations thereof were then queried against a selection of public databases such as the GenBank primate, rodent, mammalian, vertebrate, and eukaryote databases, and BLOCKS, PRINTS, DOMO, PRODOM, and hidden Markov model (HMM)-based protein family databases such as PFAM. (HMM is a probabilistic approach which analyzes consensus primary structures of gene families. See, for example, Eddy, S.R. (1996) Curr. Opin.
Struct. Biol. 6:361-365.) The queries were performed using programs based on BLAST, FASTA, BLIMPS, and HMMER.
The Incyte cDNA sequences were assembled to produce full length polynucleotide sequences.
Alternatively, GenBank cDNAs, GenBank ESTs, stitched sequences, stretched sequences, or Genscan-predicted coding sequences (see Examples IV and V) were used to extend Incyte cDNA
assemblages to full length. Assembly was performed using programs based on Phred, Phrap, and Consed, and cDNA assemblages were screened for open reading frames using programs based on GeneMark, BLAST, and FASTA. The full length polynucleotide sequences were translated to derive the corresponding full length polypeptide sequences. Alternatively, a polypeptide of the invention may begin at any of the methionine residues of the full length translated polypeptide. Full length polypeptide sequences were subsequently analyzed by querying against databases such as the GenBank protein databases (genpept), SwissProt, BLOCKS, PRINTS, DOMO, PRODOM, Prosite, and hidden Markov model (HMM)-based protein family databases such as PFAM. Full length polynucleotide sequences are also analyzed using MACDNASIS PRO software (Hitachi Software Engineering, South San Francisco CA) and LASERGENE software (DNASTAR). Polynucleotide and polypeptide sequence alignments are generated using default parameters specified by the CLUSTAL algorithm as incorporated into the MEGALIGN multisequence alignment program (DNASTAR), which also calculates the percent identity between aligned sequences.
Table 7 summarizes the tools, programs, and algorithms used for the analysis and assembly of Incyte cDNA and full length sequences and provides applicable descriptions, references, and threshold parameters. The first column of Table 7 shows the tools, programs, and algorithms used, the second column provides brief descriptions thereof, the third column presents appropriate references, all of which are incorporated by reference herein in their entirety, and the fourth column presents, where applicable, the scores, probability values, and other parameters used to evaluate the strength of a match between two sequences (the higher the score or the lower the probability value, the greater the identity between two sequences).
The programs described above for the assembly and analysis of full length polynucleotide and polypeptide sequences were also used to identify polynucleotide sequence fragments from SEQ ID
NO:S-8. Fragments from about 20 to about 4000 nucleotides which are useful in hybridization and amplification technologies are described in Table 4, column 4.
IV. Identification and Editing of Coding Sequences from Genomic DNA
Putative aminoacyl tRNA synthetases were initially identified by running the Genscan gene identification program against public genomic sequence databases (e.g., gbpri and gbhtg). Genscan is a general-purpose gene identification program which analyzes genomic DNA
sequences from a variety of organisms (See Burge, C. and S. Karlin (1997) J. Mol. Biol. 268:78-94, and Burge, C. and S. Karlin (1998) Curr. Opin. Struct. Biol. 8:346-354). The program concatenates predicted exons to form an assembled cDNA sequence extending from a methionine to a stop codon.
The output of Genscan is a FASTA database of polynucleotide and polypeptide sequences. The maximum range of sequence for Genscan to analyze at once was set to 30 kb. To determine which of these Genscan predicted cDNA sequences encode aminoacyl tRNA synthetases, the encoded polypeptides were analyzed by querying against PFAM models for aminoacyl tRNA synthetases.
Potential aminoacyl tRNA synthetases were also identified by homology to Incyte cDNA sequences that had been annotated as aminoacyl tRNA synthetases. These selected Genscan-predicted sequences were then compared by BLAST analysis to the genpept and gbpri public databases. Where necessary, the Genscan-predicted sequences were then edited by comparison to the top BLAST
hit from genpept to correct errors in the sequence predicted by Genscan, such as extra or omitted exons. BLAST
analysis was also used to find any Incyte cDNA or public cDNA coverage of the Genscan-predicted sequences, thus providing evidence for transcription. When Incyte cDNA
coverage was available, this information was used to correct or confirm the Genscan predicted sequence. Full length polynucleotide sequences were obtained by assembling Genscan-predicted coding sequences with Incyte cDNA sequences and/or public cDNA sequences using the assembly process described in Example III. Alternatively, full length polynucleotide sequences were derived entirely from edited or unedited Genscan-predicted coding sequences.
V. Assembly of Genomic Sequence Data with cDNA Sequence Data "Stitched" Sequences Partial cDNA sequences were extended with exons predicted by the Genscan gene identification program described in Example IV. Partial cDNAs assembled as described in Example ITI were mapped to genomic DNA and parsed into clusters containing related cDNAs and Genscan exon predictions from one or more genomic sequences. Each cluster was analyzed using an algorithm based on graph theory and dynamic programming to integrate cDNA and genomic information, generating possible splice variants that were subsequently confirmed, edited, or extended to create a full length sequence. Sequence intervals in which the entire length of the interval was present on more than one sequence in the cluster were identified, and intervals thus identified were considered to be equivalent by transitivity. For example, if an interval was present on a cDNA and two genomic sequences, then all three intervals were considered to be equivalent. This process allows unrelated but consecutive genomic sequences to be brought together, bridged by cDNA
sequence. Intervals thus identified were then "stitched" together by the stitching algorithm in the order that they appear along their parent sequences to generate the longest possible sequence, as well as sequence variants.
Linkages between intervals which proceed along one type of parent sequence (cDNA to cDNA or genomic sequence to genomic sequence) were given preference over linkages which change parent type (cDNA to genomic sequence). The resultant stitched sequences were translated and compared by BLAST analysis to the genpept and gbpri public databases. Incorrect exons predicted by Genscan were corrected by comparison to the top BLAST hit from genpept. Sequences were further extended with additional cDNA sequences, or by inspection of genomic DNA, when necessary.
"Stretched" Se9uences Partial DNA sequences were extended to full length with an algorithm based on BLAST
analysis. First, partial cDNAs assembled as described in Example III were queried against public databases such as the GenBankprimate, rodent, mammalian, vertebrate, and eukaryote databases using the BLAST program. The nearest GenBank protein homolog was then compared by BLAST
analysis to either Incyte cDNA sequences or GenScan exon predicted sequences described in Example IV. A chimeric protein was generated by using the resultant high-scoring segment pairs (HSPs) to map the translated sequences onto the GenB ank protein homolog.
Insertions or deletions may occur in the chimeric protein with respect to the original GenB ank protein homolog. The GenBank protein homolog, the chimeric protein, or both were used as probes to search for homologous genomic sequences from the public human genome databases. Partial DNA
sequences were therefore "stretched" or extended by the addition of homologous genomic sequences. The resultant stretched sequences were examined to determine whether it contained a complete gene.
VI. Chromosomal Mapping of ATRS Encoding Polynucleotides The sequences which were used to assemble SEQ ID N0:5-8 were compared with sequences from the Incyte LIFESEQ database and public domain databases using BLAST and other implementations of the Smith-Waterman algorithm. Sequences from these databases that matched SEQ ID NO:S-8 were assembled into clusters of contiguous and overlapping sequences using assembly algorithms such as Phrap (Table 7). Radiation hybrid and genetic mapping data available from public resources such as the Stanford Human Genome Center (SHGC), Whitehead Institute for Genome Research (WIGR), and Genethon were used to determine if any of the clustered sequences had been previously mapped. Inclusion of a mapped sequence in a clustex resulted in the assignment of all sequences of that cluster, including its particular SEQ ID NO:, to that map location.
Map locations are represented by ranges, or intervals, of human chromosomes.
The map l0 position of an interval, in centiMorgans, is measured relative to the terminus of the chromosome's p-arm. (The centiMorgan (cM) is a unit of measurement based on recombination frequencies between chromosomal markers. On average, 1 cM is roughly equivalent to 1 megabase (Mb) of DNA in humans, although this can vary widely due to hot and cold spots of recombination.) The cM distances are based on genetic markers mapped by Genethon which provide boundaries for radiation hybrid markers whose sequences were included in each of the clusters. Human genome maps and other resources available to the public, such as the NCBI "GeneMap'99" World Wide Web site (http://www.ncbi.nim.nih.gov/genemap~, can be employed to determine if previously identified disease genes map within or in proximity to the intervals indicated above.
In this manner, SEQ ID NO:S was mapped to chromosome 12 within the interval from 97.1 to 116.6 centiMorgans.
VII. Analysis of Polynucleotide Expression Northern analysis is a laboratory technique used to detect the presence of a transcript of a gene and involves the hybridization of a labeled nucleotide sequence to a membrane on which RNAs from a particular cell type or tissue have been bound. (See, e.g., Sambrook, su ra, ch. 7; Ausubel (1995) supra, ch. 4 and 16.) Analogous computer techniques applying BLAST were used to search for identical or related molecules in cDNA databases such as GenBank or LIFESEQ (Incyte Genomics). This analysis is much faster than multiple membrane-based hybridizations. In addition, the sensitivity of the computer search can be modified to determine whether any particular match is categorized as exact or similar.
The basis of the search is the product score, which is defined as:

