WO2001007628A2 - Human synthetases - Google Patents

Human synthetases Download PDF

Info

Publication number
WO2001007628A2
WO2001007628A2 PCT/US2000/019980 US0019980W WO0107628A2 WO 2001007628 A2 WO2001007628 A2 WO 2001007628A2 US 0019980 W US0019980 W US 0019980W WO 0107628 A2 WO0107628 A2 WO 0107628A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
synt
polynucleotide
polypeptide
sequence
Prior art date
Application number
PCT/US2000/019980
Other languages
French (fr)
Other versions
WO2001007628A3 (en
Inventor
Y. Tom Tang
Jennifer L. Hillman
Olga Bandman
Henry Yue
Mariah R. Baughn
Preeti Lal
Dyung Aina M. Lu
Purvi Shah
Yalda Azimzai
Original Assignee
Incyte Genomics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Incyte Genomics, Inc. filed Critical Incyte Genomics, Inc.
Priority to AU63647/00A priority Critical patent/AU6364700A/en
Priority to CA002380317A priority patent/CA2380317A1/en
Priority to JP2001512894A priority patent/JP2003533967A/en
Priority to EP00950556A priority patent/EP1200596A2/en
Publication of WO2001007628A2 publication Critical patent/WO2001007628A2/en
Publication of WO2001007628A3 publication Critical patent/WO2001007628A3/en

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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/16Central respiratory analeptics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/14Drugs for genital or sexual disorders; Contraceptives for lactation disorders, e.g. galactorrhoea
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/08Plasma substitutes; Perfusion solutions; Dialytics or haemodialytics; Drugs for electrolytic or acid-base disorders, e.g. hypovolemic shock
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • This invention relates to nucleic acid and ammo acid sequences of synthetases and to the use of these sequences in the diagnosis, treatment, and prevention of immune, neuronal, and reproductive disorders, and cell pro ferative disorders including cancer
  • a large number of cellular biosynthetic intermediary metabolism processes involve lntermolecular transfer of carbon atom-containing substrates (carbon substrates) Examples of such reactions include the tncarboxyhc acid cycle, synthesis of fatty acids and long-chain phospholipids, synthesis of alcohols and aldehydes, synthesis of intermediary metabolites, and reactions involved in the amino acid degradation pathways Many of these reactions are catalyzed by synthetases (also called hgases), which catalyze the formation of a bond between two substrate molecules Some of these reactions require input of energy, usually in the form of conversion of ATP to either ADP or AMP and pyrophosphate Synthetases are named for the products of the reaction they catalyze and are involved in such processes as metabolism and the synthesis of macromolecules Ligases forming carbon-oxygen bonds
  • Proteins make up more than half of the total dry mass of a cell
  • the synthesis of proteins is central to cell maintenance, growth, and development Synthesis occurs on ⁇ bosomes and depends on the cooperative interaction of several classes of RNA molecules
  • the process begins with transcription of the genetic code contained within the DNA to form messenger RNA (mRNA)
  • mRNA messenger RNA
  • the mRNA moves in steps through a ⁇ bosome and the nucleotide sequence of the mRNA is translated into a corresponding sequence of amino acids to construct a distinct protein chain
  • the am oacyl-transfer RNA (tRNA) synthetases are important RNA-associated enzymes with roles in translation Protein biosynthesis depends on each amino acid forming a linkage with the appropnate tRNA
  • the aminoacyl-tRNA synthetases are responsible for the activation and correct attachment of an amino acid with its cognate tRNA
  • the 20 aminoacyl-tRNA synthetase enzymes can be divided into two structural classes, and each class is charactenzed by a distinctive topology of the catalytic domain Class I enzymes contain a catalytic domain based on the nucleotide-binding Rossman 'fold' Class II enzymes contain a central catalytic domain, which consists of a seven-stranded antiparallel ⁇ -sheet motif, as well as N- and C- terminal regulatory domains Class II enzymes are separated into two groups based on the heterodimenc or homodimenc structure of the enzyme, the latter group is further subdivided by the structure
  • a carbon substrate is denved from a small molecule containing at least two carbon atoms
  • the carbon substrate is often covalently bound to a larger molecule which acts as a carbon substrate earner molecule within the cell
  • the earner molecule is coenzyme A
  • Coenzyme A (CoA) is structurally related to denvatives of the nucleotide ADP and consists of 4'- ⁇ hosphopantethe ⁇ ne linked via a phosphodiester bond to the alpha phosphate group of adenosine 3',5'-b ⁇ sphosphate
  • the terminal thiol group of 4'-phosphopantetheme acts as the site for carbon substrate bond formation
  • the predominant carbon substrates which utilize CoA as a earner molecule dunng biosynthesis and intermediary metabolism in the cell are acetyl, succinyl, and propionyl moieties, collectively referred to as acyl groups
  • Other carbon substrates include enoyl lipid
  • acyl-CoA synthetase activity 0 acetyl-CoA synthetase, which activates acetate and several other low molecular weight carboxylic acids and is found in muscle mitochondna and the cytosol of other tissues, n) medium-chain acyl-CoA synthetase, which activates fatty acids containing between four and eleven carbon atoms (predominantly from dietary sources), and is present only in liver mitochondna, and in) acyl CoA synthetase, which is specific for long chain fatty acids with between six and twenty carbon atoms, and is found m microsomes and the mitochondna Proteins associated with acyl-CoA synthetase activity have been identified from many sources including bactena, yeast, plants, mouse, and man The activity of acyl-CoA synthetase may be modulated by phosphorylation of
  • a key representative of the amide synthases is the enzyme glutamine synthetase (glutamate- ammonia hgase) that catalyzes the animation of glutamic acid to glutamme by ammonia using the energy of ATP hydrolysis
  • glutamine is the primary source for the amino group in va ⁇ ous amide transfer reactions involved in de novo pyrimidine nucleotide synthesis and in pu ⁇ ne and py ⁇ midine ⁇ bonucleotide interconversions, as well as the conversion of aspartate to asparagine
  • Overexpression of glutamine synthetase has been observed in p ⁇ mary liver cancer (Chnsta, L et al ( 1994) Gastroent 106 1312- 1320) Cyclo-hgases and other carbon-nitrogen ligases compnse va ⁇ ous enzymes and enzyme complexes that participate in the de novo pathways to pu ⁇ ne and py ⁇ midine biosynthesis Because these pathways are c ⁇ tical to the synthesis of nu
  • de novo synthesis of the py ⁇ midine nucleotides u ⁇ dylate and cytidylate also a ⁇ ses from a common precursor, in this instance the nucleotide orotidylate de ⁇ ved from orotate and phospho ⁇ bosyl pyrophosphate (PPRP)
  • PPRP phospho ⁇ bosyl pyrophosphate
  • ATCase aspartate transcarbamylase
  • carbamyl phosphate synthetase II carbamyl phosphate synthetase II
  • DHOase dihydroorotase
  • Ligases in this group are represented by the carboxylases acetyl-CoA carboxylase and pyruvate carboxylase
  • Acetyl-CoA carboxylase is a complex which includes a biotin carboxyl earner protein, biotm carboxylase, and a carboxyl transferase made up of two alpha and two beta subunits. This complex catalyzes the carboxylation of Acetyl-CoA from CO, and t O using the energy of ATP hydrolysis (PRINTS document PRO 1069).
  • Acetyl-CoA carboxylase is the rate-limiting step in the biogenesis of long-chain fatty acids
  • Two isoforms of Acetyl-CoA carboxylase, types I and types II, are expressed in humans in a tissue-specific manner (Ha, J. et al. (1994) Eur. J. Biochem. 219:297- 306).
  • Pyruvate carboxylase is a nuclear-encoded mitochond ⁇ al enzyme that catalyzes the conversion of pyruvate to oxaloacetate, a key intermediate in the cit ⁇ c acid cycle. Ligases forming phospho ⁇ c ester bonds
  • Ligases in this group are represented by the DNA ligases involved in both DNA replication and repair
  • DNA ligases seal phosphodiester bonds between two adjacent nucleotides in a DNA chain using the energy from ATP hydrolysis to first activate the free 5'-phosphate of one nucleotide and then react it with the 3'-OH group of the adjacent nucleotide.