BLAST Score x Percent Identity x minimum {length(Seq. 1), length(Seq. 2)}
The product score takes into account both the degree of similarity between two.sequences and the 5 length of the sequence match. The product score is a normalized value between 0 and 100, and is calculated as follows: the BLAST score is multiplied by the percent nucleotide identity and the product is divided by (5 times the length of the shorter of the two sequences). The BLAST score is calculated by assigning a wore of +5 for every base that matches in a high-scoring segment pair (HSP), and -4 for every mismatch. Two sequences may share more than one HSP
(separated by gaps). If there is more than one HSP, then the pair with the highest BLAST
score is used to calculate the product score. The product score represents a balance between fractional overlap and quality in a BLAST alignment. For example, a product score of 100 is produced only for 100%
identity over the entire length of the shorter of the two sequences being compared. A product score of 70 is produced either by 100% identity and 70% overlap at one end, or by 88% identity and 100% overlap at the other. A product score of 50 is produced either by 100% identity and 50%
overlap at one end, or 79%
identity and 100% overlap.
Alternatively, polynucleotide sequences encoding ATRS are analyaed with respect to the tissue sources from which they were derived. For example, some full length sequences are assembled, at least in part, with overlapping Incyte cDNA sequences (see Example III). Each cDNA
sequence is derived from a cDNA library constructed from a human tissue. Each human tissue is classified into one of the following organ/tissue categories: cardiovascular system; connective tissue;
digestive system; embryonic structures; endocrine system; exocrine glands;
genitalia, female; genitalia, male; germ cells; hemic and immune system; liver; musculoskeletal system;
nervous system;
pancreas; respiratory system; sense organs; skin; stomatognathic system;
unclassified/mixed; or urinary tract. The number of libraries in each category is counted and divided by the total number of libraries across all categories. Similarly, each human tissue is classified into one of the following disease/condition categories: cancer, cell line, developmental, inflammation, neurological, trauma, cardiovascular, pooled, and other, and the number of libraries in each category is counted and divided by the total number of libraries across all categories. The resulting percentages reflect the tissue- and disease-specific expression of cDNA encoding ATRS. cDNA sequences and eDNA
library/tissue information are found in the LIF'ESEQ GOLD database (Incyte Genomics, Palo Alto CA).
VIII. Extension of ATRS Encoding Polynucleotides Full length polynucleotide sequences were also produced by extension of an appropriate fragment of the full length molecule using oligonucleotide primers designed from this fragment. One primer was synthesized to initiate 5' extension of the known fragment, and the other primer was synthesized to initiate 3' extension of the known fragment. The initial primers were designed using OLIGO 4.06 software (National Biosciences), or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the target sequence at temperatures of about 68 °C to about 72°C. Any stretch of nucleotides which would result in hairpin structures and primer-primer dimerizations was avoided.
Selected human cDNA libraries were used to extend the sequence. If more than one extension was necessary or desired, additional or nested sets of primers were designed.
High fidelity amplification was obtained by PCR using methods well known in the art. PCR
was performed in 96-well plates using the PTC-200 thermal cycler (MJ Research, Inc.). The reaction mix contained DNA template, 200 nmol of each primer, reaction buffer containing Mg2+, (NH4)2SO4, and 2-mercaptoethanol, Taq DNA polymerase (Amersham Pharmacia Biotech), ELONGASE
enzyme (Life Technologies), and Pfu DNA polymerase (Stratagene), with the following parameters for primer pair PCI A and PCI B : Step 1: 94 ° C, 3 min; Step 2: 94 ° C, 15 sec; Step 3 : 60 ° C, 1 min;
Step 4: 68°C, 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68°C, 5 min; Step 7: storage at 4°C. In the alternative, the parameters for primer pair T7 and SK+
were as follows: Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 57°C, 1 min; Step 4:
68°C, 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68°C, 5 min; Step 7: storage at 4°C.
The concentration of DNA in each well was determined by dispensing 100 ~l PICOGREEN
quantitation reagent (0.25% (vlv) PICOGREEN; Molecular Probes, Eugene OR) dissolved in 1X TE
and 0.5 ~1 of undiluted PCR product into each well of an opaque fiuorimeter plate (Corning Costar, Acton MA), allowing the DNA to bind to the reagent. The plate was scanned in a Fluoroskan II
(Labsystems Oy, Helsinki, Finland) to measure the fluorescence of the sample and to quantify the concentration of DNA. A 5 ,u1 to 10 ~1 aliquot of the reaction mixture was analyzed by electrophoresis on a 1 % agarose gel to determine which reactions were successful in extending the sequence.
The extended nucleotides were desalted and concentrated, transferred to 384-well plates, digested with CviJI cholera virus endonuclease (Molecular Biology Research, Madison WI), and sonicated or sheared prior to religation into pUC 18 vector (Amersham Pharmacia Biotech). For shotgun sequencing, the digested nucleotides were separated on low concentration (0.6 to 0.8%) agarose gels, fragments were excised, and agar digested with Agar ACE
(Promega). Extended clones were relegated using T4 lipase (New England Biolabs, Beverly MA) into pUC 18 vector (Amersham Pharmacia Biotech), treated with Pfu DNA polymerase (Stratagene) to fill-in restriction site overhangs, and transfected into competent E. coli cells. Transformed cells were selected on antibiotic-containing media, and individual colonies were picked and cultured overnight at 37°C in 384-well plates in LBl2x carb liquid media.
The cells were lysed, and DNA was amplified by PCR using Taq DNA polymerase (Amersham Pharmacia Biotech) and Pfu DNA polymerase (Stratagene) with the following parameters: Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3:
60°C, 1 min; Step 4: 72°C, 2 min; Step 5: steps 2, 3, and 4 repeated 29 times; Step 6: 72°C, 5 min; Step 7:
storage at 4°C. DNA was quantified by PICOGREEN reagent (Molecular Probes) as described above. Samples with low DNA
recoveries were reamplified using the same conditions as described above.
Samples were diluted with 20% dimethysulfoxide (1:2, vlv), and sequenced using DYENAMIC energy transfer sequencing primers and the DYENAMIC DIRECT kit (Amersham Pharmacia Biotech) or the ABI
PRISM
BIGDYE Terminator cycle sequencing ready reaction kit (Applied Biosystems).
In like manner, full length polynucleotide sequences are verified using the above procedure or are used to obtain 5'regulatory sequences using the above procedure along with oligonucleotides designed for such extension, and an appropriate genomic library.
IX. Labeling and Use of Individual Hybridization Probes Hybridization probes derived from SEQ ID N0:5-8 are employed to screen cDNAs, genomic DNAs, or mRNAs. Although the labeling of oligonucleotides, consisting of about 20 base pairs, is specifically described, essentially the same procedure is used with larger nucleotide fragments.
Oligonucleotides are designed using state-of the-art software such as OLIGO
4.06 software (National Biosciences) and labeled by combining 50 pmol of each oligomer, 250 ~cCi of [y 32P] adenosine triphosphate (Amersham Pharmacia Biotech), and T4 polynucleotide kinase (DuPont NEN, Boston MA). The labeled oligonucleotides are substantially purified using a SEPHADEX
G-25 superfine size exclusion dextran bead column (Amersham Pharmacia Biotech). An aliquot containing 10' counts per minute of the labeled probe is used in a typical membrane-based hybridization analysis of human genomic DNA digested with one of the following endonucleases: Ase I, Bgl II, Eco RI, Pst I, Xba I, or Pvu II (DuPont NEN).
The DNA from each digest is fractionated on a 0.7% agarose gel and transferred to nylon membranes (Nytran Plus, Schleicher & Schuell, Durham NH). Hybridization is carried out for 16 hours at 40°C. To remove nonspecific signals, blots are sequentially washed at room temperature under conditions of up to, for example, 0.1 x saline sodium citrate and 0.5%
sodium dodecyl sulfate.
Hybridization patterns are visualized using autoradiography or an alternative imaging means and compared.
X. Microarrays The linkage or synthesis of array elements upon a microarray can be achieved utilizing photolithography, piezoelectric printing (ink jet printing, See, e.g., Baldeschweiler, supra.), mechanical microspotting technologies, and derivatives thereof. The substrate in each of the aforementioned technologies should be uniform and solid with a non-porous surface (Schena (1999), supra).
Suggested substrates include silicon, silica, glass slides, glass chips, and silicon wafers. Alternatively, a procedure analogous to a dot or slot blot may also be used to arrange and link elements to the surface of a substrate using thermal, UV, chemical, or mechanical bonding procedures.
A typical array may be produced using available methods and machines well known to those of ordinary skill in the art and may contain any appropriate number of elements. (See, e.g., Schena, M. et al.
(1995) Science 270:467-470; Shalom D. et al. (1996) Genome Res. 6:639-645; Marshall, A. and J. Hodgson (1998) Nat. Biotechnol. 16:27-31.) Full length cDNAs, Expressed Sequence Tags (ESTs), or fragments or oligomers thereof may comprise the elements of the microarray. Fragments or oligomers suitable for hybridization can be selected using software well known in the art such as LASERGENE software (DNASTAR). The array elements are hybridized with polynucleotides in a biological sample. The polynucleotides in the biological sample are conjugated to a fluorescent label or other molecular tag for ease of detection. .
After hybridization, nonhybridized nucleotides from the biological sample are removed, and a fluorescence scanner is used to detect hybridization at each array element.
Alternatively, laser desorbtion and mass spectrometry may be used for detection of hybridization.
The degree of complementarity and the relative abundance of each polynucleotide which hybridizes to an element on the microarray may be assessed. In one embodiment, microarray preparation and usage is described in detail below.
Tissue or Cell Sample Preparation Total RNA is isolated from tissue samples using the guanidinium thiocyanate method and poly(A)+ RNA is purified using the oligo-(dT) cellulose method. Each poly(A)+
RNA sample is reverse transcribed using MMLV reverse-transcriptase, 0.05 pg/pl oligo-(dT) primer (2lmer), 1X first strand buffer, 0.03 units/pl RNase inhibitor, 500 pM dATP, 500 pM dGTP, 500 pM
dTTP, 40 pM
dCTP, 40 pM dCTP-Cy3 (BDS) or dCTP-Cy5 (Amersham Pharmacia Biotech). The reverse transcription reaction is performed in a 25 ml volume containing 200 ng poly(A)+ RNA with GEMBRIGHT kits (Incyte). Specific control poly(A)+ RNAs are synthesized by in vitro transcription from non-coding yeast genomic DNA. After incubation at 37° C for 2 hr, each reaction sample (one with Cy3 and another with Cy5 labeling) is treated with 2.5 ml of 0.5M sodium hydroxide and incubated for 20 minutes at 85° C to the stop the reaction and degrade the RNA. Samples are purified using two successive CHROMA SPIN 30 gel filtration spin columns (CLONTECH
Laboratories, Inc.
(CLONTECH), Palo Alto CA) and after combining, both reaction samples are ethanol precipitated using 1 ml of glycogen (1 mg/ml), 60 ml sodium acetate, and 300 ml of 100%
ethanol. The sample is then dried to completion using a SpeedVAC (Savant Instruments Inc., Holbrook NY) and resuspended in 14 ~tl 5X SSC/0.2% SDS.
Microarray Preparation Sequences of the present invention are used to generate array elements. Each array element is amplified from bacterial cells containing vectors with cloned cDNA inserts.
PCR amplification uses primers complementary to the vector sequences flanking the cDNA insert. Array elements are amplified in thirty cycles of PCR from an initial quantity of 1-2 ng to a final quantity greater than 5 ~ g.
Amplified array elements are then purified using SEPHACRYL-400 (Amersham Pharmacia Biotech).
Purified array elements are immobilized on polymer-coated glass slides. Glass microscope slides (Corning) are cleaned by ultrasound in 0.1 % SDS and acetone, with extensive distilled water washes between and after treatments. Glass slides are etched in 4%
hydrofluoric acid (VWR
Scientific Products Corporation (VWR), West Chester PA), washed extensively in distilled water, and coated with 0.05% aminopropyl silane (Sigma) in 95% ethanol. Coated slides are cured in a 110°C
oven.
Array elements are applied to the coated glass substrate using a procedure described in US
Patent No. 5,807,522, incorporated herein by reference. 1 ~1 of the array element DNA, at an average concentration of 100 ng/~1, is loaded into the open capillary printing element by a high-speed robotic apparatus. The apparatus then deposits about 5 n1 of array element sample per slide.
Microarrays are UV-crosslinked using a STRATALINI~ER UV-crosslinker (Stratagene).
Microarrays are washed at room temperature once in 0.2% SDS and three times in distilled water.
Non-specific binding sites are blocked by incubation of microarrays in 0.2%
casein in phosphate buffered saline (PBS) (Tropix, Inc., Bedford MA) for 30 minutes at 60°
C followed by washes in 0.2%
SDS and distilled water as before.
Hybridization Hybridization reactions contain 9 ~l of sample mixture consisting of 0.2 ~g each of Cy3 and Cy5 labeled cDNA synthesis products in 5X SSC, 0.2% SDS hybridization buffer.
The sample mixture is heated to 65° C for 5 minutes and is aliquoted onto the microarray surface and covered with an 1.8 cm2 coverslip. The arrays are transferred to a waterproof chamber having a cavity just slightly larger than a microscope slide. The chamber is kept at 100% humidity internally by the addition of 140 ~ 1 of 5X SSC in a corner of the chamber. The chamber containing the arrays is incubated for about 6.5 hours at 60° C. The arrays are washed for 10 min at 45° C in a first wash buffer (1X SSC, 0.1 %
SDS), three times for 10 minutes each at 45° C in a second wash buffer (0.1X SSC), and dried.
Detection Reporter-labeled hybridization complexes are detected with a microscope equipped with an Innova 70 mixed gas 10 W laser (Coherent, Inc., Santa Clara CA) capable of generating spectral lines at 488 nm for excitation of Cy3 and at 632 nm for excitation of CyS. The excitation laser light is focused on the array using a 20X microscope objective (Nikon, Inc., Melville NY). The slide containing the array is placed on a computer-controlled X-Y stage on the microscope and raster-scanned past the objective. The 1.8 cm x 1.8 cm array used in the present example is scanned with a resolution of 20 micrometers.
In two separate scans, a mixed gas multiline laser excites the two fluorophores sequentially.
Emitted light is split, based on wavelength, into two photomultiplier tube detectors (PMT 81477, Hamamatsu Photonics Systems, Bridgewater NJ) corresponding to the two fluorophores. Appropriate filters positioned between the array and the photomultiplier tubes are used to filter the signals. The emission maxima of the fluorophores used are 565 nm for Cy3 and 650 nm for CyS. Each array is typically scanned twice, one scan per fluorophore using the appropriate filters at the laser source, although the apparatus is capable of recording the spectra from both fluorophores simultaneously.
The sensitivity of the scans is typically calibrated using the signal intensity generated by a cDNA control species added to the sample mixture at a known concentration. A
specific location on the array contains a complementary DNA sequence, allowing the intensity of the signal at that location to be correlated with a weight ratio of hybridizing species of 1:100,000. When two samples from different sources (e.g., representing test and control cells), each labeled with a different fluorophore, are hybridized to a single array for the purpose of identifying genes that are differentially expressed, the calibration is done by labeling samples of the calibrating cDNA with the two fluorophores and adding identical amounts of each to the hybridization mixture.
The output of the photomultiplier tube is digitized using a 12-bit RTI-835H
analog-to-digital (A/D) conversion board (Analog Devices, Inc., Norwood MA) installed in an IBM-compatible PC
computer. The digitized data are displayed as an image where the signal intensity is mapped using a linear 20-color transformation to a pseudocolor scale ranging from blue (low signal) to red (high.
signal). The data is also analyzed quantitatively. Where two different fluorophores are excited and measured simultaneously, the data are first corrected for optical crosstalk (due to overlapping emission spectra) between the fluorophores using each fluorophore's emission spectrum.
A grid is superimposed over the fluorescence signal image such that the signal from each spot is centered in each element of the grid. The fluorescence signal within each element is then integrated to obtain a numerical value corresponding to the average intensity of the signal. The software used for signal analysis is the GEMTOOLS gene expression analysis program (Incyte).
XI. Complementary Polynucleotides Sequences complementary to the ATRS-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occurring ATRS.
Although use of oligonucleotides comprising from about 15 to 30 base pairs is described, essentially the same procedure is used with smaller or with larger sequence fragments. Appropriate oligonucleotides are designed using OLIGO 4.06 software (National Biosciences) and the coding sequence of ATRS. To inhibit transcription, a complementary oligonucleotide is designed from the most unique 5' sequence and used to prevent promoter binding to the coding sequence. To inhibit translation, a complementary oligonucleotide is designed to prevent ribosomal binding to the ATRS-encoding transcript.
XII. Expression of ATRS
Expression and purification of ATRS is achieved using bacterial or virus-based expression systems. For expression of ATRS in bacteria, cDNA is subcloned into an appropriate vector containing an antibiotic resistance gene and an inducible promoter that directs high levels of cDNA
transcription. Examples of such promoters include, but are not limited to, the tf p-lac (tac) hybrid promoter and the TS or T7 bacteriophage promoter in conjunction with the lac operator regulatory element. Recombinant vectors are transformed into suitable bacterial hosts, e.g., BL21 (DE3).
Antibiotic resistant bacteria express ATRS upon induction with isopropyl beta-D-thiogalactopyranoside (IPTG). Expression of ATRS in eukaryotic cells is achieved by infecting insect or mammalian cell lines with recombinant Auto~raphica californica nuclear polyhedrosis virus (AcMNPV), commonly known as baculovirus. The nonessential polyhedrin gene of baculovirus is replaced with cDNA
encoding ATRS by either homologous recombination or bacterial-mediated transposition involving transfer plasmid intermediates. Viral infectivity is maintained and the strong polyhedrin promoter drives high levels of cDNA transcription. Recombinant baculovirus is used to infect ~odoptera fru~iperda (Sf9) insect cells in most cases, or human hepatocytes, in some cases. Infection of the latter requires additional genetic modifications to baculovirus. (See Engelhard, E.K. et al. (1994) Proc.
Natl. Acad. Sci. USA 91:3224-3227; Sandig, V. et al. (1996) Hum. Gene Ther.
7:1937-1945.) In most expression systems, ATRS is synthesized as a fusion protein with, e.g., glutathione S-transferase (GST) or a peptide epitope tag, such as FLAG or 6-His, permitting rapid, single=step, affinity-based purification of recombinant fusion protein from crude cell lysates. GST, a 26-kilodalton enzyme from Schistosoma Lponicum, enables the purification of fusion proteins on immobilized glutathione under conditions that maintain protein activity and antigenicity (Amersham Pharmacia Biotech). Following purification, the GST moiety can be proteolytically cleaved from ATRS at specifically engineered sites. FLAG, an 8-amino acid peptide, enables immunoaffinity purification using commercially available monoclonal and polyclonal anti-FLAG antibodies (Eastman Kodak). 6-His, a stretch of six consecutive histidine residues, enables purification on metal-chelate resins (QIAGEN). Methods for protein expression and purification are discussed in Ausubel (1995, sera, ch. 10 and 16). Purified ATRS obtained by these methods can be used directly in the assays shown in Examples XVI, XVII, and XVIII, where applicable.
XIII. Functional Assays o ATRS function is assessed by expressing the sequences encoding ATRS at physiologically elevated levels in mammalian cell culture systems. cDNA is subcloned into a mammalian expression vector containing a strong promoter that drives high levels of cDNA
expression. Vectors of choice include PCMV SPORT (Life Technologies) and PCR3.1 (Invitrogen, Carlsbad CA), both of which contain the cytomegalovirus promoter. 5-10 ~g of recombinant vector are transiently transfected into a human cell line, for example, an endothelial or hematopoietic cell line, using either liposome formulations or electroporation. 1-2 ~c.g of an additional plasmid containing sequences encoding a marker protein are co-transfected. Expression of a marker protein provides a means to distinguish transfected cells from nontransfected cells and is a reliable predictor of cDNA expression from the recombinant vector. Marker proteins of choice include, e.g., Green Fluorescent Protein (GFP;
Clontech), CD64, or a CD64-GFP fusion protein. Flow cytometry (FCM), an automated, laser optics-based technique, is used to identify transfected cells expressing GFP or CD64-GFP and to evaluate the apoptotic state of the cells and other cellular properties. FCM detects and quantifies the uptake of fluorescent molecules that diagnose events preceding or coincident with cell death. These events include changes in nuclear DNA content as measured by staining of DNA with propidium iodide;
changes in cell size and granularity as measured by forward light scatter and 90 degree side light scatter; down-regulation of DNA synthesis as measured by decrease in bromodeoxyuridine uptake;
alterations in expression of cell surface and intracellular proteins as measured by reactivity with specific antibodies; and alterations in plasma membrane composition as measured by the binding of fluorescein-conjugated Annexin V protein to the cell surface. Methods in flow cytometry are discussed in Ormerod, M.G. (1994) Flow Cytometry, Oxford, New York NY.
The influence of ATRS on gene expression can be assessed using highly purified populations of cells transfected with sequences encoding ATRS and either CD64 or CD64-GFP.
CD64 and CD64-GFP are expressed on the surface of transfected cells and bind to conserved regions of human immunoglobulin G (IgG). Transfected cells are efficiently separated from nontransfected cells using magnetic beads coated with either human IgG or antibody against CD64 (DYNAL, Lake Success NY). mRNA can be purified from the cells using methods well known by those of skill in the art.
Expression of mRNA encoding ATRS and other genes of interest can be analyzed by northern analysis or microarray techniques.
XIV. Production of ATRS Specific Antibodies ATRS substantially purified using polyacrylamide gel electrophoresis (PAGE;
see, e.g., Harrington, M.G. (1990) Methods Enzymol. 182:488-495), or other purification techniques, is used to . immunize rabbits and to produce antibodies using standard protocols.
Alternatively, the ATRS amino acid sequence is analyzed using LASERGENE
software (DNASTAR) to determine regions of high immunogenicity, and a corresponding oligopeptide is synthesized and used to raise antibodies by means known to those of skill in the art. Methods for selection of appropriate epitopes, such as those near the C-terminus or in hydrophilic regions are well described in the art. (See, e.g., Ausubel, 1995, supra, ch. 11.) Typically, oligopeptides of about 15 residues in length are synthesized using an ABI 431A
peptide synthesizer (Applied Biosystems) using FMOC chemistry and coupled to KLH (Sigma-Aldrich, St. Louis MO) by reaction with N-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS) to increase immunogenicity. (See, e.g., Ausubel, 1995, supra.) Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant. Resulting antisera are tested for antipeptide and anti-ATRS activity by, for example, binding the peptide or ATRS to a substrate, blocking with 1 % BSA, reacting with rabbit antisera, washing, and reacting with radio-iodinated goat anti-rabbit IgG.
XV. Purification of Naturally Occurring ATRS Using Specific Antibodies Naturally occurring or recombinant ATRS is substantially purified by immunoaffinity chromatography using antibodies specific for ATRS. An immunoaffinity column is constructed by covalently coupling anti-ATRS antibody to an activated chromatographic resin, such as CNBr-activated SEPHAROSE (Amersham Pharmacia Biotech). After the coupling, the resin is blocked and washed according to the manufacturer's instructions.
Media containing ATRS are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of ATRS (e.g., high.ionic strength buffers in the presence of detergent). The column is eluted under conditions that disrupt antibody/ATRS binding (e.g., a buffer of pH 2 to pH 3, or a high concentration of a chaotxope, such as urea or thiocyanate ion), and ATRS is collected.
XVI. Identification of Molecules Which Interact with ATRS
ATRS, or biologically active fragments thereof, are labeled with 1~I Bolton-Hunter reagent.
(See, e.g., Bolton A.E. and W.M. Hunter (1973) Biochem. J. 133:529-539.) Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled ATRS, washed, and any wells with labeled ATRS complex are assayed. Data obtained using different concentrations of ATRS are used to calculate values for the number, affinity, and association of ATRS with the candidate molecules.
Alternatively, molecules interacting with ATRS are analyzed using the yeast two-hybrid system as described in Fields, S. and O. Song ( 1989) Nature 340:245-246, or using commercially available kits based on the two-hybrid system, such as the MATCHMAKER system (Clontech).
ATRS may also be used in the PATHCALLING process (CuraGen Corp., New Haven CT) which employs the yeast two-hybrid system in a high-throughput manner to determine all interactions between the proteins encoded by two large libraries of genes (Nandabalan, K.
et al. (2000) U.S.
Patent No. 6,057,101).
XVII. Demonstration of ATRS Activity tRNA synthetase activity is measured as the azninoacylation of a substrate tRNA in the presence of ['4C]-labeled amino acid. ATRS is incubated with ['4C]-labeled amino acid and the appropriate cognate tRNA (for example, [14C]alanine and tRNA~'a) in a buffered solution. '4C-labeled product is separated from free [14C]amino acid by chromatography, and the incorporated 14C
is quantified by scintillation counter. The amount of 14C-labeled product detected is proportional to the activity of ATRS in this assay.
XVIII. Identification of ATRS Agonists and Antagonists Agonists or antagonists of ATRS activation or inhibition may be tested using the assay described in section XVII. Agonists cause an increase in ATRS activity and antagonists cause a decrease in ATRS activity.
Various modifications and variations of the described methods and systems of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention.
Although the invention has been described in connection with certain embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments.
Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in molecular biology or related fields are intended to be within the scope of the following claims.