  • This reseahng reaction is used in both DNA replication to join small DNA fragments called "Okazaki" fragments that are transiently formed in the process of replicating new DNA, and in DNA repair.
  • DNA repair is the process by which accidental base changes, such as those produced by oxidative damage, hydrolytic attack, or uncontrolled methylation of DNA, are corrected before replication or transc ⁇ ption of the DNA can occur.
  • Bloom's syndrome is an inhe ⁇ ted human disease in which individuals are partially deficient in DNA hgation and consequently have an increased incidence of cancer (Alberts, B et al. ( 1994) The Molecular Biology of the Cell, Garland Publishing Inc., New York, NY, p. 247)
  • the discovery of new synthetases and the polynucleotides encoding them satisfies a need in the art by providing new compositions which are useful m the diagnosis, prevention, and treatment of immune, neuronal, and reproductive disorders, and cell prohferative disorders including cancer.
  • the invention features punfied polypeptides, human synthetases, referred to collectively as
  • the invention provides an isolated polypepttde compnsing an amino acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, b) a naturally occumng amino acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO.1-15, and d) an immunogenic fragment of an ammo acid sequence selected from the group consisting of SEQ ID NO: 1-15.
  • the invention provides an isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 1-15.
  • the invention further provides an isolated polynucleotide encoding a polypeptide compnsing an amino acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, b) a naturally occumng amino acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, and d) an immunogenic fragment of an am o acid sequence selected from the group consisting of SEQ ID NO: 1-15.
  • the polynucleotide encodes a polypeptide selected from the group consisting of SEQ ID NO- 1-15
  • the polynucleotide is selected from the group consisting of SEQ ID NOT 6-30.
  • the invention provides a recombinant polynucleotide compnsing a promoter sequence operably linked to a polynucleotide encoding a polypeptide compnsing an amino acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO: l-15, b) a naturally occumng amino acid sequence having at least 90% sequence identity to an ammo acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO: 1- 15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15.
  • the invention provides a cell transformed with the recombinant polynucleotide.
  • the invention provides a transgenic organism compnsing the recombinant polynucleotide
  • the invention also provides a method for producing a polypeptide compnsing an am o acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, b) a naturally occurring amino acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOT-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO- 1-15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15.
  • the method compnses a) cultu ⁇ ng a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide compnsing a promoter sequence operably linked to a polynucleotide encoding the polypeptide, and b) recove ⁇ ng the polypeptide so expressed.
  • the invention provides an isolated antibody which specifically binds to a polypeptide compnsing an amino acid sequence selected from the group consisting of a) an ammo acid sequence selected from the group consisting of SEQ ID NO: 1-15, b) a naturally occumng ammo acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, and d) an immunogenic fragment of an ammo acid sequence selected from the group consisting of SEQ ID NO 1-15
  • the invention further provides an isolated polynucleotide compnsing a polynucleotide sequence selected from the group consisting of a) a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, b) a naturally occu ng polynucleotide sequence having at least 90% sequence identity to a polynucleotide sequence selected from the
  • the invention provides a method for detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide comprising a polynucleotide sequence selected from the group consisting of a) a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, b) a naturally occumng polynucleotide sequence having at least 90% sequence identity to a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, c) a polynucleotide sequence complementary to a), d) a polynucleotide sequence complementary to b), and e) an RNA equivalent of a)-d)
  • the method comprises a) hybndizing the sample with a probe compnsing at least 20 contiguous nucleotides compnsing a sequence complementary to said target polynucleotide in the sample, and which probe specifically
  • the invention also provides a method for screening a compound for effectiveness as an agonist of a polypeptide compnsing an amino acid sequence selected from the group consisting of a) an ammo acid sequence selected from the group consisting of SEQ ID NO 1-15, b) a naturally occurring amino acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15
  • the method compnses a) exposing a sample compnsing the polypeptide to a compound, and b) detecting agonist activity in the sample
  • the invention provides a pharmaceutical composition comprising an agonist compound identified by the method and a pharmaceutically acceptable excipient
  • the invention provides a method of treating a disease or condition associated with decreased expression of functional SYNT, compnsing admimstenng
  • the invention provides a method for screening a compound for effectiveness as an antagonist of a polypeptide compnsing an ammo acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, b) a naturally occumng ammo acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15
  • the method compnses a) exposing a sample compnsing the polypeptide to a compound, and b) detecting antagonist activity in the sample
  • the invention provides a pharmaceutical composition compnsing an antagonist compound identified by the method and a pharmaceutically acceptable excipient
  • the invention provides a method of treating a disease or condition associated with overexpression of functional SYNT, compnsing
  • the invention further provides a method of screening for a compound that modulates the activity of a polypeptide comprising an ammo acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO 1- 15, b) a naturally occumng amino acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an ammo acid sequence selected from the group consisting of SEQ ID NO 1- 15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1- 15
  • the method compnses 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) companng 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 invention further provides a method for screening a compound for effectiveness in alternativeng expression of a target polynucleotide, wherein said target polynucleotide compnses a sequence selected from the group consisting of SEQ ID NO 16-30, the method compnsing a) exposing a sample compnsing 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 compnsing a) treating a biological sample containing nucleic acids with the test compound, b) hybndizing the nucleic acids of the treated biological sample with a probe compnsing at least 20 contiguous nucleotides of a polynucleotide compnsing a polynucleotide sequence selected from the group consisting of I) a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, n) a naturally occumng polynucleotide sequence having at least 90% sequence identity to a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, in) a polynucleotide sequence complementary to I), iv) a polynucleotide sequence complementary to n), and v) an RNA equivalent of ⁇ )- ⁇ v) Hybndization occurs under conditions whereby a specific hyb
  • Table 1 shows polypeptide and nucleotide sequence identification numbers (SEQ ID NOs), clone identification numbers (clone IDs), cDNA hbranes, and cDNA fragments used to assemble full- length sequences encoding SYNT
  • Table 2 shows features of each polypeptide sequence, including potential motifs, homologous sequences, and methods, algorithms, and searchable databases used for analysis of SYNT
  • Table 3 shows selected fragments of each nucleic acid sequence, the tissue-specific expression patterns of each nucleic acid sequence as determined by northern analysis, diseases, disorders, or conditions associated with these tissues, and the vector into which each cDNA was cloned
  • Table 4 describes the tissues used to construct the cDNA hbranes from which cDNA clones encoding SYNT were isolated
  • Table 5 shows the tools, programs, and algonthms used to analyze the polynucleotides and polypeptides of the invention, along with applicable descriptions, references, and threshold parameters
  • SYNT refers to the ammo acid sequences of substantially pu ⁇ fied SYNT obtained from any species, particularly a mammalian species, including bovine, ovine, porcine, munne, equine, and human, and from any source, whether natural, synthetic, semi-synthetic, or recombinant
  • agonist refers to a molecule which intensifies or mimics the biological activity of SYNT Agonists may include proteins, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of SYNT either by directly interacting with SYNT or by acting on components of the biological pathway in which SYNT participates
  • Allehc va ⁇ ant is an alternative form of the gene encoding SYNT Allehc va ⁇ ants 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 allehc vanants of its naturally occumng form
  • Common mutational changes which give nse to allehc va ⁇ ants 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 SYNT include those sequences with deletions, insertions, or substitutions of different nucleotides, resulting in a polypeptide the same as SYNT or a polypeptide with at least one functional characte ⁇ stic of SYNT Included within this definition are polymorphisms which may or may not be readily detectable using a particular oligonucleotide probe of the polynucleotide encoding SYNT, and improper or unexpected hybndization to allehc vanants, with a locus other than the normal chromosomal locus for the polynucleotide sequence encoding SYNT
  • 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 SYNT Deliberate amino acid substitutions may be made on the basis of simila ⁇ ty in polarity, charge, solubility, hydrophobicity,
  • Amino acids with uncharged polar side chains having similar hydrophilicity values may include: asparagme and glutamine; and senne 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.