A

H

N

h O a-Ir1c-I.-I

N POalWPO

r-I UU UU

U N~1~.-I

N r1l0l~N
~

.N 01l~L~N
~

~y cPt17tnN

U Ll C~M
r-I

~'.,Lt1d~d~M
O

H d~L~l~
W

N

J~

O

4l O

r~
''~.,' U

H

r-I
O<

O
W

P.i Gn1~l~~
C!~

q H

r '~l ~l~l~l~l .t->UU UU

S~ N~ l0r1 O ~-Il0l~N
N

.1~ 01l~L~N

~y cNLP1t!7N
~

U lI~L~M
r-I

i,''~d~d~M
O

H d~t~L~i.n W

N

-r1 O

1W, N
f-~

H

r-1 O~

O
W

W r-1N Md~
CQ

A

H

.4.1NLf)l0r1 N r1~Ol~N
U

-4-101L~L~N
N

~y d'i.f1InN
'n U LL lM
O

~i u7'd~d~M
?-I

H d~L~l~In W

I

O 1 ~ .1~

~
O

H U7 01 ~,''U1 ~ ~ H ~I 'C,' -a .1-~ ~ -rl r1 M W-1 c~ rtS .h U -~

Q) L~ 01 N .J~ (O tU O N (J) r1 .N

U M O 4-I ~-I U M G' r~l ~C ~ cd N
-r1 -r1 o1 CIA~,' o ~I O I ~', o N , ,.S~ r-I ~I
1~ U1 N Gl o~ ,.~

U7 N o0 ~-I N W ~,' N (d Ca r-I r-I ~
v-I .!-1 '~

N to ~ dP o1 N to N U1 N ~ -rl J.~
N P; N v N
~ ~ ~ ~ ~ N ~ ~
~
:

-I P U
s; U ?a N .-I O .!-' I
.~ U ~ O cti ttf ~
N N .u U o~ N .u U .L~ U1 N
O ~;

Ul Ul W c--I , C/~ W -~, ~ U Pa N
f~'7-~ .h -S-', -4 -I ~
~ 4 l ~ N - ~ W-I

~ O ~
1 f~

N 0 . .
m -I r . r.
- x r1 -~I o ~ -~I
cCi ~, W o, -I ~
-N .. o -~I ~ N .I-~ ~
~ ~ o -~i ~ o ~u ~f N m ~I -~I ~ W o ~ ~I U + N oo b~
-a o .h m ,.~ o, O O ~ .N a~ I .R ~S -r-Iar 0~
o, O O rd s~ 1n ~ .N ~
,-I

N .1..1 01 ~., I~ ~1 di N 10.I (CS c-I
M O1 ~., .!-~ ~I O L~ r-I I N I
.1-~ ~-, .1~

r1 O U .!, yo tn r1 U1 ~ U' .!~ ~-O U .!~ -ri ~,' O --I U~ r-I t~
ttf o~

~ cij cd O
~' c~ UI -rl ~--I ~- .~? $-1 ~,'' zS U! 1-~ 4J rt h .-IU1 N ,~ U1 N W6 b~ l-I U N ,-G U7 ' -.-1 r~; .I-~
> ~ oo 00 -' 1 W i O
~ O ~ W
'' l -I

N s O U7 ~, . r ~n i ~
~n N ~ O S N ~ .1--, , . rt ~I
H ~ ~ l g ..
a~ o ~ ~ a~
l c~ ~
~ tn ~I
FC m ~ C

>315 . r ~ 'J
, , N
r > ~ CO
f 1 ~ 1> >_ (d N (is UI ~I -r-1 ' X37 ~
U1 (S O (d U7 -r1 O y 1 N . y ~ L - fly ~ Ul ~ , !~ 1W' ~ . N
~ ~ 1 l0 N
U ~i ~ tti ~

r1 ca - - .
~ . . , r ~y -1~ N O
r O r-I
. a-I
.h -r-I .J_1 .1-1 U
fn -1~ -rl Q', .!.J
.1.) U U1 N U1 1=( O I .!-~ N r-i c>3 U1 U1 rt1 ~ 4-I
O .-I .I-W d -'-i U ~y o0 Ul ~y .u ,~ ~n 4.a ~I .I~ a~ a ~ o ~I . o ~I
m o ~C 4-I ~I .u N ~ I r, ~I
~ u ~
~ ~ o u ~ ~c a a o ~

o I u ~, z . o ~a ~n . I
x ~ ~, ~ o ~s m , .u ~a .H >~ U N r-I ~, c~ H ~ r~ . >~ m U ~ ~ U ~ us ~n ~ o ~y ~-I O U1 ~ U O i U7 Cl~ N ~-1 I S~' U ~ U N cd I m ~', O
C1rri ,.Sa,~',~ .=i ~ -r1 r1 O U (D -t~ U ~i r1 O -rl O . .-I r-I O U ~,' U7 ~, .-icti U O u~ ~-I ~ ~,' I ~,' N ~ .t~
U ~ N O U1 r-I ,.c,' oo N ~ .1-~
~,' N .-1 O N

td N ~1 O ,'~ ~y N U N ~ r1 ,.C, U7 O U7 O ",-~ ~r N U N ~ O N ~-1 cd U
'd L ,~,' ~;

W -1~~ N n-1 U N ~., O N ,5y N ~ O 'C3 ~-Ir1 U N O .i-1 O O N N w1 w ~ ~ ~ U U
~ ~ O
~ ~

O ~ ~ N ',~ -,' ~' ~-I N N s~ ~ ~ ca s-..' 5., ca O ~I M O of (d t7 U ~a ~ x ca ~ rn cn ~I ~ x w ;~ N U h ~ a3, x ~ ~ rn cn tn a~ h oa ~ ~s >~, .-I a~

-.-I~-I N

~ ~ O ~.,~
~, -r-II I , I
~-1 ~ ~ 00 lD M

N ~ l~ M N

O

M L~
N Lf1 Lf~

H ~ M d~

OD '-I Lt~ M
.'iLC7O r1 , tSl Pa ~ b~

N

O

H

H

N f-~q f~ Ca N U U U U
''d .1JN L(~ l0 v-1 -r1 ~r r1 to t~ N
-L~

U O1 l~ L~ N

N d~ ~ u7 N

H I t~ L M

~ u7 d~ di M
~y O

N

O

J-~

N -1 N M d~
y W

O
~

rr~ w x o ~ ~ q a ~n ~ l H w E-a N

a a x a w w m m I

~ w 1 ..

in x ~1 cd Gi ~ ~ ~~
m ~ ~ H

" N Q ~I H H N
I N ~ W E-~ O U] 'HI Lf7 Ul ~ ~ O ~ C7 E-~ ~ W N
~ i"

a~N : u~axwzwH~ ~"ax w ~,.uH , ~c~wxHNa ~ x~H H-~1~ , ~ I ~r I .u .u ~N~' H ca ..~Nw ~
q I

U ~ ., H :~ t U H m H ~ N E-~ U H ~ ~~ 1 N
(a 1 i4 r.(; ~ ~~ I
J-1 Ul Cr In W ~ W ~ E-~
N 1 '-I E-~ H 00 N ~ ' M

O Ul U1 r1 w-I x O Ul H P: O Ul U2 , P.~ '~Ul M
N d, fR U1 a O ~~ U1 ~,' O ~ M Ea ~ ~ U7 O
U1 ~ fYl ~' FC H
~,' ~ ~ O
N

r1 f~',(!S O H H CJ O W P.t 'H FC
Q'S 00 ' W U7 O FC O
b7 ~ ~ N
~

U U7 W H ~ a Pa r ~ U fa U ' ~ FC F4 a ~ ~n ' W

- .
-a C~

~~~n~ a ~ ~ ~

a a U r~ as w o w v ~ q ~

rn l I

?i P,' W W H P;
O
.i~

~ W H H W

cd ~ a ry . a J-~

~~q ~ W P

~ N r N ~ I M I 1 -I

d~ N - M

~

~ ~
'Z~ M H
~ ~ U

~I "I ' c,->

tn U .u ~ 1~ W ~ ~-I O q >~ ~-. I !-I ~ ~ a, ~ ,. ..
~; ' ' N . U W t U ~ o~ ~ O
~ ~

N -1~ r-I f~ r1 Ul Ul M -l-~
U7 v ' N OI ~I ~-, -,~
-r1 M ~ s~ M N . ~ W x ~I ~' ' ~ ra r~ I
a~ H ~ ~I -as ~ N W ~
~ ' I
' U I-I U bW .1~ ~ ~C H I 1-I cd ~ I I W ~-~
G d ~

S ~ O u1 ~ W H u1 N m l N ~o f~ ~

rt d, r ,~ H H OM FI,' Ul ~,' Ul ~i O .!-) r-I ~ '~ '-1 ~ ~ d~ J~ S=,' ~ ' W-I
~ N
A
~

b) l~ -r1 F( UI Lr ~ Ul O ~ (~
~ -I , O N O N W l - N l O ~
l l S ~
N ~
' -I

r r M r .nr ~, .!~ ~- , '~ U1 ~ -.
U7 U ~ ~ al y U ~-1 .1 ~ U
U7 '~-~ -r ~ Z
c0 U U7 W ~

O

O M

M

rd M
r1 -r1 M
,5y In .1-1 00 '~.I
.1-1 W
[7 in S~

O O1 l~
c--1 N
L~

-rI O1 O di N
N U7 c-I
l~
M
c-1 l0 .1-1r1 O l0 N
M d~
M
d~
c-1 d~
dW
-I

!f5 U1 N N M N
Ul N

U]
H
H
H
~

.-I H u~ try u~ ~o H

r1 di Ul N
,5y L~
c-I
w-I

L~
l0 (($ l~ M In 01 ~1 l0 d~ L~
Lt1 O
d~
N
ri -ri ~-I O N N LO

M
d~
Lf~
M
cH
Lf1 ~ u~u~u1u1u1EnE-~H a ~~
~
H

,~ r l nH

N w-I 01 ~ 01 U1 v-f N O

Ln OD
N

1.1 di O di l0 O N N O d~
.1-~d1 In O
N

O v-I W -I N N
,i,''N Lf7 01 ri M M I
M
dW
-I
di Lc w u1 v~ try w v~ v1 u1 u1 U1 m H
try v1 H
H
H

U

N

-W o ' ~ uo ~ !I
N

O I

i ~i N U U
-'~

'I'~N Lf) ~r m H L~ l~
~

0 ~

d~ L

~

q r1 N
~

rn U 'N~, O
°a ~ o w Ix~ wl wl r-~~
H W ~', ~,' U7 PO f~ W
..
H I N o~ to H U7 ~,' W .-. 4-I l ~ I CIA
N Lf~ N W Ol M l0 p', m ~a ~ ~'i ~ t~ao oo~,o-t W
~a .u -~.I . a~ ~I ~ a x a x N ~ (IS tf7 Ln .1J .t~ Pa M QI I [7 H
~rl ~° .~ H ~ I ~ I N N aC N '~ W W E-~ H LJ~ C7 r.C N
r-I N .1..~ -- O N u-i r-f ,.L~, O N U1 Q7 ~° ~o cn a a w ~ FC a W ,.~ in a ~.,.u ~I ~ ~ m ~I ~'~ r-I rC ~ x H
I ~H M II sJ ~ I .u.u ~~~ ~c.WN ~HU~
.. N ~ W ~ (d .u M 1 N .!-~ I W r.~ H ~ E-~ W ~ d~ '~
4-I ~ U1 ,'~ I ~I N O Tl7 cd '~ r-i ~', c~ ~ .~ x O W ~ H i-l 7 W O ~
.I-~ W N <;C m '-I O ~ ~ s~ "~ ~, .t~ s~ N oo E~ ~ E-~ ~n at ~ H a o ~
~ W ~ U cd ~ ~ D ~ ~ .u7 ~ ~ ~ U1 ~ W ~ ~ W W ~ ~ ~ O ~ ~
M
H
00 01 N l~ M
r1 01 O 61 OJ
N N d~ In tn L(7 In ~ ~ ~ l' H
~IMO~Hm r-I N M LfY tn L(7 Cl] U2 U] U1 N E-~
c-~1 M N d~ N H N
v-I M M l0 C~ N
u-I N M d~ 01 Ln M
~ ~ I~ In ~ (1 c-~
N
H
q U
M

x o U

o o <x a w ~

W
H

FC FC FC H ~

a a a o a r~ w r~ ~ x r~

I
p'..,l0 I 1 L~ N ~-I
, W O W a l0 1 r1 ~
N

~' ~

>n~n ~a~Wx = ~N a~~ A>n a W O ~(', ''~-, N ~ U1 I '~,' U1d~ ,5-t t!7 ~(,' O1 lay N I
1 Ln a (d ~ (d ~ H il~ ~-I Ul x ~(', M
~

Q,'lD W U] O W ~ 01 ,y.J.1J L~ ?-I
E-~mo H ~ l0 I ~L f($
E- E-~ E-~ o I 00 H N H
4 O W W ~ 1~ ~
N

~'p M N I .

1 N W O U7 ~ C-~ ~ ,.~ I N
W ~"H x o ~ H
W ry axw u~axwNwH w I -~Iw.u ..
H ~'~ w v~ ur~~
~ ~
wx ~
I

~ - r -oo v ~ o U H ~~ ''~' H ,.
d~ ~,- O M !d J R U
' t!~ ~ O~
E-~ U N N
C7 Ri -1 I~ , a~ . ..
w~Cw Hw~ra cn ~1 as zNrn~ -- ~
~~

O U1 ~~ Ul O U1 , Cl7In x O Ul H U7 U~ I O
W I N U!
c-I
I

~ H ~ ~
H ~t,' ~H C7 O '~$ ~ r1 FCO N~ Z H H O ,7y Z 01 ~ ~ ~ W ~
~ a ~
,~," ~
O a N

~~

~w~A~-I ~~aoaatr-~Nwr-~aAM~~-t.G.~ ~n Ur-~ wE-M

N

H

z cN t!7 N
Ln l4 O
N

~ M v-1 l0 l0 L~
O d~

v-I u-I
N N M M
d~ d~

.-I Ei tn H u1 uW
n E-~

O Ln N

L~ N l0 Ln 01 t~
CO r1 L(W -i r1 N N M M
di E-~ u1 E-~
tn u1 H
u1 H

n ~H

H

q U

I
H

N

N

N

M

M

N

di -~ -m as w I

a m H W
f~

O H
rx a w w ~.1 N ~ O N
-r1 ,'~ I
U7 U1 ~I
~ NI

~," 1 M U L~
N .!.J l0 c-I
U1 O lfl (~ U1 ~ Ul (~ C7 O W
IS ~
M
.1-~
J-~
~ -~
~-1 I N W ~-1 -.-1 r1 U' ?~ .~u Ei ~ FC
~ c~~ a1 ~ ~
~o Ca cd hl ~ W d~
S-1 d~ ~ W
f~ d~

-r~l (Y., E
FI,' O O N
Id M O
cdl ,1t ~ R;
~ I~.' N
r1 (~ N P,' ~1-1 U ~- H u~ W
~I w ~1 Ot w M

N

H

-rl l0 v-1InO01d1 r100 Od~L~t17 v-IM01d~LI701N ~H
~~ O ~~ O ~ '-1 .!~ N ~'~0101N01OO Nl0~~ 00M Md~W-Il0M r-IN L(7l~'~~ ppLC7 O

M N ~ 000001O v--IO ON ~~ d~d~Nc-I~L!)l0O L~O1Nl0~ ~ dia1 r1 U7 u-I W -Iu-W-Iv-IN Nw-I Nv-Ic-1c-I N Nr1Nv-Iw-Ic-I ,-I

O

W

~, O

r1 ~ 010001 M CO 01 M Ld't11N O1 M . M N O C~L~L~ O O 10~
~ OI -I -I f 0 lL l -h L r 00 c c ~ Lf~ 1 rn 01N 0 O r1 N 117 l0 v-I 01OL~r-I O s-I01L~N
L 00 N Ilk d~ d~ O OJ01-I
l n N N v MN N
-r ~ ~ N ~ ~ t~d~ ~ ~ O1N

U1 r1~-Ic--I r1 r1 N r1u-Iu-INr1 r1 O

W

O c-I d~00NO N Lf1 Nw1w-I MN r1M

r1O r-IO OO O N OO O OO O O

u1 H U HH HH ~ H ~H H HH H ~

.u ~ ~ OO OO H O OO W ~~ O O

N H H ~U U q ~ ZN HH Z ~

~aC- C_~ ~ C q f~ ~ uur ~-a ~ 7 7 ~ I 1 ~ ~ ~ o An o ~e a w~ a aw Hw a x a~m ~ ,-I ~-I

v v v --U,. --~

w ~-I ~-I~ ~-Ia ~ ~

l0c-1 l0l0 l0,~N ,'~,'W-Ic-I,5,~,5lW-I,W-Ic-1l0,~ c-ii-1l0v-I
c-I r1 w xN HH .~w ~~ o~ xx ~~ ~Iw h~ xx r~~ ~.~~cHx rxxH

U d~~~ d~c-IW-Ilf1r1d~M Ov-INv-ICOO Lf1M Odil0cH(3'.,p'.,01L~~-1Ol O O

0101~-1L~N L~N WH Nd~NLC7L~N O1Ln0001l0d~NO Lf101O N ~OO
,~Z., Z, N l0~O~-Il0N In~ L~01LI~l0r10100L~O00LC7M Ld~cHl0MN O InL~O

O l0Lf7c-IL~L~L~01d~010101M O101Nd~LflN Od~ON O01N r1NN
O L~d WM NM 01N OlOlMN 0101LC7M lbdiInL~00d~Oll0O d~NO
-I r1 c-I
P.,' p;

N L~c-Il0Ind~LOcftOO OO L~M OO c-Il!1Oc-IIn01l0v-1O'd~C~~ NL~
W W

Ll~ N l~~7r1r1d~c--IL~00l~L~L~l0C~L~l~M 00l~l0L(1c-IL~L~l0c-1N Mc---i ~-' v M

N O M L~

J-1 dW In O
~ -I H 00 U CO
-I

c c N I c-I u-I I
I

.-I ~-1 I I
N

ltI c-I w-i ~W

N

U
,.~

.S''.,O O ~H N
.N

N 00 ~-I L~

N r1 N N r1 a~
a as as x x N U U U U
O

.1-~N LC7 l0 r1 ~v -I l0 L~ N
~
PW

U o1 L~ I~ N
U
H

~, d~ Ln Lf7 N
~

H ~ L~ L M
~

LC7 d~ d~ M

r-1 d~ L~ ~ t1~

O' W

'~

O
'Z, r-I ~ l0 L~ 00 ~

UH

O<

W

O

~ c a ~ U ' ~ ?, U ~

~ 1-I -.- ~-I
~ i 'T5 ,~
~ (d U

~

N ~ N U2 -r-I 1~
N 1 ->

N '~ ~ ~
~

~-I ~.,O ~,' N
~ ~
~-I

N ctS U O .!~ S-I ',~
i .l~ ~ N

~ In o a~ -~I x o o ~n o r1 ~

1~f1 ~~-1~ ,.~ O U7 N
b1 r1T~J1 U
~,' -U2 ~.,'1-IU7 N N .h S-I
O . tiiU O

r-I ~ r1 U1 ,~
-ri ~ J-I ~1 N -rl -1~ ~-I !-IL~ N 'd ~ N
~ t~ . U N

~ -rl N ~-1 O N N
U .~.'-rl ,~

~-I '~.''N -L.) i~ U7 '~ U r1 J.) .h U~
?-I
, O ~ c0~ b7 cd .1~ U1 ca O cd ~

r1 S'-,c0U W N ~ .1~
f--,' ~ S-,' ~

~

M r1 ~iS~ ~" I 4-I O
Ul N U1 (!S Ul b1 ~

~i >=i O ~ (IS (~ r1 (I$ J..~.~., O r1 ;j (d N N .1~ U7 O O
-r1 ~-1~-I -.-i ''' N U
d ' r1 -rl O ( ~ C5 ~.(d N ~, O r1 ~, m c~ ,s~a~ 3 ~ -~I
~I ~ ~
U

~ at ~ ~ ~ .u v ~
~I ~ .u o ~ U ~ ~ O
N W

O U O O O ~-I
tr ~-I U
U c>3 ,5W-t N ~ ~-I
~-1 c6 ~-I
~ O
,-R

cd r1O U '~ cd N
r-I
'Jy O

U
~ ~

"~ 't3'~ (d -rl ~
~ r1 ''~ f', O

N''~'O N N -4~ r1 N
N'd O ~ U

.!-~ .t~.h ~1 O r1 J.-1 r--I N N r-1 -rl cd !~
~.,"

cd O UScd ~ U1 O
to f~U U D
~ cd .-I r-Ir-1 r-I -ri r1 I N (dr1 ~,' r-I N
U

O ~I O O ~-1 ,~ O
~-I .!-~O U1 -.-i ,~
cCj 1~

tn ~ ~ ~ ~ ~ ~ ~ :n ~ ~ ~

.-I -rl--I ''d 1.~ .-i N '~ O O ~ N cd 'z3 ~
~

o~ cd ~
S~

to U
.-i b7b7 ~ ~ O b1 O 4-I~ .~ ~
L5~
td '~
f ~' -, ''d~ G' S-~' -i O ~: G' U '~
"
'' .> .>~ -r-1 ., ~., .,-ri cIS -.-i ~ -ri1 ~y O
-r-I

-rl ~,' U1 O rf~U1 .-1 a U1 -.-I .~'.,.L-~ M r1 N
-l ,S,'' ~ ~ U1 O -rl ~
-r1U7 r-I

N O ,~.'cd U N ~ ai U1 ~ >~

~ O '~''~ U U7 ''C5 J-1 Ulr1 ''t~
~ N N

N O N N ;~ O ,.Q
r-I of C~ -I-1 .h 1~ r1 .h.4-1 (a ~-1 .1-~
O -rl CJ7c~ ~-I ~I -L,' ~', tt~

U ,~' U U U U r1 U
~', N b~ N N cd ~d ~,'~ ~-I ~ ~ r-1 ,5 ~
J-1 r1Cd U7 -r1 ~,' ~

O S-I ~-Irl f-I
N ctS ~I'~ ~ N U1 .1~ -~.
t51 -~I.u 3 .u.u ~-I ~ .u ~ >~ a~~ o ~ ~ ca -~

1JU1 O UlU1 O ~i p U1 ~ r-1 ,S~..nW -~ U

i S.,' ~'.,>-'., I O ~,' r1 -l,r1 TR ~
-rl ~'., ,5y (d -.-IO s: O O ~-I at O
.1~ ~ bW-1 cti ~

f-IU ~ U U cd ,.~ U
O ai ~ ~ N r1 O U

U 4-I O N ~r .1.~
-rl 1~ ~
W

U1U7 O U7U1 ',~, S~ U1 !-~ ~-IW r1 O O '~
(d ~1 a~ca r1 a~as I o 3 ai U ~u,~ ~.t c>3 r1 ~I

a 3 a~ 3 3 ~ U '~ 3 ~a 0 ~

a~ u~ ~ ~ tn U a~ I
~ ~a ~ ~ ~ ~, I ~ a~ a~ ~

~ ~''~ ~,?~ o -~ , a~ a~u ~a ~ ~n ~I
o r1 ~ as rt ~ ~-, as ~ a ~

I ~ td. cd ~ I s -.-I, N
cd -r1 aS U O U
~-I ~ ~ -f~

~I~I ~., ~-I~1 ~., >-r' ~1 ~ U ~'SS-I .-i ~ r1 N O

O ~ .R'Q O O U ,~
~-1 1~'~ U U1 ~-I

-rl~ rir-I ~-I ~-I r1 -rl .~RO 1=i .5~. o ~-i ri O
~ cCl a a 4-I a a 4-1 .~ a r1 O S=,' -r1 U ~' -ri 'ZS
U

E-i N p a ' 1 Z, O

a ~'' w a~

w M v-IM u-I

O O v-1 O O

.RO

o a a s x a ~ h, ~ i o ' I~

o p ~ ~ ~b ~, ~ ~
' N o i1 W M c~
I P O _N t., U
' ~ >
~ "o b ~ ~, ~ .N
~ 5 ~ y ~ 5 N ~ U
W

p ~ U II ~ ~ ~ U
~

O ~ "

~ O O ~ ~ N ~~
o c~ 5 cct ,.O
' E' ~ ..~~ W ~ ~ ~ 1 ~ ~
~ ~ O ~, o c~
"

cd .n.~ 7 C_i V w W N e v~
~ , ' 5 ~ ~
''"

O .- >, ~' H .
O ti ~ c~"~
~
m ~

U i-W' c ..~.r 1 ' C'~' ,y"OC
W ~~.C'O
~

~ N 'i i' O U ~ ~
N W ,~ M
~

(~ (~
~
~
~

W W O W Q,' a~ ~ P-W Y
~Y w GL w ~
~ ~

U
.b _ U
O O 00 .,-~i U cOC r -.
-; ~ U M U
-' 0 ~ 00 P-~ ~ ~ ~, ,--i N
" 4:
' M d' a U U U ~ .. ov ~ o ~ N
o c~ ~ ~ ~ c~ ~
~1, o a o b o.

n _ P-l N ~ ~ :~ ~ ~ W
~ M
O

U U U s~ ~ .
~ . o ~,.., s~ ~
~, ~,,r', a~
,-~ O U W wt O
s s ~ 1y ~ .r., cd r~ N
Ov ~ y H ~ N

~ . . ( ~ d. >, pp .
a ~ v O~ , ~
~ C/~ .a N N 'b M N~' ~

r U ~ ~ . a~ ,.o ~;
n O M . o , a~
O w w p~ . ~ t! N r~
w , O U ~', W ~ P~i I--0~0 ONO

~ v N :: U".~G~ ~
~~,~~,~, ~C/~ ~~
~

_ ~

~~ ~~ ~ ~~ ~b ' ~ '.~

~, >, >, c ' V ~! ~ c~
p., O cn ~ Q\ ~
' ~ O
'~

0 0 o ~ ~ v~ ~ ~' ,-~ ~
fs! U ~n o :
: ?
i ' vy-' ,b . v~ w 'n Pa Pa Pa ~ ~ N O O r' ~ d ~ x~' U
U

i~ U ~ , vw.~
f,. O 0 ~' ~~
p~'' ~

O~ ~ ~ ,.~
' ~ ~
~.C

a~'' ~ ~ ~ '' 'o -d ~ ' m ~ '~' ~ ~ R '~'' ~ ov ~ ~

f3~ ., ~ ~ ~ W Z ~ ~ ~ . ~
~ ~, , x ~ vi c~
~ rt c~ U ~
Z A
Z

o .c '~
o r~ ~ 'o ~ '~
o ~

>~ V O ~ .O y cpn cct N N ~
~

O 'G Q cH ~' ,~, cn ~ ~ U ~
' N E"~t-~ V c~
b G fl v~ Q a' N >~ L ~n y ~ L3 p .
a ~ c~

U~ ~~ ~ ~~U ~~pa~ U~~~ O c~CU

O ,-pO ~ ~ cC U c~C
,~ ~
, ~

ca U O O ~ Q ~ ~ ~ .i~ ~ ~
.'~ U ~
~ vi H O

...Ur~ U ..r ~ ~ 4r c~ ' ~ ,~ ~

~ 'y ~~ . o ~ c~ ~ U ~ ~
U (-~ ,~.s O ''~ 'o ~ ~ a~ ~ W ~ O >~
O
'1 "
~

CC ~ , 7-r ~ ~

>~ Cj O U
O ~ 'i"' ~
a by U y ~ cn ~ ~
'~

'~'.3' N ~~ ~ N ttt ~
it ' ~-' U ~ ~"
~

. .n O
, Q . .~, ~ N ~ U ~ ~ ~ O U
~ '~

O O ~ ~ ~--c O N b ~ ~ y U C N
U cH
y., c~

N ~ ~ ~ ' N ~ N N
~ N > .~ ~ r vi w ;~ ~ ~' ~ ~
~ a' A o w ~ 3 ~ ~ ~ ~o >~ ,~ >~ N 4-i .~ .
~ ~ ' U .~. ~ ~
a o ~ p m w ~ '~' ~
4.n O 0 'vW' .f.' 4=,~ ~ ~ O ~
U V
~ ~
' m ~ ai ~,.,ccf N a O ~ b0~0c~
a.~ O a' ~ O ~ c~ ~ ,b w ~ a'' ~ Y
U ~ -o ~

~ ~~~ . o,~ o A ~ s ~ d ~~~ ~ ~
~ ~ ~ ~ ~
A :~

~ ~ a ~ ~ .--~ ~ ~ s~, w ~ 4, H

U ~ e~ R

~ ~ ~ a a; ~ w m U ~
C7 ~, ni ~ '"; N
o ~ '~ ~o ' i ~c O
~ ~ ~i FW " ~ .o ° o ,~ o N ~
'LS ~ 'r ~ N M .., c'~ .O, '~ c'~ p '~ II r-i O N N