  • amino acid sequence refers to an ohgopeptide, peptide, polypeptide, or protein sequence, or a fragment of any of these, and to naturally occumng or synthetic molecules. Where "amino acid sequence” is recited to refer to a sequence of a naturally occumng protein molecule, “ammo 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 earned out using polymerase chain reaction (PCR) technologies well known in the art.
  • PCR polymerase chain reaction
  • Antagonist refers to a molecule which inhibits or attenuates the biological activity of SYNT Antagonists may include proteins such as antibodies, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of SYNT either by directly interacting with SYNT or by acting on components of the biological pathway in which SYNT participates
  • 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 SYNT polypeptides can be prepared using intact polypeptides or using fragments containing small peptides of interest as the immunizing antigen.
  • the polypeptide or ohgopeptide used to immunize an animal e.g., a mouse, a rat, or a rabbit
  • an animal e.g., a mouse, a rat, or a rabbit
  • an animal e.g., a mouse, a rat, or a rabbit
  • peptides Commonly used earners that are chemically coupled to peptides include bovine serum albumin, thyroglobu n, and keyhole limpet hemocyanin (KLH).
  • KLH keyhole limpet hemocyanin
  • the coupled peptide is then used to immunize the animal.
  • antigenic determinant refers to that region of a molecule (i.e., an epitope) that makes contact with a particular antibody.
  • an antigenic determinant may compete with the intact antigen (i.e., the lmmunogen used to elicit the immune response) for binding to an antibody
  • antisense refers to any composition capable of base-pai ⁇ ng with the "sense" (coding) strand of a specific nucleic acid sequence.
  • Antisense compositions may include DNA, RNA; peptide nucleic acid (PNA); ohgonucleotides having modified backbone linkages such as phosphorothioates, methylphosphonates, or benzylphosphonates; o gonucleotides having modified sugar groups such as 2 -methoxyethyl sugars or 2'-methoxyethoxy sugars; or ohgonucleotides 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 occumng nucleic acid sequence produced by the cell to form duplexes which block either transc ⁇ ption 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.
  • biologically active refers to a protein having structural, regulatory, or biochemical functions of a naturally occumng molecule.
  • immunologically active or “immunogenic” refers to the capability of the natural, recombinant, or synthetic SYNT, or of any ohgopeptide thereof, to induce a specific immune response in appropnate animals or cells and to bind with specific antibodies.
  • “Complementary” descnbes 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'
  • 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 comp ⁇ se a dry formulation or an aqueous solution.
  • Compositions compnsing polynucleotide sequences encoding SYNT or fragments of SYNT may be employed as hybndization probes.
  • the probes may be stored in freeze-dned form and may be associated with a stabilizing agent such as a carbohydrate
  • a stabilizing agent such as a carbohydrate
  • 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 XL-PCR kit (PE 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 WI) or Phrap (University of Washington, Seattle WA) Some sequences have been both extended and assembled to produce the consensus sequence.
  • GELVIEW fragment assembly system GELVIEW fragment assembly system
  • Phrap Universality of Washington, Seattle WA
  • Constant amino acid substitutions are those substitutions that are predicted to least interfere with the properties of the onginal 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 onginal amino acid in a protein and which are regarded as conservative amino acid substitutions Ongmal Residue Conservative Substitution
  • Conservative ammo 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 ammo acid residues or nucleotides
  • denvative refers to a chemically modified polynucleotide or polypeptide
  • a denvative polynucleotide encodes a polypeptide which retains at least one biological or immunological function of the natural molecule
  • a denvative 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 de ⁇ ved
  • 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
  • a “fragment” is a unique portion of SYNT or the polynucleotide encoding SYNT which is identical in sequence to but shorter in length than the parent sequence
  • a fragment may comp ⁇ se up to the entire length of the defined sequence, minus one nucleotide/amino acid residue
  • a fragment may comp ⁇ se from 5 to 1000 contiguous nucleotides or amino acid residues
  • a fragment used as a probe, p ⁇ mer, antigen, therapeutic molecule, or for other purposes, may be at least 5, 10, 15, 16, 20, 25, 30, 40, 50, 60.
  • Fragments may be preferentially selected from certain regions of a molecule
  • a polypeptide fragment may comp ⁇ se 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
  • 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 NO 16-30 compnses a region of unique polynucleotide sequence that specifically identifies SEQ ID NO 16-30, for example, as distinct from any other sequence in the genome from which the fragment was obtained
  • a fragment of SEQ ID NO 16-30 is useful, for example, in hybndization and amplification technologies and in analogous methods that distinguish SEQ ID NO 16-30 from related polynucleotide sequences
  • the precise length of a fragment of SEQ ID NO 16-30 and the region of SEQ ID NO 16-30 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 1- 15 is encoded by a fragment of SEQ ID NO 16-30
  • a fragment of SEQ ID NO 1-15 compnses a region of unique amino acid sequence that specifically identifies SEQ ID NO 1-15
  • a fragment of SEQ ID NO 1- 15 is useful as an immunogenic peptide for the development of antibodies that specifically recognize SEQ ID NO
  • 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 sinulanty or, interchangeably, sequence identity, between two or more polynucleotide sequences or two or more polypeptide sequences
  • 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 algonthm Such an algo ⁇ thm 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 compa ⁇ son of the two sequences
  • Percent identity between polynucleotide sequences may be determined using the default parameters of the CLUSTAL V algonthm 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 descnbed 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.
  • Biol 215:403-410 which is available from several sources, including the NCBI, Bethesda, MD, and on the Internet at http://www.ncbi nlm.mh gov/BLAST/ The BLAST software suite includes vanous sequence analysis programs including "blastn,” that is used to align a known polynucleotide sequence with other polynucleotide sequences from a va ⁇ ety of databases.
  • BLAST 2 Sequences 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 2.0.12 (Ap ⁇ l-21-2000) set at default parameters. Such default parameters may be, for example-
  • Percent identity may be measured over the length of an entire defined 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 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 algonthm. 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.
  • NCBI BLAST software suite may be used.
  • BLAST 2 Sequences Version 2.0.12 (Apr-21-2000) with blastp set at default parameters.
  • Such default parameters may be, for example.
  • Gap x drop-off 50
  • 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 desc ⁇ be a length over which percentage identity may be measured.
  • HACs 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
  • 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 onginal binding ability
  • “Hybndization” refers to the process by which a polynucleotide strand anneals with a complementary strand through base painng under defined hybndization conditions
  • Specific hybndization is an indication that two nucleic acid sequences share a high degree of complementarity Specific hybndization complexes form under permissive annealing conditions and remain hybridized after the "washing" step(s)
  • the washing step(s) is particularly important in determining the stnngency of the hybndization process with more stnngent conditions allowing less non-specific binding, 1 e , binding between pairs of nucleic acid strands that are not perfectly matched Permissive conditions for annealing of nucleic acid sequences are routinely determinable
  • stnngency of hybndization is expressed, in part, with reference to the temperature under which the wash step is earned out
  • wash temperatures are typically selected to be about 5°C to 20°C lower than the thermal melting point (T m ) for the specific sequence at a defined ionic strength and pH
  • T m is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe
  • High stnngency conditions for hybndization 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 %
  • blocking reagents are used to block non-specific hybndization
  • Such blocking reagents include, for instance, sheared and denatured salmon sperm DNA at about 100-200 ⁇ g/ml 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 hyb ⁇ dizations Useful vanations on these wash conditions will be readily apparent to those of ordinary skill in the art Hybndization, particularly under high stnngency conditions, may be suggestive
  • 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 , C 0 t or R ⁇ analysis) or formed between one nucleic acid sequence present m solution and another nucleic acid sequence immobilized on a solid support (e g , paper, membranes, filters, chips, pins or glass slides, or any other appropnate substrate to which cells or their nucleic acids have been fixed)
  • 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
  • Immuno 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 va ⁇ ous factors, e g., cytokines, chemokines, and other signaling molecules, which may affect cellular and systemic defense systems
  • an “immunogenic fragment” is a polypeptide or ohgopeptide fragment of SYNT which is capable of eliciting an immune response when introduced into a living organism, for example, a mammal
  • immunogenic fragment also includes any polypeptide or ohgopeptide fragment of SYNT which is useful in any of the antibody production methods disclosed herein or known in the art.
  • microarray refers to an arrangement of a plurality of polynucleotides, polypeptides, or other chemical compounds on a substrate
  • element and “array element” refer to a polynucleotide, polypeptide, or other chemical compound having a unique and defined position on a microarray
  • nucleic acid and “nucleic acid sequence” refer to a nucleotide, ohgonucleotide, polynucleotide, or any fragment thereof These phrases also refer to DNA or RNA of genomic or synthetic ongin 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-hke or RNA-hke mate ⁇ al
  • 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
  • a promoter is operably linked to a coding sequence if the promoter affects the transcnption 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
  • PNA protein nucleic acid
  • PNA refers to an antisense molecule or anti-gene agent which compnses an ohgonucleotide of at least about 5 nucleotides in length linked to a peptide backbone of ammo acid residues ending m lysine.
  • the terminal lysme confers solubility to the composition PNAs preferentially bind complementary single stranded DNA or RNA and stop transcnpt elongation, and may be pegylated to extend their hfespan in the cell
  • Post-translational modification of an SYNT may involve hpidation, 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 SYNT
  • Probe refers to nucleic acid sequences encoding SYNT, their complements, or fragments thereof, which are used to detect identical, allehc or related nucleic acid sequences
  • Probes are isolated ohgonucleotides or polynucleotides attached to a detectable label or reporter molecule Typical labels include radioactive isotopes, hgands, chemiluminescent agents, and enzymes
  • P ⁇ mers are short nucleic acids, usually DNA ohgonucleotides, which may be annealed to a target polynucleotide by complementary base-painng 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 p ⁇ mers as used in the present invention typically comp ⁇ se at least 15 contiguous nucleotides of a known sequence.
  • PCR polymerase chain reaction
  • probes and pnmers may also be employed, such as probes and p ⁇ mers that comp ⁇ se 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 p ⁇ mers 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
  • PCR p ⁇ mer pairs can be de ⁇ ved from a known sequence, for example, by using computer programs intended for that purpose such as P ⁇ mer (Version 0.5, 1991, Whitehead Institute for Biomedical Research, Cambndge MA).
  • O gonucleotides for use as p ⁇ mers are selected using software known in the art for such purpose. For example, OLIGO 4 06 software is useful for the selection of PCR pnmer pairs of up to 100 nucleotides each, and for the analysis of ohgonucleotides 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.
  • the P ⁇ mOU pnmer 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 pnmers from megabase sequences and is thus useful for designing pnmers on a genome-wide scope.
  • the P ⁇ mer3 primer selection program (available to the public from the Whitehead Institute/MIT Center for Genome Research, Cambndge MA) allows the user to input a "misp ⁇ ming library," m which sequences to avoid as p ⁇ mer binding sites are user-specified.
  • P ⁇ mer3 is useful, in particular, for the selection of o gonucleotides for microarrays.
  • the source code for the latter two p ⁇ mer selection programs may also be obtained from their respective sources and modified to meet the user's specific needs
  • the P ⁇ meGen program available to the public from the UK Human Genome Mapping Project Resource Centre, Cambndge UK) designs p ⁇ mers based on multiple sequence alignments, thereby allowing selection of p ⁇ mers that hybndize 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 ohgonucleotides and polynucleotide fragments identified by any of the above selection methods are useful in hybridization technologies, for example, as PCR or sequencing p ⁇ mers, microarray elements, or specific probes to identify fully or partially complementary polynucleotides in a sample of nucleic acids Methods of o gonucleotide selection are not limited to those described above A "recombinant nucle
  • 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 denved 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, am o acid, or antibody Reporter molecules include radionuc des, enzymes, fluorescent, chemiluminescent, or chromogemc 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 nbose instead of deoxy ⁇ bose
  • sample is used in its broadest sense
  • 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
  • substantially pu ⁇ fied 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
  • Substrate refers to any suitable ngid or semi-ngid support including membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, tubing, plates, polymers, microparticles and capillanes
  • the substrate can have a va ⁇ ety 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 desc ⁇ bes a process by which exogenous DNA is introduced into a recipient cell Transformation may occur under natural or artificial conditions according to va ⁇ ous 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, bactenophage or viral infection, electroporation, heat shock, hpofection, 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 pe ⁇ ods of time
  • a "transgenic organism,” as used herein, is any organism, including but not limited to animals and plants, m which one or more of the cells of the organism contains heterologous nucleic acid introduced by way
  • 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 2 0 9 (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 95% or at least 98% or greater sequence identity over a certain defined length
  • a vanant may be descnbed as, for example, an "allehc” (as defined above), "splice,” “species,” or “polymorphic” vanant
  • a splice vanant 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 du ⁇ ng mRNA processing The corresponding polypeptide may
  • a “vanant" 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 2 0 9 (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 95%, or at least 98% 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 synthetases (SYNT), the polynucleotides encoding SYNT, and the use of these compositions for the diagnosis, treatment, or prevention of immune, neuronal, and reproductive disorders, and cell prohferative disorders including cancer
  • Table 1 lists the Incyte clones used to assemble full length nucleotide sequences encoding
  • SYNT Columns 1 and 2 show the sequence identification numbers (SEQ ID NOs) of the polypeptide and nucleotide sequences, respectively
  • Column 3 shows the clone IDs of the Incyte clones in which nucleic acids encoding each SYNT were identified
  • column 4 shows the cDNA hbranes from which these clones were isolated
  • Column 5 shows Incyte clones and their corresponding cDNA libraries Clones for which cDNA libraries are not indicated were de ⁇ ved from pooled cDNA hbranes
  • GenBank sequence identifiers are also shown in column 5.
  • the Incyte clones and GenBank cDNA sequences, where indicated, in column 5 were used to assemble the consensus nucleotide sequence of each SYNT and are useful as fragments in hybridization technologies.
  • column 1 references the SEQ ID NO;
  • column 2 shows the number of amino acid residues in each polypeptide,
  • column 3 shows potential phosphorylation sites,
  • column 4 shows potential glycosylation sites;
  • column 5 shows the amino acid residues compnsing signature sequences and motifs;
  • column 6 shows homologous sequences as identified by BLAST analysis, and
  • column 7 shows analytical methods and in some cases, searchable databases to which the analytical methods were applied The methods of column 7 were used to characte ⁇ ze each polypeptide through sequence homology and protein motifs.
  • the columns of Table 3 show the tissue-specificity and diseases, disorders, or conditions associated with nucleotide sequences encoding SYNT.
  • the first column of Table 3 lists the nucleotide SEQ ID NOs.
  • Column 2 lists fragments of the nucleotide sequences of column 1 These fragments are useful, for example, in hybndization or amplification technologies to identify SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO-22, SEQ ID N0 23, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO.29, and SEQ ID NO 30 and to distinguish between SEQ ID NO: 16, SEQ ID NO.17, SEQ ID NO: 18, SEQ ID NO- 19 SEQ ID NO:20, SEQ ID NO:21 , SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:26, S
  • Column 3 lists tissue categories which express SYNT as a fraction of total tissues expressing SYNT
  • Column 4 lists diseases, disorders, or conditions associated with those tissues expressing SYNT as a fraction of total tissues expressing SYNT.
  • Column 5 lists the vectors used to subclone each cDNA library The columns of Table 4 show descnptions of the tissues used to construct the cDNA hbranes from which cDNA clones encoding SYNT were isolated.
  • Column 1 references the nucleotide SEQ ID NOs
  • column 2 shows the cDNA hbranes from which these clones were isolated
  • column 3 shows the tissue origins and other descriptive information relevant to the cDNA hbranes m column 2.
  • SEQ ID NO 16 maps to chromosome 5 within the interval from 147 10 to 150.00 centiMorgans
  • SEQ ID NO: 17 maps to chromosome 10 within the interval from 137.60 to 139.20 centiMorgans This interval also contains gene MXI1, a member of the MYC family.