O ~ N by P~I Z ~ ~1 ~J ~ ~ CJ~
N
~ ~ ~ ~ 00 V1 01 ~O O ' f ~ ~ ~ i~ ~ ~ ~ ~ : ~ ~ v~ ,4 O
U~v~.~r 4; ~ ~ ~ ~ ~ ~ ~ .
.--'tea G ~ ~ G~U'.'~ ~ p ~ C/~ ~ y0 ø, rn W 0~1 v~ ~ ~ ~ >~ U .~ U ~ b0-0 U .-i V
.-n .~', °
~ ~L
_ U 7-y.' E:"i ~ ~ r..' ~ ~ i.., ~ c~ O
c~ N ~, ~ ~ ~ .r: O .N ~ C1 ~ °~ 4°-i ~ x .,~
~ N O N ~ O ~ ~ p Vj U ~ ~ ~ ~ ~ .~ ~ ~ ~ ~ ~ a Y U
o°°, o°°, ~ N ~ ~ oo ~ o; ° ~ ~ r '~
° ~ - ~ >, .~ '~ i o ~ v N ,°oh, ~ oo ~ N
O Cv ~ ~ ~ ~ h .~ ~ ~ ~ M ~ ~ M ~ .~ N .~ y U
c~G c~ ~~'~' N '_'a b0 ~r ~ N ~ ~ ~ a d' ~ m a N 'b vi " bA w i i Pa ~ .~ y ~ ~ N ~ ''~~ ''~ '~ .~ d -~ ~ i° W ~ °' y W ~ '~ ~ ~ ~ °' ~ _~ N '~ N ~ '~ ~ y °' P, N
~' '~ pa ono w ~ ~ N ~ ~ ~' U ~ ~ ~ ~ ~ N ~ ~ ~ C7 O O ~
U ~ x , ~x ~r ~~ ~''-' C7 E-a ~ ~ a~ ~ o o ~ ~ o ~ .~ .~ ° ~
.°~~° ~ o °i ~ ~n co .~ .~ a~ L7 ~ .ty ~ . v~ ~ as ~ ~, bo ,~ o °
~ .O ~ . ~ ay, .,J ~ .-~ ~n +-. G ~' i ~ ° 3 ,~ ~ '~ ~, ~ b ~ o ~ o ~ ~ M ° o o .~
~4 c7 C7 ,~ Z w ~o ,~ ~ ~ ~ ~ v~ C7 z .~ U w N w v~ U ~7 ~ ra ~ ~
,i c ~o '~ ~ a.
P. ~t7 ~ ~ °~ ~ ~ ~ .~ ~ N ~ ~ w V ~ ~ V~ ~ N o" a. ~ cW n ~ c~
a~ ~' .", b 'c~ ~~ ~~"~WZ b0 rUn'n '~U~O.., O~
U ~ W by b 'a~3 ~ ~ -~', yi.., ° "d . ~ ~' . .~ ~ .,~ U ~ U U .~ cG ~ U O
O ~ O P~
~b 4~., ~ a.
P.~ ~ 'O ~ bD O O ~,~'' >~ a~.~ ~
cn N '~ .~ >yn tr4 ~n ~ p, U .C O 'O cn 'C
N U pp ..--v c~ '~ CCf ~ U bA v~ 'd U ~.' N
O
b°0 ~3 '~ ~ ~ p 4-i cct U ~ ~ ..~ ~ ~ .~', cUn "yn O ctY ~ .-, O
'~? o ~ ~ ~ ~~ ~ o o ~P; ;~ h y v;Y.~ 'c~ ~'''' ~,n o ~ ~. a~.~
N Ix cyC ~ N ~ ''-' N ~ ~ U ~ N U
v '~ '~ ~ ~ °' :~ ~ ~ a~ ~ y ~ o ~ .~ o ° .b o A ~ ~ b ~ ~ w o ~ ~ ~ ~ ~ ~ b ~u ~ b ~ ~ ."°.~.~
d U o >~
U
'd >=L ~ U
4a N c~
P~-i P.i p., U ~ H E-a <110> INCYTE GENOMICS, INC.
YUE, Henry TANG, Y. Tom PATTERSON, Chandra GANDHI, Ameena R.
TRIBOULEY, Catherine M.
LEE, Ernestine A.
YAO, Monique G.
BANDMAN, Olga LU, Dyung Aina M.
<120> AMINOACYL tRNA SYNTHETASES
<130> PI-0109 PCT
<140> To Be Assigned <141> Herewith <150> 60/207,248; 60/208,791; 60/210,585 <151> 2000-05-25; 2000-06-01; 2000-06-08 <160> 8 <170> PERL Program <210> 1 <211> 564 <212> PRT
<213> Homo Sapiens <220>
<221> misc_feature <223> Incyte ID No: 4574912CD1 <400> 1 Met Leu Arg Thr Thr Arg G1y Pro Gly Leu Gly Pro Pro Leu Leu Gln Ala Ala Leu Gly Leu Gly Arg Ala Gly Trp His Trp Pro Ala G1y Arg Ala Ala Ser Gly G1y Arg Gly Arg Ala Trp Leu Gln Pro Thr Gly Arg Glu Thr Gly Val Gln Val Tyr Asn Ser Leu Thr Gly Arg Lys Glu Pro Leu Ile Val Ala His Ala Glu Ala Ala Ser Trp Tyr Ser Cys Gly Pro Thr Val Tyr Asp His Ala His Leu Gly His Ala Cys Ser Tyr Val Arg Phe Asp Ile Tle Arg Arg Ile Leu Thr Lys Val Phe Gly Cys Ser Ile Va1 Met Val Met Gly Ile Thr Asp Val Asp Asp Lys Ile Ile Lys Arg Ala Asn G1u Met Asn Ile Ser Pro Ala Ser Leu Ala Ser Leu Tyr Glu Glu Asp Phe Lys Gln Asp Met Ala Ala Leu Lys Val Leu Pro Pro Thr Val Tyr Leu Arg Val Thr Glu Asn Ile Pro Gln Ile Ile Ser Phe Ile Glu Gly Ile Ile Ala Arg Gly Asn A1a Tyr Ser Thr Ala Lys Gly Asn Val Tyr Phe Asp Leu Lys Ser Arg Gly Asp Lys Tyr Gly Lys Leu Val Gly Val Val Pro Gly Pro Val Gly Glu Pro Ala Asp Ser Asp Lys Arg His Ala Ser Asp Phe Ala Leu Trp Lys Ala Ala Lys Pro Gln Glu Val Phe Trp Ala Ser Pro Trp Gly Pro Gly Arg Pro Gly Trp His Ile Glu Cys Ser Ala Ile Ala Ser Met Val Phe Gly Ser Gln Leu Asp Ile His Ser Gly Gly Ile Asp Leu Ala Phe Pro His His Glu Asn Glu Ile Ala Gln Cys Glu Va1 Phe His Gln Cys Glu Gln Trp G1y Asn Tyr Phe Leu His Ser G1y His Leu His Ala Lys G1y Lys Glu Glu Lys Met Ser Lys Ser Leu Lys Asn Tyr Ile Thr Ile Lys Asp Phe Leu Lys Thr Phe Ser Pro Asp Val Phe Arg Phe Phe Cys Leu Arg Ser Ser Tyr Arg 5er Ala Ile Asp Tyr Ser Asp Ser Ala Met Leu Gln Ala Gln Gln Leu Leu Leu G1y Leu Gly Ser Phe Leu Glu Asp Ala Arg Ala Tyr Met Lys Gly Gln Leu Ala Cys Gly Ser Val Arg Glu Ala Met Leu Trp Glu Arg Leu Ser Ser Thr Lys Arg Ala Val Lys Ala Ala Leu Ala Asp Asp Phe Asp Thr Pro Arg Val Val Asp Ala Ile Leu Gly Leu Ala His His Gly Asn Gly Gln Leu Arg Ala Ser Leu Lys Glu Pro Glu Gly Pro Arg Ser Pro Ala Val Phe Gly Ala Ile Ile Ser Tyr Phe Glu Gln Phe Phe Glu Thr Val Gly Ile Ser Leu Ala Asn Gln Gln Tyr Val Ser Gly Asp Gly Ser Glu Ala Thr Leu His Gly Va1 Val Asp Glu Leu Val Arg Phe Arg G1n Lys Va1 Arg Gln Phe Ala Leu Ala Met Pro Glu Ala Thr Gly Asp Ala Arg Arg Gln Gln Leu Leu Glu Arg Gln Pro Leu Leu Glu Ala Cys Asp Thr Leu Arg Arg Gly Leu Thr Ala His Gly Ile Asn Ile Lys Asp Arg Ser Ser Thr Thr Ser Thr Trp Glu Leu Leu Asp Gln Arg Thr Lys Asp Gln Lys Ser A1a G1y <210> 2 <211> 477 <212> PRT
<213> Homo sapiens <220>
<221> misc_feature <223> Incyte ID No: 7475765CD1 <400> 2 Met Leu Gly Va1 Arg Cys Leu Leu Arg Ser Val Arg Phe Cys Ser Ser Ala Pro Phe Pro Lys His Lys Pro Ser Ala Lys Leu Ser Val Arg Asp Ala Leu Gly Ala Gln Asn Ala Ser Gly Glu Arg Ile Lys Ile Gln Gly Trp I1e Arg Ser Val Arg Ser Gln Lys Glu Val Leu Phe Leu His Val Asn Asp Gly Ser Ser Leu Glu Ser Leu Gln Val Val Ala Asp Ser Gly Leu Asp Ser Arg Glu Leu Thr Phe Gly Ser Ser Val Glu Val Gln Gly Gln Leu Ile Lys Ser Pro Ser Lys Arg Gln Asn Val Glu Leu Lys Ala Glu Lys Ile Lys Val Ile Gly Asn Cys Asp Ala Lys Asp Phe Pro Ile Lys Tyr Lys G1u Arg His Pro Leu Glu Tyr Leu Arg Gln Tyr Pro His Phe Arg Cys Arg Thr Asn Val Leu Gly Ser Ile Leu Arg Ile Arg Ser Glu Ala Thr Ala Ala Ile His Ser Phe Phe Lys Asp Ser Gly Phe Va1 His I1e His Thr Pro Ile Ile Thr Ser Asn Asp Ser Glu G1y Ala Gly Glu Leu Phe Gln Leu Glu Pro Ser Gly Lys Leu Lys Val Pro Glu Glu Asn Phe Phe Asn Val Pro Ala Phe Leu Thr Val Ser Gly Gln Leu His Leu Glu Val Met Ser Gly Ala Phe Thr Gln Val Phe Thr Phe Gly Pro Thr Phe Arg A1a Glu Asn Ser Gln Ser Arg Arg His Leu A1a Glu Phe Tyr Met Ile Glu A1a Glu Ile Ser Phe Val Asp Ser Leu Gln Asp Leu Met Gln Val Ile Glu Glu Leu Phe Lys A1a Thr Thr Met Met Val Leu Ser Lys Cys Pro Glu Asp Val Glu Leu Cys His Lys Phe Ile Ala Pro Gly G1n Lys Asp Arg Leu Glu His Met Leu Lys Asn Asn Phe Leu Ile Ile Ser Tyr Thr Glu Ala Val Glu Ile Leu Lys Gln A1a Ser Gln Asn Phe Thr Phe Thr Pro Glu Trp Gly Ala Asp Leu Arg Thr Glu His Glu Lys Tyr Leu Val Lys His Cys Gly Asn Ile Pro Val Phe Val Ile Asn Tyr Pro Leu Thr Leu Lys Pro Phe Tyr Met Arg Asp Asn Glu Asp Gly Pro Gln His Thr Val Ala A1a Val Asp Leu Leu Val Pro Gly Val Gly Glu Leu Phe Gly Gly Gly Leu Arg Glu Glu Arg Tyr His Phe Leu Glu Glu Arg Leu Ala Arg Ser Gly Leu Thr G1u Val Tyr Gln Trp Tyr Leu Asp Leu Arg Arg Phe Gly Ser Val Pro His Gly Gly Phe Gly Met Gly Phe Glu Arg Tyr Leu G1n Cys Ile Leu Gly Val Asp Asn Ile Lys Asp Val I1e Pro Phe Pro Arg Phe Pro His Ser Cys Leu Leu 3l9 <210> 3 <211> 621 <212> PRT
<213> Homo Sapiens <220>
<221> misc_feature <223> Incyte ID No: 7475776CD1 <400> 3 Met Gly Ser Cys Ala Pro Gly Arg Ser His Arg Ala Gly Arg Ala Val Asp Ser Pro Cys Leu Ser Phe Ser Arg Ser Gly Asp Val Ile Glu Tyr Leu Leu Lys Asn Gln Trp Phe Val Arg Cys Gln Glu Met Gly Ala Arg Ala Ala Lys Ala Val Glu Ser Gly A1a Leu Glu Leu Ser Pro Ser Phe His Gln Lys Asn Trp Gln His Trp Phe Ser His I1e Gly Asp Trp Cys Va1 Ser Arg Gln Leu Trp Trp G1y His Gln Ile Pro Ala Tyr Leu Val Val Glu Asp His A1a Gln Gly Glu Glu Asp Cys Trp Val Val Gly Arg Ser Glu Ala Glu Ala Arg Glu Val Ala Ala Glu Leu Thr Gly Arg Pro Gly Ala Glu Leu Thr Leu Glu Arg Asp Pro Asp Val Leu Asp Thr Trp Phe Ser Ser A1a Leu Phe Pro Phe Ser Ala Leu Gly Trp Pro Gln Glu Thr Pro Asp Leu Ala Arg Phe Tyr Pro Leu Ser Leu Leu Glu Thr Gly Ser Asp Leu Leu Leu Phe Trp Val Gly Arg Met Val Met Leu G1y Thr G1n Leu Thr Gly Gln Leu Pro Phe Ser Lys Val Leu Leu His Pro Met Val Arg Asp Arg Gln G1y Arg Lys Met Ser Lys Ser Leu Gly Asn Val Leu Asp Pro Arg Asp Ile Ile Ser Gly Val Glu Met Gln Leu Leu G1n Glu Lys Leu Arg Ser Gly Asn Leu Asp Pro Ala Glu Leu Ala Ile Val Ala Ala Ala Gln Lys Lys Asp Phe Pro His G1y Ile Pro Glu Cys Gly Thr Asp Ala Leu Arg Phe Thr Leu Cys Ser His Gly Val Gln Ala Gly Asp Leu His Leu Ser Val Ser Glu Val Gln Ser Cys Arg His Phe Cys Asn Lys Ile Trp Asn Ala Leu Arg Phe Ile Leu Asn Ala Leu Gly Glu Lys Phe Val Pro Gln Pro Ala Glu Glu Leu Ser Pro Ser Ser Pro Met Asp Ala Trp Ile Leu Ser Arg Leu Ala Leu Ala A1a G1n Glu Cys Glu Arg Gly Phe Leu Thr Arg Glu Leu Ser Leu Val Thr His Ala Leu His His Phe Trp Leu His Asn Leu Cys Asp Val Tyr Leu Glu Ala Val Lys Pro Val Leu Trp His Ser Pro Arg Pro Leu Gly Pro Pro Gln Val Leu Phe Ser Cys Ala Asp Leu Gly Leu Arg Leu Leu Ala Pro Leu Met Pro Phe Leu Ala Glu Glu Leu Trp Gln Arg Leu Pro Pro Arg Pro Gly Cys Pro Pro Ala Pro Ser Ile Ser Va1 Ala Pro Tyr Pro Ser Ala Cys Ser Leu Glu His Trp Arg Gln Pro Glu Leu Glu Arg Arg Phe Ser Arg Val Gln Glu Val Val Gln Val Leu Arg Ala Leu Gln Ala Thr Tyr Gln Leu Thr Lys Ala Arg Pro Arg Val Leu Leu Gln Ser Ser Glu Pro Gly Asp Gln Gly Leu Phe Glu Ala Phe Leu Glu Pro Leu Gly Thr Leu Gly Tyr Cys Gly Ala Val Gly Leu Leu Pro Pro Gly Thr A1a Ala Pro Ser Gly Trp Ala Gln Ala Pro Leu Ser Asp Thr Ala Gln Val Tyr Met Glu Leu Gln Gly Leu Val Asp Pro Gln Ile G1n Leu Pro Leu Leu Ala Ala Arg Arg Tyr Lys Leu Gln Lys Gln Leu Asp Ser Leu Thr Ala Arg Thr Pro Ser Glu Gly Glu Ala Gly Thr Gln Arg Gln Gln Lys Leu Ser Ser Leu Gln Leu Glu Leu Ser Lys Leu Asp Lys Ala Ala Ser His Leu Arg Gln Leu Met Asp Glu Pro Pro Ala Pro Gly Ser Pro Glu Leu <210> 4 <211> 477 <212> PRT
<213> Homo sapiens <220>
<221> misc_feature <223> Incyte ID No: 5332221CD1 <400> 4 Met Ala Ala Pro Ile Leu Arg Ser Phe Ser Trp Gly Arg Trp Ser Gly Thr Leu Asn Leu Ser Val Leu Leu Pro Leu Gly Leu Arg Lys Ala His Ser Gly Ala Gln Gly Leu Leu Ala Ala Gln Lys Ala Arg Gly Leu Phe Lys Asp Phe Phe Pro Glu Thr Gly Thr Lys Tle Glu Leu Pro Glu Leu Phe Asp Arg Gly Thr Ala Ser Phe Pro Gln Thr I2e Tyr Cys Gly Phe Asp Pro Thr Ala Asp Ser Leu His Val Gly His Leu Leu Ala Leu Leu Gly Leu Phe His Leu Gln Arg Ala Gly His Asn Val Ile Ala Leu Val Gly Gly Ala Thr Ala Arg Leu Gly Asp Pro Ser Gly Arg Thr Lys Glu Arg Glu Ala Leu Glu Thr Glu Arg Val Arg Ala Asn Ala Arg Ala Leu Arg Leu Gly Leu Glu Ala Leu Ala Ala Asn His Gln Gln Leu Phe Thr Asp Gly Arg Ser Trp Gly Ser Phe Thr Val Leu Asp Asn Ser Ala Trp Tyr Gln Lys Gln His Leu Val Asp Phe Leu Ala Ala Val Gly Gly His Phe Arg Met Gly Thr Leu Leu Ser Arg Gln Ser Val Gln Leu Arg Leu Lys Ser Pro Glu Gly Met Ser Leu Ala Glu Phe Phe Tyr Gln Val Leu Gln Ala Tyr Asp Phe Tyr Tyr Leu Phe Gln Arg Tyr Gly Cys Arg Val Gln Leu Gly Gly Ser Asp Gln Leu Gly Asn Ile Met Ser Gly Tyr Glu Phe Ile Asn Lys Leu Thr Gly Glu Asp Val Phe Gly Ile Thr Val Pro Leu Ile Thr Ser Thr Thr Gly Ala Lys Leu Gly Lys Ser Ala Gly Asn Ala Val Trp Leu Asn Arg Asp Lys Thr Ser Pro Phe Glu Leu Tyr Gln Phe Phe Val Arg Gln Pro Asp Asp Ser Val Glu Arg Tyr Leu Lys Leu Phe Thr Phe Leu Pro Leu Pro Glu Ile Asp His Ile Met Gln Leu His Val Lys Glu Pro Glu Arg Arg Gly Pro Gln Lys Arg Leu Ala Ala Glu Val Thr Lys Leu Va1 His Gly Arg Glu Gly Leu Asp Ser Ala Lys Arg Cys Thr Gln Ala Leu Tyr His Ser Ser Tle Asp A1a Leu G1u Val Met Ser Asp Gln Glu Leu Lys G1u Leu Phe Lys Glu Ala Pro Phe Ser Glu Phe Phe Leu Asp Pro Gly Thr Ser Val Leu Asp Thr Cys Arg Lys Ala Asn Ala Ile Pro Asp Gly Pro Arg Gly Tyr Arg Met Ile Thr G1u Gly G1y Val Ser Ile Asn His Gln Gln Val Thr Asn Pro Glu Ser Val Leu I1e Val Gly Gln His I1e Leu Lys Asn Gly Leu Ser Leu Leu Lys Ile Gly Lys Arg Asn Phe Tyr Ile Ile Lys Trp Leu Gln Leu <210> 5 <211> 1920 <212> DNA