  • SEQ ID NO 18 maps to chromosome 2 within the interval from 228.80 to 230.10 centiMorgans This interval also contains a gene for a proto-oncogene encoding a tyrosine-protem kinase
  • SEQ ID NO 21 maps to chromosome 5 within the interval from 172.6 to 184 7 centiMorgans
  • SEQ ID NO'24 maps to chromosome 2 within the interval from 1 18 0 to 127.4 centiMorgans.
  • SEQ ID NO 26 maps to chromosome 3 within the interval from 157 4 to 162.0 centiMorgans.
  • SEQ ID NO 27 maps to chromosome 12 within the interval from 97 1 to 1 16.6 centiMorgans.
  • SEQ ID NO 28 maps to chromosome 4 within the interval from 77 3 to 99.2 centiMorgans and to chromosome 5 within the intervals from 79.2 to 92.3 centiMorgans, from 1 16.3 to 127.9 centiMorgans, and from 157.6 to 163 0 centiMorgans
  • SEQ ID NO:29 maps to chromosome 1 within the interval from 242.5 to 258.7 centiMorgans and to chromosome 19 within the interval from 69.9 to 104.9 centiMorgans.
  • SEQ ID NO:30 maps to chromosome 1 within the interval from 57.2 to 57.5 centiMorgans.
  • a preferred SYNT vanant is one which has at least about 80%, or alternatively at least about 90%, or even at least about 95% ammo acid sequence identity to the SYNT ammo acid sequence, and which contains at least one functional or structural charactenstic of SYNT
  • the invention also encompasses polynucleotides which encode SYNT.
  • the invention encompasses a polynucleotide sequence compnsing a sequence selected from the group consisting of SEQ ID NO: 16-30, which encodes SYNT.
  • the polynucleotide sequences of SEQ ID NO: 16-30 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 ⁇ bose instead of deoxynbose.
  • the invention also encompasses a vanant of a polynucleotide sequence encoding SYNT.
  • a vanant polynucleotide sequence will have at least about 70%, or alternatively at least about 85%, or even at least about 95% polynucleotide sequence identity to the polynucleotide sequence encoding SYNT.
  • a particular aspect of the invention encompasses a variant of a polynucleotide sequence compnsing a sequence selected from the group consisting of SEQ ID NO: 16-30 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 NO: 16-30.
  • Any one of the polynucleotide va ⁇ ants desc ⁇ bed above can encode an amino acid sequence which contains at least one functional or structural characte ⁇ stic of SYNT.
  • nucleotide sequences which encode SYNT and its vanants are generally capable of hybndizing to the nucleotide sequence of the naturally occumng SYNT under appropnately selected conditions of st ⁇ ngency, it may be advantageous to produce nucleotide sequences encoding SYNT or its de ⁇ vatives possessing a substantially different codon usage, e.g., inclusion of non-naturally occumng 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.
  • RNA transc ⁇ pts having more desirable properties, such as a greater half-life, than transcnpts produced from the naturally occumng sequence
  • the invention also encompasses production of DNA sequences which encode SYNT and SYNT denvatives, or fragments thereof, entirely by synthetic chemistry.
  • the synthetic sequence may be inserted into any of the many available expression vectors and cell systems using reagents well known in the art.
  • synthetic chemistry may be used to introduce mutations into a sequence encoding SYNT or any fragment thereof.
  • polynucleotide sequences that are capable of hybridizing to the claimed polynucleotide sequences, and, in particular, to those shown in SEQ ID NO: 16-30 and fragments thereof under various conditions of stnngency.
  • Hybridization conditions including annealing and wash conditions, are described in "Definitions.”
  • sequence preparation is automated with machines such as the MICROLAB 2200 liquid transfer system (Hamilton, Reno NV), PTC200 thermal cycler (MJ Research, Watertown MA) and ABI CATALYST 800 thermal cycler (PE Biosystems).
  • Sequencing is then earned out using either the ABI 373 or 377 DNA sequencing system (PE 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 vanety of algonthms which are well known in the art. (See, e.g.,
  • the nucleic acid sequences encoding SYNT 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
  • restriction-site PCR uses universal and nested pnmers to amplify unknown sequence from genomic DNA within a cloning vector (See, e g , Sarkar, G ( 1993) PCR Methods Apphc 2 318-322 )
  • Another method, inverse PCR uses primers that extend in divergent directions to amplify unknown sequence from a circula ⁇ zed template The template is denved from restnction fragments compnsing a known genomic locus and surrounding sequences (See, e g , Tngha, T et al ( 1988) Nucleic Acids Res 16 8186 )
  • capture PCR involves PCR amplification of DNA fragments adjacent to known sequences in human and yeast artificial chromosome DNA (See, e g , Tngha
  • hbranes When screening for full-length cDNAs, it is preferable to use hbranes that have been size-selected to include larger cDNAs.
  • random-p ⁇ med hbranes which often include sequences containing the 5' regions of genes, are preferable for situations in which an ohgo d(T) library does not yield a full-length cDNA Genomic hbranes may be useful for extension of sequence into 5' non-transcnbed 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
  • 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
  • Output light intensity may be converted to elect ⁇ cal signal using appropnate software (e g , GENOTYPER and SEQUENCE NAVIGATOR, PE 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
  • polynucleotide sequences or fragments thereof which encode SYNT may be cloned in recombinant DNA molecules that direct expression of SYNT, or fragments or functional equivalents thereof, in appropnate 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 SYNT
  • nucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter SYNT-encoding sequences for a vanety 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 ohgonucleotides may be used to engineer the nucleotide sequences.
  • ohgonucleotide- mediated site-directed mutagenesis may be used to introduce mutations that create new rest ⁇ ction sites, alter glycosylation patterns, change codon preference, produce splice vanants, and so forth
  • the nucleotides of the present invention may be subjected to DNA shuffling techniques such as MOLECULARBREEDING (Maxygen Inc., Santa Clara CA: desc ⁇ bed in U.S Patent Number 5,837,458, Chang, C -C. et al ( 1999) Nat. Biotechnol. 17:793-797; Ch ⁇ stians, F.C. et al. (1999) Nat. Biotechnol. 17.259-264; and Crame ⁇ , A. et al. ( 1996) Nat. Biotechnol 14-315-319) to alter or improve the biological properties of SYNT, such as its biological or enzymatic activity or its ability to bind to other molecules or compounds.
  • MOLECULARBREEDING Maxygen Inc., Santa Clara CA: desc ⁇ bed in U.S Patent Number 5,837,458, Chang, C -C. et al ( 1999) Nat. Biotechnol. 17:793-797; Ch ⁇ stians, F
  • DNA shuffling is a process by which a library of gene va ⁇ ants is produced using PCR-mediated recombination of gene fragments. The library is then subjected to selection or screening procedures that identify those gene vanants with the desired properties. These preferred vanants may then be pooled and further subjected to recursive rounds of DNA shuffling and selection/screening.
  • 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.
  • 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 occumng genes in a directed and controllable manner.
  • sequences encoding SYNT 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 )
  • SYNT itself or a fragment thereof may be synthesized using chemical methods.
  • peptide synthesis can be performed using vanous 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.
  • the nucleotide sequences encoding SYNT or de ⁇ vatives thereof may be inserted into an appropnate expression vector, I e., a vector which contains the necessary elements for transcnptional and translational control of the inserted coding sequence in a suitable host
  • appropnate expression vector I e., a vector which contains the necessary elements for transcnptional 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 SYNT
  • Such elements may vary in their strength and specificity Specific initiation signals may also be used to achieve more efficient translation of sequences encoding SYNT.
  • Such signals include the ATG initiation codon and adjacent sequences, e.g the Kozak sequence.
  • sequences encoding SYNT and its initiation codon and upstream regulatory sequences are inserted into the appropriate expression vector, no additional transcnptional or translational control signals may be needed
  • 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 vanous o ⁇ gins, 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.
  • a va ⁇ ety of expression vector/host systems may be utilized to contain and express sequences encoding SYNT
  • microorganisms such as bactena transformed with recombinant bactenophage, 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 bacte ⁇ al expression vectors (e.g., Ti or pBR322 plasmids); or animal cell systems.
  • microorganisms such as bactena transformed with recombinant bactenophage, 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, Ca
  • Expression vectors denved from retroviruses, adenoviruses, or herpes or vaccinia viruses, or from va ⁇ ous bactenal plasmids may be used for delivery of nucleotide sequences to the targeted organ, tissue, or cell population.
  • the invention is not limited by the host cell employed.
  • a number of cloning and expression vectors may be selected depending upon the use intended for polynucleotide sequences encoding SYNT.
  • routine cloning, subcloning, and propagation of polynucleotide sequences encoding SYNT can be achieved using a multifunctional E. coli vector such as PBLUESCRIPT (Stratagene, La Jolla CA) or PSPORT1 plasmid (Life Technologies). Ligation of sequences encoding SYNT into the vector's multiple cloning site disrupts the lac gene, allowing a colo ⁇ met ⁇ c screening procedure for identification of transformed bactena containing recombinant molecules.
  • 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.
  • vectors which direct high level expression of SYNT may be used.
  • vectors containing the strong, inducible T5 or T7 bactenophage promoter may be used.
  • Yeast expression systems may be used for production of SYNT.
  • 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 pastons.
  • 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.
  • Plant systems may also be used for expression of SYNT.
  • Transcnption of sequences encoding SYNT may be d ⁇ ven 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 6:307-31 1).
  • plant promoters such as the small subunit of RUBISCO or heat shock promoters may be used.
  • These constructs can be introduced into plant cells by direct DNA transformation or pathogen-mediated transfection. (See, e.g., The McGraw Hill Yearbook of Science and Technology ( 1992) McGraw Hill, New York NY, pp. 191-196 )
  • a number of viral-based expression systems may be utilized.
  • sequences encoding SYNT may be gated into an adenovirus transcnption/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential El or E3 region of the viral genome may be used to obtain infective virus which expresses SYNT in host cells.
  • transcnption enhancers such as the Rous sarcoma virus (RSV) enhancer
  • RSV Rous sarcoma virus
  • SV40 or EBV- based vectors may also be used for high-level protein expression
  • HACs Human artificial chromosomes
  • 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, polycatio c amino polymers, or vesicles) for therapeutic purposes.
  • liposomes, polycatio c amino polymers, or vesicles for therapeutic purposes.
  • stable expression of SYNT in cell lines is preferred.
  • sequences encoding SYNT can be transformed into cell lines using expression vectors which may contain viral ongins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector.
  • cells may be allowed to grow for about 1 to 2 days ennched 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 appropnate 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 phosphonbosyltransferase genes, for use in tk and apr cells, respectively (See, e.g., Wigler, M. et al. (1977) Cell 1 1 :223-232, Lowy, I. et al. (1980) Cell 22.817-823 )
  • antimetabohte, antibiotic, or herbicide resistance can be used as the basis for selection.
  • dhfr confers resistance to methotrexate
  • neo confers resistance to the aminoglycosides neomycm and G-418
  • als and pat confer resistance to chlorsulfuron and phosphinotncin acetyltransferase, respectively.
  • Visible markers e.g., anthocyamns, green fluorescent proteins (GFP; Clontech), ⁇ glucuromdase and its substrate ⁇ -glucuronide, or luciferase and its substrate lucife ⁇ n 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, CA. ( 1995) Methods Mol. Biol. 55 121-131.)
  • marker gene expression suggests that the gene of interest is also present, the presence and expression of the gene may need to be confirmed.
  • sequence encoding SYNT is inserted within a marker gene sequence
  • transformed cells containing sequences encoding SYNT can be identified by the absence of marker gene function.
  • a marker gene can be placed in tandem with a sequence encoding SYNT 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
  • SYNT 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 hybndizations, 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 SYNT using either specific polyclonal or monoclonal antibodies are known in the art.
  • ELISAs enzyme-linked immunosorbent assays
  • RIAs radioimmunoassays
  • FACS fluorescence activated cell sorting
  • va ⁇ ety of labels and conjugation techniques are known by those skilled in the art and may be used in va ⁇ ous nucleic acid and amino acid assays.
  • Means for producing labeled hybndization or PCR probes for detecting sequences related to polynucleotides encoding SYNT include oligolabehng, nick translation, end-labeling, or PCR amplification using a labeled nucleotide
  • the sequences encoding SYNT, or any fragments thereof may be cloned into a vector for the production of an mRNA probe.
  • RNA probes are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by addition of an appropnate RNA polymerase such as T7, T3, or SP6 and labeled nucleotides
  • an appropnate RNA polymerase such as T7, T3, or SP6 and labeled nucleotides
  • Host cells transformed with nucleotide sequences encoding SYNT 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
  • expression vectors containing polynucleotides which encode SYNT may be designed to contain signal sequences which direct secretion of SYNT through a prokaryotic or eukaryotic cell membrane
  • 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
  • modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, hpidation, 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 charactenstic mechanisms for post-translational activities (e.g., CHO, HeLa, MDCK, HEK293, and WI38) 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.
  • ATCC American Type Culture Collection
  • natural, modified, or recombinant nucleic acid sequences encoding SYNT may be hgated to a heterologous sequence resulting in translation of a fusion protein in any of the aforementioned host systems.
  • a chimeric SYNT protein containing a heterologous moiety that can be recognized by a commercially available antibody may facilitate the screening of peptide hbranes for inhibitors of SYNT activity.
  • Heterologous protein and peptide moieties may also facilitate purification of fusion proteins using commercially available affinity matnces.
  • Such moieties include, but are not limited to, glutathione S-transferase (GST), maltose binding protein (MBP), thioredoxin (Trx), calmodulin binding peptide (CBP), 6-H ⁇ s, FLAG, c-myc, and hemagglutimn (HA) GST, MBP, Trx, CBP, and 6-H ⁇ s enable punfication of their cognate fusion proteins on immobilized glutathione, maltose, phenylarsine oxide, calmodulin, and metal-chelate resins, respectively.
  • GST glutathione S-transferase
  • MBP maltose binding protein
  • Trx thioredoxin
  • CBP calmodulin binding peptide
  • 6-H ⁇ s 6-H ⁇ s
  • FLAG c-myc
  • hemagglutimn (HA) GST, MBP, Trx, CBP, and 6-H ⁇ s enable punfication of their cognate fusion proteins on
  • FLAG, c-myc, and hemagglutimn (HA) enable immunoaffinity punfication 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 SYNT encoding sequence and the heterologous protein sequence, so that SYNT may be cleaved away from the heterologous moiety following purification Methods for fusion protein expression and punfication are discussed in Ausubel ( 1995, supra, ch. 10).
  • a va ⁇ ety of commercially available kits may also be used to facilitate expression and punfication of fusion proteins.
  • synthesis of radiolabeled SYNT may be achieved in vitro using the TNT rabbit reticulocyte lysate or wheat germ extract system (Promega). These systems couple transcnption and translation of protein-coding sequences operably associated with the T7, T3, or SP6 promoters. Translation takes place in the presence of a radiolabeled amino acid precursor, for example, 35 S-meth ⁇ on ⁇ ne.
  • SYNT of the present invention or fragments thereof may be used to screen for compounds that specifically bind to SYNT.
  • At least one and up to a plurality of test compounds may be screened for specific binding to SYNT.
  • Examples of test compounds include antibodies, o gonucleotides, proteins (e.g., receptors), or small molecules
  • the compound thus identified is closely related to the natural ligand of SYNT, e.g , a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a natural binding partner (See, Cohgan, J.E. et al. ( 1991) Current Protocols in Immunology 1(2) Chapter 5.)
  • the compound can be closely related to the natural receptor to which SYNT binds, or to at least a fragment of the receptor, e.g., the ligand binding site.
  • screening for these compounds involves producing appropnate cells which express SYNT, either as a secreted protein or on the cell membrane Preferred cells include cells from mammals, yeast, Drosophila, or E. coli. Cells expressing SYNT or cell membrane fractions which contain SYNT are then contacted with a test compound and binding, stimulation, or inhibition of activity of either SYNT 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.
  • the assay may comp ⁇ se the steps of combining at least one test compound with SYNT, either in solution or affixed to a solid support, and detecting the binding of SYNT to the compound.