<213> Homo Sapiens <220>
<221> misc_feature <223> Incyte ID No: 4574912CB1 <400> 5 tgcgggtgtc actctttatt gcggggtcca cactgtgggt gctggggccc ctcccactga 60 gggaaggctg agcctctagc cagggctgac atgttgagga ctacgcgcgg cccaggcctg 120 ggccccccgc tgctccaggc cgcgctgggc cttgggcggg ctgggtggca ctggcctgcg 280 ggccgggcgg cgagcggggg gcgcgggcgg gcctggctgc agcccacggg ccgggagacg 240 ggtgtgcagg tgtacaacag cctcaccggg aggaaggaac ccctaatcgt ggcgcacgcc 300 gaagccgcct cctggtatag ctgtggacca actgtatatg atcatgcgca ccttggccat 360 gcttgctcat atgttagatt tgatatcatt cgaaggatcc taaccaaggt ttttggatgc 420 agcatagtca tggtgatggg tattacagat gtagatgata aaatcatcaa aagagccaat 480 gagatgaata tttcccccgc ttccctcgcc agtctttatg aggaagactt caagcaggac 540 atggcagccc tgaaggttct cccacccacg gtgtacctga gggtaaccga aaatattcct 600 cagataattt ctttcattga aggaatcatt gctcgtggga acgcttattc aacggcaaaa 660 ggcaatgtct acttcgatct gaagtctaga ggagacaagt atggcaaatt ggtcggcgtg 720 gtccctggtc cagtcggaga gccagcggac tctgacaagc gtcatgccag tgacttcgcc 780 ctgtggaagg cggccaaacc ccaggaggtg ttctgggcct ctccctgggg acccgggagg 840.
ccgggctggc acatcgagtg ctctgccatc gctagtatgg tatttggaag tcaactggat 900 atccattcag gtgggataga tttagctttt ccacatcatg agaacgaaat tgcacagtgc 960 gaagtctttc atcagtgcga gcagtgggga aattattttc tgcattctgg gcatttgcac 1020 gccaaaggca aagaagaaaa aatgtccaaa tcattaaaga actacattac tattaaggac 1080 tttctgaaga ccttttcccc cgatgtcttc cggttcttct gcctgcggag cagctaccgc 1140 tcagccatcg actacagtga cagcgccatg ctccaagctc agcagctgct cctggggctg 1200 ggctctttcc tggaggacgc acgtgcctac atgaaggggc agctggcctg cggctccgtc 1260 agggaagcga tgctgtggga gaggctctcc agcaccaaga gggccgtgaa ggcggccttg 1320 gcagatgact ttgacacacc cagggtggtt gatgccatcc tgggccttgc acaccacggg 1380 aatggacagc tcagggcgtc cctgaaggaa cctgaagggc cgagaagtcc tgctgtgttt 1440 ggtgccatca tctcttactt tgaacagttt tttgaaactg ttggaatttc tctggcaaat 1500 caacagtacg tttcaggaga cggcagcgag gctaccttgc atggtgtggt ggacgagctg 1560 gtgcggttcc ggcagaaggt ccggcagttt gcgctggcca tgcccgaggc cacgggggac 1620 gcccggcggc agcagctcct agaaaggcag cccctgctgg aagcatgcga caccctgcgc 1680 cggggcctga ctgcccacgg catcaacatc aaggacagaa gcagtacaac atccacgtgg 1740 gaactgctgg atcaaaggac aaaagaccaa aaatcagcgg gctgaggatg gagcacagcc 1800 atgaacctgc tcacgacaag acgcacccat gcttctcagg gtcaaggctt tatgttaaag 1860 cttcctgtcg gggctgctag gtcagcatta aagtaaggca accaacagtg aaaaaaaaaa 1920 <210> 6 <211> 2480 <212> DNA
<213> Homo Sapiens <220>
<221> misc_feature <223> Incyte ID No: 7475765CB1 <400> 6 gggctcgagg tggttgcgcg tgttgcggcc cgggttctgt tcgtaggttt acaattgtcg 60 cgggtagtgg ggcgccccag ctgccaactg cacgtggttt ctttctggcg tttgcctagt 120 gtcctgtcca ttccgccccg ttgggaagtg ggaagccgaa aaggtcctgt ggggacagag 180 ctgggagtcc tgaaggagaa agcgcgcagc cgaggggctc tggagtgcct tagagcagag 240 ctgcggccgc ggaaggggag cagctgagaa aggagggccg ctgcaggcgg ggttcgaacc 300 gtggggtctg ggctgctccc gcggagggcc tgggcggacg cgggatgctg ggggtccgct 3&0 gcctgctgcg gtccgtgcgc ttctgttcct ccgccccctt ccccaagcac aaaccttcag 420 ccaaactgag cgtgcgggac gctctcgggg ctcagaacgc gagtggggag cgcattaaga 480 tccagggatg gattcgttct gtccgatccc agaaggaagt cttgttcctg catgtaaatg 540 atgggtcatc tttggaaagc cttcaggttg ttgcagattc aggccttgac agtagagaat 600 taacttttgg gagttctgtg gaagtacaag ggcagctgat aaaaagtcca tccaaaaggc 660 aaaatgtgga actgaaggca gaaaaaatta aagttattgg aaattgtgat gccaaggatt 720 tccccatcaa atataaagag aggcatcctc tggagtatct gcgacaatat cctcacttta 780 ggtgtaggac taacgttctg ggttctatat tgaggattcg cagtgaagcg acagctgcta 840 ttcattcttt ctttaaggac agtggctttg tacatattca tactccaata atcacatcca 900 atgactctga gggagctgga gaactttttc aacttgaacc ttcaggcaaa cttaaggtac 960 ctgaggagaa tttcttcaat gttcctgctt tcttaactgt ctcaggacaa cttcatctag 1020 aagtgatgtc aggagctttt actcaagtgt ttacctttgg tccgaccttc cgagctgaaa 1080 attctcagag ccggaggcac ctggcagagt tttatatgat agaagcagag atttcttttg 1140 ttgacagcct tcaagatctt atgcaggtta tagaggaact gttcaaggct acaacaatga 1200 tggttctctc aaaatgtcct gaagatgttg aactctgtca caaattcata gcacctggcc 1260 aaaaggacag attagaacat atgctaaaaa acaacttttt aatcatttct tatactgaag 1320 cagtggagat cttaaagcaa gcatcccaga acttcacctt taccccagag tggggtgctg 1380 acctacggac tgaacatgaa aagtacctgg tgaagcactg tggcaacata cctgtcttcg 1440 ttattaatta tccattaaca ctcaagcctt tctacatgag ggataatgaa gatggccctc 1500 7l9 agcacacggt tgctgctgtt gatcttctgg ttcctggagt tggggaactc tttggaggag 1560 gcctcagaga agaacgatac catttcttag aggagcgctt agccagatcg ggacttacag 1620 aagtctacca atggtatctg gaccttcgtc gatttggatc tgtgccacat ggaggttttg 1680 ggatgggatt tgaacgctac ctgcagtgca tcttgggtgt tgacaatatc aaagatgtta 1740 tccctttccc aaggtttcct cattcatgcc ttttatagct ggaagattgg ttaaggaaaa 1800 gcacccccca tggcagagac actgcacatg attgtgcata cagcagaatg catgtttgga 1860 ttttagaaat gcagatttca atatgtaatt gttgtgccat aagatatcat agaaaaaata 1920 taagtggttg tgattttctt agaaagttga gggtatttca cgtaaggatg agctcccgca 1980 agaagaggta cttatagcaa ggggactctc aaatccatta cctcaattaa gaaatgaaga 2040 aattgaatta gtctcaaagt ttcttttaaa ctctaaaaca gaatgagata atgtatttta 2100 cgttgtctat aatcattaaa tcactccctg tgtaatttgt gagaaccatc tagtagctcg 2160 aaataaaata atgttgcatc ttttctccct tgccatatac tttgtgataa atccttatct 2220 cattttcagt acttcattaa acattgcaga aaaaaatatt ccttaaggtc ttaattgatt 2280 taaagaagta gctattctga attgaaatct cctttcattg aactggataa aaaaatcatg 2340 tttaataact gttgcttttc aattttcaaa gctgttgaga tattacatta agtatttcaa 2400 ctctttaatc actgttgtta taatttgttt atatttgatg tttataattt gtctaactaa 2460 aatagatttt tttaatagac 2480 <210> 7 -<211> 2714 <212> DNA
<213> Homo Sapiens <220>
<221> misc_feature <223> Incyte ID No: 7475776CB1 <400> 7 cagggcggtg caggtgatga tgatacatct ggaaaagcaa aagccaaggt caggttcagt 60 actcaccatg gctgtgctcc ccaaggggca gtgaaggtga ctccagctca cagtcctgcc 120 gatgctgaga tgggggcccg acatggcttg agccccttga atgtcattgc ggaggatggg 180 accatgacct ccctctgcgg ggactggttg caggtggtac caccctatgt taccccatcc 240 tttgggggct ctctgtcccc ctaatcctcc tcctagtttc ttatttctct agaggccttc 300 agtctttact cttgccgctt tttctccagg gtcttcaccg gtttgtggcc cgggaaaaga 360 taatgtctgt gctgagtgaa cggggcctat tccggggcct ccagaaccac cccatggtac 420 tgcccatctg caggtacctc attttaactc ctttactaag ggctacccca aaagggaatg 480 tatggagctt aagggtgaca ataggatggg ctctgcaccc ctccgttaga atacgagctc 540 cgtgtcggtt ttattcgcta ttgtatcctc agtaccaagg gcctggcatg gcatggggtc 600 ttgtgcccct gggagaagtc acagggccgg aagagcagtg gactcaccct gtctctcttt 660 cagccgttct ggggatgtga tagaatacct gctgaagaac cagtggtttg tccgctgcca 720 ggaaatgggg gcccgagctg ccaaggctgt ggagtcgggg gccctggagc tcagtccctc 780 cttccaccag aagaactggc agcactggtt ttcccatatt ggggactggt gtgtctcccg 840 gcagctgtgg tggggccatc agattccagc ctacctggtt gtagaggacc atgcgcaggg 900 agaagaggac tgttgggtgg ttgggcggtc agaggctgag gccagagagg tagcagcgga 960 actgacaggg aggccagggg cagagctgac cctggagagg gatcctgatg tcctagacac 1020 atggttttct tctgccctgt tccccttttc tgccctgggc tggccccaag agaccccaga 1080 ccttgctcgt ttctaccccc tgtcactttt ggaaacgggc agcgaccttc tgctgttctg 1140 ggtgggccgc atggtcatgt tggggaccca gctcacaggg cagctgccct tcagcaaggt 1200 gcttcttcat cccatggttc gggacaggca gggccggaag~atgagcaagt ccctggggaa 1260 tgtgctggac ccaagagaca tcatcagtgg ggtggagatg cagttgctgc aggaaaagct 1320 gagaagcgga aatttggacc ctgcagagct ggccattgtg gctgcagcac agaaaaagga 1380 ctttcctcac gggatccctg agtgtgggac agatgccctg agattcacac tctgctccca 1440 tggagttcag gcgggcgact tgcacctgtc agtctctgag gtccagagct gccgacattt 1500 ctgcaacaag atctggaatg ctcttcgctt tatcctcaat gctttagggg agaaatttgt 1560 gccacagcct gctgaggagc tgtctccctc ctccccgatg gatgcctgga tcctgagccg 1620 ccttgccctg gctgcccagg agtgtgagcg gggcttcctc acccgagagc tctcgctcgt 1680 cactcatgcc ctgcaccact tctggcttca caacctctgt gacgtctacc tggaggctgt 1740 gaagcccgtg ctgtggcact cgccccgccc cctggggccc cctcaggtcc tgttctcctg 1800 cgctgacctc ggcctccgcc tcctggcccc actgatgccc ttcctggctg aagagctctg 1860 gcagaggctg ccccccaggc ctggttgccc ccctgccccc agcatctcgg ttgcccccta 1920 ccctagcgcc tgcagcttgg agcactggcg ccagccagag ctggagcggc gcttctcccg 1980 ggtccaagag gtcgtgcagg tgctaagggc tctccaagcc acgtaccagc tcaccaaagc 2040 ccggccccga gtgctgctgc agagctcaga gcctggggac cagggcctct tcgaggcctt 2100 cttggagccc ctgggcaccc tgggctactg tggggctgtg ggcctgttac ccccaggcac 2160 agcagctccc tccggctggg cccaggctcc actcagtgac acggctcaag tctacatgga 2220 gctgcagggc ctggtggacc cgcagatcca gctacctctg ttagccgccc gaaggtacaa 2280 gttgcagaag cagcttgata gcctcacagc caggacccca tcagaagggg aggcagggac 2340 tcagaggcaa caaaagcttt cttccctcca gctggaattg tcaaaactgg acaaggcagc 2400 ctctcacctc cggcagctga tggatgagcc tccagcccca gggagcccgg agctctaact 2460 catcatcccc atcagttttc ctccctctca gacctgtctt tgaggacaaa cagatttgtc 2520 agctgtcagg gtgcagtggg acgtcagaga ctatgtggtc catcgccttc attgtgtaaa 2580 tgaggacaca gactggcttg gtcgcagtga ctgtggtgtc cttgagatgc tcacattact 2640 gcccggcctg cctcccacct ggaagtctgg gaatgaggag attgagataa acttttgaaa 2700 tcccaaaaaa aaaa 2714 <210> 8 <211> 1672 <212> DNA
<213> Homo Sapiens <220>
<221> misc_feature <223> Incyte ID No: 5332221CB1 <400> 8 gtgggagcct ctgcaatccc agcattccct tcgtgccgct accaagatgg cggcgcccat 60 cttgcggtcc ttttcctggg gccggtggtc tggtacccta aatctctcag tattgttgcc 120 cttggggctg cgtaaggccc actcgggcgc tcaggggtta ctggcagcgc agaaggctcg 180 aggtctgttc aaggacttct tcccggagac ggggacgaaa atagagctcc cagagctctt 240 cgaccgtggc acggcgagtt ttccccaaac catttactgt ggcttcgacc ccacggcaga 300 ctcgcttcat gtgggtcatc tacttgcgct gctgggcctg. tttcatttgc agcgagcggg 360 ccacaacgtg atcgcgctgg tgggaggcgc cacggcgcgc ctgggagacc cgagcggccg 420 taccaaggaa cgcgaggcgc tggagacaga gcgcgtgcga gccaacgcgc gagctctgcg 480 cctagggctt gaggccctgg cggctaatca ccagcagctt ttcactgatg ggcgctcctg 540 gggcagcttc actgtgctgg acaactcggc ctggtaccag aagcagcacc tggtggactt 600.