  • the assay may detect or measure binding of a test compound in the presence of a labeled competitor.
  • the assay may be earned out using cell-free preparations, chemical hbranes, or natural product mixtures, and the test compound(s) may be free in solution or affixed to a solid support.
  • SYNT of the present invention or fragments thereof may be used to screen for compounds that modulate the activity of SYNT.
  • Such compounds may include agonists, antagonists, or partial or inverse agonists.
  • an assay is performed under conditions permissive for SYNT activity, wherein SYNT is combined with at least one test compound, and the activity of SYNT in the presence of a test compound is compared with the activity of SYNT in the absence of the test compound.
  • a change in the activity of SYNT in the presence of the test compound is indicative of a compound that modulates the activity of SYNT
  • a test compound is combined with an in vitro or cell-free system compnsing SYNT under conditions suitable for SYNT activity, and the assay is performed In either of these assays, a test compound which modulates the activity of SYNT 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
  • polynucleotides encoding SYNT or their mammalian homologs may be "knocked out" in an animal model system using homologous recombination in embryonic stem (ES) cells
  • ES embryonic stem
  • 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 No 5, 175,383 and U S Patent No 5,767,337 )
  • mouse ES cells such as the mouse 129/SvJ cell line, are de ⁇ ved from the early mouse embryo and grown m 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
  • Polynucleotides encoding SYNT 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 1 145-1 147)
  • Polynucleotides encoding SYNT can also be used to create "knockin" humanized animals (pigs) or transgenic animals (mice or rats) to model human disease
  • knockin technology a region of a polynucleotide encoding SYNT 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 descnbed above Transgenic progeny or inbred lines are studied and treated with potential pharmaceutical agents to obtain information on treatment of a human disease
  • a mammal inbred to overexpress SYNT, e g by secreting SYNT 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
  • SYNT appears to play a role in an immune disorder such as inflammation, actinic keratosis, acquired immunodeficiency syndrome (AIDS), Addison s disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, arteriosclerosis, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, bronchitis, bursitis, cholecystitis, cirrhosis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes melhtus, emphysema,
  • an immune disorder such as inflammation, actinic keratosis, acquired immunodeficiency syndrome (AIDS), Addison s disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, arteriosclerosis, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis
  • a vector capable of expressing SYNT or a fragment or denvative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of SYNT including, but not limited to, those described above
  • a pharmaceutical composition comprising a substantially punfied
  • SYNT in conjunction with a suitable pharmaceutical earner may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of SYNT including, but not limited to, those provided above
  • an agonist which modulates the activity of SYNT may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of SYNT including, but not limited to, those listed above
  • an antagonist of SYNT may be administered to a subject to treat or prevent a disorder associated with increased expression or activity of SYNT Examples of such disorders include, but are not limited to, those immune, neuronal, reproductive, and cell prohferative disorders descnbed above
  • an antibody which specifically binds SYNT may be used directly as an antagonist or indirectly as a targeting or delivery mechanism for bnnging a pharmaceutical agent to cells or tissues which express SYNT
  • a vector expressing the complement of the polynucleotide encoding SYNT may be administered to a subject to treat or prevent a disorder associated with increased expression or activity of SYNT including, but not limited to, those descnbed above
  • any of the proteins, antagonists, antibodies, agonists, complementary sequences, or vectors of the invention may be administered m combination with other appropriate therapeutic agents
  • Selection of the appropnate 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 vanous disorders desc ⁇ bed 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 SYNT may be produced using methods which are generally known in the art
  • punfied SYNT may be used to produce antibodies or to screen hbranes of pharmaceutical agents to identify those which specifically bind SYNT
  • Antibodies to SYNT may also be generated using methods that are well known in the art
  • Such antibodies may include, but are not limited to, polyclonal, monoclonal, chimenc, and single chain antibodies, Fab fragments, and fragments produced by a Fab expression library
  • Neutralizing antibodies I e , those which inhibit dimer formation
  • va ⁇ ous hosts including goats, rabbits, rats, mice, humans, and others may be immunized by injection with SYNT or with any fragment or ohgopeptide thereof which has immunogenic properties
  • 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.
  • BCG Bacilli Calmette-Guenn
  • Corynebacte ⁇ um parvum are especially preferable
  • the ohgopeptides, peptides, or fragments used to induce antibodies to SYNT have an ammo acid sequence consisting of at least about 5 amino acids, and generally will consist of at least about 10 amino acids.
  • these ohgopeptides, peptides, or fragments are identical to a portion of the amino acid sequence of the natural protein Short stretches of SYNT ammo acids may be fused with those of another protein, such as KLH, and antibodies to the chimenc molecule may be produced
  • Monoclonal antibodies to SYNT 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 hybndoma technique, the human B-cell hyb ⁇ doma technique, and the EBV-hyb ⁇ doma technique (See, e.g., Kohler, G. et al. (1975) Nature 256 495-497; Kozbor, D. et al. ( 1985) J
  • chimenc 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.
  • Antibodies may also be produced by inducing in vivo production m the lymphocyte population or by screening immunoglobulin hbranes 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 SYNT may also be generated
  • 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 bndges of the F(ab')2 fragments.
  • Fab expression hbranes 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.
  • K a which is defined as the molar concentration of SYNT-antibody complex divided by the molar concentrations of free antigen and free antibody under equihb ⁇ um conditions
  • K a association constant
  • the K a determined for a preparation of monoclonal antibodies, which are monospecific for a particular SYNT epitope represents a true measure of affinity High-affinity antibody preparations with K, ranging from about 10 9 to 10 12 L/mole are preferred for use in immunoassays in which the SYNT-antibody complex must withstand ⁇ gorous manipulations
  • Low-affinity antibody preparations with K a ranging from about 10 6 to 10 7 L/
  • polyclonal antibody preparations may be further evaluated to determine the quality and suitability of such preparations for certain downstream applications
  • 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 requi ⁇ ng precipitation of SYNT-antibody complexes
  • Procedures for evaluating antibody specificity, titer, and avidity, and guidelines for antibody quality and usage in vanous applications are generally available (See, e g , Catty, supra, and Cohgan et al , supra )
  • the polynucleotides encoding SYNT, or any fragment or complement thereof may be used for therapeutic purposes
  • modifications of gene expression can be achieved by designing complementary sequences or antisense molecules (DNA, RNA, PNA, or modified ohgonucleotides) to the coding or regulatory regions of the gene encoding SYNT
  • DNA, RNA, PNA, or modified ohgonucleotides DNA, RNA, P
  • Antisense sequences can be delivered mtracellularly 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.
  • Antisense sequences can also be introduced mtracellularly 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.)
  • viral vectors such as retrovirus and adeno-associated virus vectors
  • Other gene delivery mechanisms include hposome-de ⁇ ved systems, artificial viral envelopes, and other systems known m the art (See, e.g., Rossi, J.J. ( 1995) Br. Med. Bull. 51(l):217-225; Boado, R.J. et al. ( 1998) J. Pharm. Sci. 87(1 1)- 1308-1315; and Moms, M.C. et al (1997) Nucleic Acids Res 25(14):2730-2736.)
  • polynucleotides encoding SYNT may be used for somatic or germline gene therapy.
  • Gene therapy may be performed to (l) correct a genetic deficiency (e.g., in the cases of severe combined immunodeficiency (SCID)-Xl disease characte ⁇ zed by X- hnked inhentance (Cavazzana-Calvo, M. et al. (2000) Science 288-669-672), severe combined immunodeficiency syndrome associated with an inhented 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
  • conditionally lethal gene product e.g., in the case of cancers which result from unregulated cell proliferation
  • 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.
  • hepatitis B or C virus HBV, HCV
  • fungal parasites such as Candida albicans and Paracoccidioides brasihensis, and protozoan parasites such as Plasmodium falciparum and Trypanosoma cruzi.
  • the expression of SYNT from an appropnate population of transduced cells may alleviate the clinical manifestations caused by the genetic deficiency
  • diseases or disorders caused by deficiencies in SYNT are treated by constructing mammalian expression vectors encoding SYNT and introducing these vectors by mechanical means into SYNT-deficient cells.