cctggcggca gtggggggtc acttccgcat ggggacgctg ctgagccggc agagcgtgca 660 gctgcggctc aagagccccg agggcatgag cttggccgag ttcttttacc aggtgctcca 720 ggcctatgac ttctattacc tcttccagcg ttatggatgc agggtccagc tgggcggatc 780 tgatcaacta ggcaacatca tgtccggata tgagttcatc aacaagttga ctggagaaga 840 tgtatttgga atcaccgttc ctctaattac aagtacaact ggagcaaagc tgggaaagtc 900 tgctggcaac gctgtttggc taaacagaga taagacatct ccatttgaat tgtatcaatt 960 ctttgtcagg caaccggacg attcagtgga aaggtacctg aagctgttca ctttcctgcc 1020 ccttccagag attgatcata tcatgcagct gcatgtcaaa gagccagaaa ggcggggtcc 1080 tcagaaacga ctggcagcag aagtaacaaa gcttgttcat ggacgagaag gattggattc 1140 tgctaaaagg tgtacacaag ccctttatca cagtagcata gatgcactgg aggtcatgtc 1200 tgatcaggag ttaaaagagt tgtttaaaga agctccattt tctgaatttt ttctcgatcc 1260 tggaacaagt gtcctagata cttgccgcaa agcaaatgcc attccagatg gtccccgagg 1320 gtatcgaatg ataacagaag gcggagtcag cataaatcac caacaagtaa caaatcctga 1380 gagtgtttta attgttggac aacatattct caagaatgga ctttccttac ttaaaatagg 1440 aaaaagaaat ttctacatta taaaatggct tcagttgtga tgaaaagtcc ttctggttgt 1500 ccaaataaac ttacccatca ttcattctca agacctctga agggttggct ccagaactta 1560 gacctttgct tatgcaaatc agaaaaacag aatggactag gactcagtgt gagtaacttc 1620 attattttta tgggccggtt aataaatatt tgtttaataa aaaaaaaaaa as 1672

Claims (52)

What is claimed is:
1. An isolated polypeptide selected from the group consisting of:
a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1-4, b) a naturally occurring polypeptide comprising an amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:1-4, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-4, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-4.
2. An isolated polypeptide of claim 1 selected from the group consisting of SEQ ID NO:1-4.
3. An isolated polynucleotide encoding a polypeptide of claim 1.
4. An isolated polynucleotide encoding a polypeptide of claim 2.
5. An isolated polynucleotide of claim 4 selected from the group consisting of SEQ ID NO:S-8.
6. A recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide of claim 3.
7. A cell transformed with a recombinant polynucleotide of claim 6.
8. A transgenic organism comprising a recombinant polynucleotide of claim 6.
9. A method for producing a polypeptide of claim 1, the method comprising:
a) culturing a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide, and said recombinant polynucleotide comprises a promoter sequence operably linked to a polynucleotide encoding the polypeptide of claim 19 and b) recovering the polypeptide so expressed.
10. An isolated antibody which specifically binds to a polypeptide of claim 1.
11. An isolated polynucleotide selected from the group consisting of:
a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:5-8, b) a naturally occurring polynucleotide comprising a polynucleotide sequence at least 90%
identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:5-8, c) a polynucleotide complementary to a polynucleotide of a), d) a polynucleotide complementary to a polynucleotide of b), and e) an RNA equivalent of a)-d).
12. An isolated polynucleotide comprising at least 60 contiguous nucleotides of a polynucleotide of claim 11.
13. A method for detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide of claim 11, the method comprising:
a) hybridizing the sample with a probe comprising at least 20 contiguous nucleotides comprising a sequence complementary to said target polynucleotide in the sample, and which probe specifically hybridizes to said target polynucleotide, under conditions whereby a hybridization complex is formed between said probe and said target polynucleotide or fragments thereof, and b) detecting the presence or absence of said hybridization complex, and, optionally, if present, the amount thereof.
14. A method of claim 13, wherein the probe comprises at least 60 contiguous nucleotides.
15. A method for detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide of claim 11, the method comprising:
a) amplifying said target polynucleotide or fragment thereof using polymerase chain reaction amplification, and b) detecting the presence or absence of said amplified target polynucleotide or fragment thereof, and, optionally, if present, the amount thereof.
16. A composition comprising a polypeptide of claim 1 and a pharmaceutically acceptable excipient.
17. A composition of claim 16, wherein the polypeptide has an amino acid sequence selected from the group consisting of SEQ ID NO:1-4.
18. A method for treating a disease or condition associated with decreased expression of functional ATRS, comprising administering to a patient in need of such treatment the composition of claim 16.
19. A method for screening a compound for effectiveness as an agonist of a polypeptide of claim 1, the method comprising:
a) exposing a sample comprising a polypeptide of claim 1 to a compound, and b) detecting agonist activity in the sample.
20. A composition comprising an agonist compound identified by a method of claim 19 and a pharmaceutically acceptable excipient.
21. A method for treating a disease or condition associated with decreased expression of functional ATRS, comprising administering to a patient in need of such treatment a composition of claim 20.
22. A method for screening a compound for effectiveness as an antagonist of a polypeptide of claim 1, the method comprising:
a) exposing a sample comprising a polypeptide of claim 1 to a compound, and b) detecting antagonist activity in the sample.
23. A composition comprising an antagonist compound identified by a method of claim 22 and a pharmaceutically acceptable excipient.
24. A method for treating a disease or condition associated with overexpression of functional ATRS, comprising administering to a patient in need of such treatment a composition of claim 23.
25. A method of screening for a compound that specifically binds to the polypeptide of claim 1, said method comprising the steps of:
a) combining the polypeptide of claim 1 with at least one test compound under suitable conditions, and b) detecting binding of the polypeptide of claim 1 to the test compound, thereby identifying a compound that specifically binds to the polypeptide of claim 1.
26. A method of screening for a compound that modulates the activity of the polypeptide of claim 1, said method comprising:
a) combining the polypeptide of claim 1 with at least one test compound under conditions permissive for the activity of the polypeptide of claim 1, b) assessing the activity of the polypeptide of claim 1 in the presence of the test compound, and c) comparing the activity of the polypeptide of claim 1 in the presence of the test compound with the activity of the polypeptide of claim 1 in the absence of the test compound, wherein a change in the activity of the polypeptide of claim 1 in the presence of the test compound is indicative of a compound that modulates the activity of the polypeptide of claim 1.
27. A method for screening a compound for effectiveness in altering expression of a target polynucleotide, wherein said target polynucleotide comprises a sequence of claim 5, the method comprising:
a) exposing a sample comprising the target polynucleotide to a compound, under conditions suitable for the expression of the target polynucleotide, b) detecting altered expression of the target polynucleotide, and c) comparing the expression of the target polynucleotide in the presence of varying amounts of the compound and in the absence of the compound.
28. A method for assessing toxicity of a test compound, said method comprising:
a) treating a biological sample containing nucleic acids with the test compound;
b) hybridizing the nucleic acids of the treated biological sample with a probe comprising at least 20 contiguous nucleotides of a polynucleotide of claim 11 under conditions whereby a specific hybridization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide comprising a polynucleotide sequence of a polynucleotide of claim 11 or fragment thereof;

c) quantifying the amount of hybridization complex; and d) comparing the amount of hybridization complex in the treated biological sample with the amount of hybridization complex in an untreated biological sample, wherein a difference in the amount of hybridization complex in the treated biological sample is indicative of toxicity of the test compound.
29. A diagnostic test fox a condition or disease associated with the expression of ATRS in a biological sample comprising the steps of:
a) combining the biological sample with an antibody of claim 10, under conditions suitable for the antibody to bind the polypeptide and form an antibody:polypeptide complex;
and b) detecting the complex, wherein the presence of the complex correlates with the presence of the polypeptide in the biological sample.
30. The antibody of claim 10, wherein the antibody is:
a) a chimeric antibody, b) a single chain antibody, c) a Fab fragment, d) a F(ab')2 fragment, or e) a humanized antibody.
31. A composition comprising an antibody of claim 10 and an acceptable excipient.
32. A method of diagnosing a condition or disease associated with the expression of ATRS in a subject, comprising administering to said subject an effective amount of the composition of claim 31.
33. A composition of claim 31, wherein the antibody is labeled.
34. A method of diagnosing a condition or disease associated with the expression of ATRS in a subject, comprising administering to said subject an effective amount of the composition of claim 33.
35. A method of preparing a polyclonal antibody with the specificity of the antibody of claim comprising:
a) immunizing an animal with a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-4, or an immunogenic fragment thereof, under conditions to elicit an antibody response;
b) isolating antibodies from said animal; and c) screening the isolated antibodies with the polypeptide, thereby identifying a polyclonal antibody which binds specifically to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-4.
36. An antibody produced by a method of claim 35.
37. A composition comprising the antibody of claim 36 and a suitable carrier.
38. A method of making a monoclonal antibody with the specificity of the antibody of claim comprising:
a) immunizing an animal with a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-4, or an immunogenic fragment thereof, under conditions to elicit an antibody response;
b) isolating antibody producing cells from the animal;
c) fusing the antibody producing cells with immortalized cells to form monoclonal antibody-producing hybridoma cells;
d) culturing the hybridoma cells; and e) isolating from the culture monoclonal antibody which binds specifically to a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID
NO:1-4.
39. A monoclonal antibody produced by a method of claim 38.
40. A composition comprising the antibody of claim 39 and a suitable carrier.
41. The antibody of claim 10, wherein the antibody is produced by screening a Fab expression library.
42. The antibody of claim 10, wherein the antibody is produced by screening a recombinant immunoglobulin library.
43. A method for detecting a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-4 in a sample, comprising the steps of:
a) incubating the antibody of claim 10 with a sample under conditions to allow specific binding of the antibody and the polypeptide; and b) detecting specific binding, wherein specific binding indicates the presence of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID
NO:1-4 in the sample.
44. A method of purifying a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-4 from a sample, the method comprising:
a) incubating the antibody of claim 10 with a sample under conditions to allow specific binding of the antibody and the polypeptide; and b) separating the antibody from the sample and obtaining the purified polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-4.
45. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID
NO:1.
46. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID
NO:2.
47. A polypeptide of claim 1, comprising the amino acid sequence of SEQ 1D
NO:3.
48. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID
NO:4.
49. A polynucleotide of claim 11, comprising the polynucleotide sequence of SEQ ID NO:5.
50. A polynucleotide of claim 11, comprising the polynucleotide sequence of SEQ ID NO:6.
51. A polynucleotide of claim 11, comprising the polynucleotide sequence of SEQ ID NO:7.
52. A polynucleotide of claim 11, comprising the polynucleotide sequence of SEQ ID NO:8.
CA002408315A 2000-05-25 2001-05-22 Aminoacyl trna synthetases Abandoned CA2408315A1 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US20724800P 2000-05-25 2000-05-25
US60/207,248 2000-05-25
US20879100P 2000-06-01 2000-06-01
US60/208,791 2000-06-01
US21058500P 2000-06-08 2000-06-08
US60/210,585 2000-06-08
PCT/US2001/016808 WO2001090330A2 (en) 2000-05-25 2001-05-22 AMINOACYL tRNA SYNTHETASES

Publications (1)

Publication Number Publication Date
CA2408315A1 true CA2408315A1 (en) 2001-11-29

Family

ID=27395048

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002408315A Abandoned CA2408315A1 (en) 2000-05-25 2001-05-22 Aminoacyl trna synthetases

Country Status (5)

Country Link
EP (1) EP1290148A2 (en)
JP (1) JP2004510407A (en)
AU (1) AU2001263404A1 (en)
CA (1) CA2408315A1 (en)
WO (1) WO2001090330A2 (en)

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1349939A2 (en) * 2000-12-15 2003-10-08 Incyte Genomics, Inc. Aminoacyl trna synthetases
EP2307046B1 (en) 2008-06-11 2017-01-25 aTyr Pharma, Inc. Thrombopoietic activity of tyrosyl-trna synthetase polypeptides
CN106434576A (en) 2008-06-26 2017-02-22 Atyr 医药公司 Compositions and methods comprising glycyl-trna synthetases having non-canonical biological activities
US20120004185A1 (en) 2009-02-27 2012-01-05 Atyr Pharma, Inc. Polypeptide structural motifs associated with cell signaling activity
CN102428175A (en) 2009-03-16 2012-04-25 盘古生物制药有限公司 Compositions and methods comprising histidyl-trna synthetase splice variants having non-canonical biological activities
CA2757289A1 (en) 2009-03-31 2010-10-21 Atyr Pharma, Inc. Compositions and methods comprising aspartyl-trna synthetases having non-canonical biological activities
US8828395B2 (en) 2009-12-11 2014-09-09 Atyr Pharma, Inc. Antibodies that bind tyrosyl-tRNA synthetases
US20110150885A1 (en) * 2009-12-11 2011-06-23 Atyr Pharma, Inc. Aminoacyl trna synthetases for modulating hematopoiesis
EP2509625B1 (en) 2009-12-11 2015-01-28 Atyr Pharma, Inc. Histidyl trna synthetases for reducing inflammation
AU2011248625B2 (en) 2010-04-26 2017-01-05 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-tRNA synthetase
EP2563381B1 (en) 2010-04-27 2017-08-09 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl trna synthetases
EP2563382B1 (en) 2010-04-27 2017-06-07 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of threonyl trna synthetases
CN103097524B (en) 2010-04-28 2016-08-03 Atyr医药公司 The innovation for the treatment of, diagnosis and the antibody compositions relevant to the protein fragments of Alanyl-tRNA synthetase finds
CN103118693B (en) 2010-04-29 2017-05-03 Atyr 医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of valyl tRNA synthetases
AU2011248490B2 (en) 2010-04-29 2016-11-10 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Asparaginyl tRNA synthetases
CN103096925A (en) 2010-05-03 2013-05-08 Atyr医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of arginyl-tRNA synthetases
AU2011248357B2 (en) 2010-05-03 2017-02-23 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of seryl-tRNA synthetases
ES2668207T3 (en) 2010-05-03 2018-05-17 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic and antibody compositions related to fragments of methionyl-tRNA synthetases proteins
WO2011140132A2 (en) 2010-05-03 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-alpha-trna synthetases
CN103096909A (en) 2010-05-04 2013-05-08 Atyr医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutamyl-prolyl-trna synthetases
US9062302B2 (en) 2010-05-04 2015-06-23 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-tRNA synthetase complex
CN103200953B (en) 2010-05-14 2017-02-15 Atyr 医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-trna synthetases
CN103096914B (en) 2010-05-17 2015-08-12 Atyr医药公司 The innovation of the treatment relevant to the protein fragments of Leucyl-tRNA synthetase, diagnosis and antibody compositions finds
AU2011258106B2 (en) 2010-05-27 2017-02-23 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutaminyl-tRNA synthetases
EP2575857B1 (en) 2010-06-01 2018-01-24 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of lysyl-trna synthetases
ES2653718T3 (en) 2010-07-12 2018-02-08 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic and antibody compositions related to histidyl-tRNA synthetase protein fragments
WO2012021247A2 (en) 2010-07-12 2012-02-16 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-trna synthetases
US8999321B2 (en) 2010-07-12 2015-04-07 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-tRNA synthetases
KR102099730B1 (en) 2010-07-12 2020-04-16 에이티와이알 파마, 인코포레이티드 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of aspartyl-trna synthetases
WO2012027611A2 (en) 2010-08-25 2012-03-01 Atyr Pharma, Inc. INNOVATIVE DISCOVERY OF THERAPEUTIC, DIAGNOSTIC, AND ANTIBODY COMPOSITIONS RELATED TO PROTEIN FRAGMENTS OF TYROSYL-tRNA SYNTHETASES
EP2624857B1 (en) 2010-10-06 2017-08-02 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of tryptophanyl trna synthetases
US9714419B2 (en) 2011-08-09 2017-07-25 Atyr Pharma, Inc. PEGylated tyrosyl-tRNA synthetase polypeptides
WO2013086228A1 (en) 2011-12-06 2013-06-13 Atyr Pharma, Inc. Pegylated aspartyl-trna synthetase polypeptides
US9816084B2 (en) 2011-12-06 2017-11-14 Atyr Pharma, Inc. Aspartyl-tRNA synthetases
CA2858613A1 (en) 2011-12-29 2013-08-08 Atyr Pharma, Inc. Aspartyl-trna synthetase-fc conjugates
NZ628126A (en) 2012-02-16 2016-10-28 Atyr Pharma Inc Histidyl-trna synthetases for treating autoimmune and inflammatory diseases
EP3460054B1 (en) 2013-03-15 2020-10-21 aTyr Pharma, Inc. Histidyl-trna synthetase-fc conjugates
US11767520B2 (en) 2017-04-20 2023-09-26 Atyr Pharma, Inc. Compositions and methods for treating lung inflammation
CN109266734A (en) * 2018-09-25 2019-01-25 深圳市人民医院 Autoimmune disease diagnostic kit and application

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5759833A (en) * 1994-05-27 1998-06-02 Cubist Pharmaceuticals, Inc. Human isoleucyl-tRNA synthetase proteins, nucleic acids and tester strains comprising same

Also Published As

Publication number Publication date
JP2004510407A (en) 2004-04-08
WO2001090330A2 (en) 2001-11-29
WO2001090330A3 (en) 2002-08-08
EP1290148A2 (en) 2003-03-12
AU2001263404A1 (en) 2001-12-03

Similar Documents

Publication Publication Date Title
CA2408315A1 (en) Aminoacyl trna synthetases
US20040018505A1 (en) Aminoacyl trna synthetases
US20030215827A1 (en) Aminoacyl trna synthetases
US20040048290A1 (en) Aminoacyl trna synthetases
CA2431493A1 (en) Aminoacyl trna synthetases
CA2380317A1 (en) Human synthetases
EP1203016A2 (en) Proteases and protease inhibitors
WO2001020004A2 (en) Protein phosphatase and kinase proteins
EP1498480A2 (en) Human hydrolytic enzymes
WO2001032888A2 (en) Human transferase molecules
CA2409315A1 (en) Protein phosphatases
CA2410953A1 (en) Adenylyl and guanylyl cyclases
US20020160950A1 (en) Cytochrome P450 variant
CA2398003A1 (en) Phosphodiesterases
CA2413810A1 (en) Aminoacyl trna synthetases
EP1206553A2 (en) Isomerase proteins
EP1183370A2 (en) Human oxidoreductase proteins
EP1124951A2 (en) Human sorting nexins
CA2381338A1 (en) Human lyases
CA2425767A1 (en) Lipocalins
EP1242590A2 (en) Human lyases and associated proteins
CA2406264A1 (en) Protein phosphatases
EP1192250A2 (en) Human rna metabolism proteins (rmep)
EP1477564A2 (en) Human synthetases
US20030121061A1 (en) Human lyases

Legal Events

Date Code Title Description
FZDE Dead