  • Mechanical transfer technologies for use with cells in vivo or ex vitro include (I) direct DNA micromjection into individual cells, (n) ballistic gold particle delivery, (in) hposome-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.
  • Expression vectors that may be effective for the expression of SYNT 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).
  • SYNT may be expressed using (I) a constitutively active promoter, (e.g., from cytomegalovirus (CMV), Rous sarcoma virus (RSV), SV40 virus, thymidine kinase (TK), or ⁇ -actin genes), (n) an inducible promoter (e.g , the tetracychne-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) Curr. Opin. Biotechnol.
  • a constitutively active promoter e.g., from cytomegalovirus (CMV), Rous sarcoma virus (RSV), SV40 virus, thymidine kinase (TK), or ⁇ -actin genes
  • liposome transformation kits e.g., the PERFECT LIPID TRANSFECTION KIT, available from Invitrogen
  • PERFECT LIPID TRANSFECTION KIT available from Invitrogen
  • 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 pnmary cells requires modification of these standardized mammalian transfection protocols.
  • diseases or disorders caused by genetic defects with respect to SYNT expression are treated by constructing a retrovirus vector consisting of (1) the polynucleotide encoding SYNT under the control of an independent promoter or the retrovirus long terminal repeat (LTR) promoter, (n) appropnate RNA packaging signals, and (in) a Rev-responsive element (RRE) along with additional retrovirus cu-acting RNA sequences and coding sequences required for efficient vector propagation.
  • Retrovirus vectors e.g., PFB and PFBNEO
  • Retrovirus vectors are commercially available (Stratagene) and are based on published data (Riviere, I. et al. (1995) Proc Natl. Acad. Sci.
  • the vector is propagated 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.
  • VPCL vector producing cell line
  • U.S. Patent Number 5,910,434 to Rigg 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, ML.
  • an adenovirus-based gene therapy delivery system is used to deliver polynucleotides encoding SYNT to cells which have one or more genetic abnormalities with respect to the expression of SYNT.
  • 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.
  • adenoviral vectors are descnbed in U.S. Patent Number 5,707,618 to Armentano ("Adenovirus vectors for gene therapy"), hereby incorporated by reference.
  • Antmozzi P.A. et al. ( 1999) Annu. Rev. Nutr. 19:51 1-544; and Verma, I.M. and N. Somia ( 1997) Nature 18:389:239-242, both incorporated by reference herein.
  • a herpes-based, gene therapy delivery system is used to deliver polynucleotides encoding SYNT to target cells which have one or more genetic abnormalities with respect to the expression of SYNT
  • the use of herpes simplex virus (HSV)-based vectors may be especially valuable for introducing SYNT 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 p ⁇ mates (Liu, X. et al. (1999) Exp. Eye Res.169:385-395).
  • HSV-1 virus vector has also been disclosed m 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 appropnate promoter for purposes including human gene therapy
  • recombinant HSV strains deleted for ICP4, ICP27 and ICP22 are also taught by this patent.
  • For HSV vectors see also Goins, W F. et al. ( 1999) J Virol.
  • an alphavirus (positive, single-stranded RNA virus) vector is used to deliver polynucleotides encoding SYNT to target cells
  • SFV Semliki Forest Virus
  • 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 SYNT into the alphavirus genome in place of the capsid-coding region results in the production of a large number of SYNT-codmg RNAs and the synthesis of high levels of SYNT in vector transduced cells.
  • enzymatic activity e g., protease and polymerase
  • 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 vanant of Smdbis 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 SYNT into a vanety of cell types
  • the specific transduction of a subset of cells in a population may require the sorting of cells p ⁇ or 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 de ⁇ ved from the transcnption initiation site may also be employed to inhibit gene expression.
  • inhibition can be achieved using t ⁇ ple helix base-painng methodology T ⁇ ple helix pai ⁇ ng is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transc ⁇ ption factors, or regulatory molecules.
  • t ⁇ plex DNA Recent therapeutic advances using t ⁇ plex DNA have been descnbed in the literature. (See, e.g., Gee, J.E. et al. (1994) in Huber, B.E. and B.I. Carr, Molecular and Immunologic 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 transcnpt from binding to nbosomes.
  • Ribozymes enzymatic RNA molecules
  • Ribozymes may also be used to catalyze the specific cleavage of RNA
  • the mechanism of ⁇ bozyme action involves sequence-specific hybndization of the nbozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • engineered hammerhead motif nbozyme molecules may specifically and efficiently catalyze endonucleolytic cleavage of sequences encoding SYNT
  • nbozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for nbozyme cleavage sites, including the following sequences GUA, GUU, and GUC Once identified, short RNA sequences of between 15 and 20 ⁇ bonucleotides, 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 hybndization with complementary o gonucleotides using nbonuclease protection assays
  • RNA molecules may be generated by in vitro and in vivo transcnption of DNA sequences encoding SYNT Such DNA sequences may be incorporated into a wide vanety 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
  • An additional embodiment of the invention encompasses a method for screening for a compound which is effective m alternateng expression of a polynucleotide encoding SYNT
  • Compounds which may be effective in alternativeng expression of a specific polynucleotide may include, but are not limited to, ohgonucleotides, antisense ohgonucleotides, tnple helix-forming ohgonucleotides, transc ⁇ ption factors and other polypeptide transcnptional 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
  • a compound which specifically inhibits expression of the polynucleotide encoding SYNT may be therapeutically useful, and in the treament of disorders associated with decreased SYNT expression
  • Alterations in the expression of a polynucleotide encoding SYNT are assayed by any method commonly known in the art Typically, the expression of a specific nucleotide is detected by hybndization with a probe having a nucleotide sequence complementary to the sequence of the polynucleotide encoding SYNT The amount of hybndization may be quantified, thus forming the basis for a compa ⁇ son 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 alternativeng the expression of the polynucleotide.
  • a screen for a compound effective in alternativeng expression of a specific polynucleotide can be earned out, for example, using a Schizosaccharomyces 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 Commun. 268 8- 13).
  • a particular embodiment of the present invention involves screening a combinato ⁇ al library of ohgonucleotides (such as deoxynbonucleotides, ⁇ bonucleotides, peptide nucleic acids, and modified ohgonucleotides) 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)
  • ohgonucleotides such as deoxynbonucleotides, ⁇ bonucleotides, peptide nucleic acids, and modified ohgonucleotides
  • 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. Biotechnol 15-462-466 ) Any of the therapeutic methods descnbed 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 pharmaceutical composition which generally compnses an active ingredient formulated with a pharmaceutically acceptable excipient
  • Excipients may include, for example, sugars, starches, celluloses, gums, and proteins Va ⁇ ous formulations are commonly known and are thoroughly discussed in the latest edition of Remington's Pharmaceutical Sciences (Maack Publishing, Easton PA).
  • Such pharmaceutical compositions may consist of SYNT, antibodies to SYNT, and mimetics, agonists, antagonists, or inhibitors of SYNT
  • compositions utilized in this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, lntra-arte ⁇ al, intramedullary, intrathecal, intravent ⁇ cular, pulmonary, transdermal, subcutaneous, intrapentoneal, 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 pnor to inhalation by the patient.
  • aerosol delivery of fast-acting formulations is well-known in the art.
  • macromolecules e g larger peptides and proteins
  • 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 m 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 pharmaceutical compositions may be prepared for direct intracellular delivery of macromolecules compnsing SYNT or fragments thereof.
  • liposome preparations containing a cell-impermeable macromolecule may promote cell fusion and intracellular delivery of the macromolecule.
  • SYNT 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).
  • 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 SYNT or fragments thereof, antibodies of SYNT, and agonists, antagonists or inhibitors of SYNT, which ameliorates the symptoms or condition.
  • Therapeutic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or with expe ⁇ mental animals, such as by calculating the ED 50 (the dose therapeutically effective in 50% of the population) or LD 50 (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 LD 50 /ED, 0 ratio
  • Pharmaceutical 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 ED 50 with little or no toxicity
  • 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 seventy of the disease state, the general health of the subject, the age, weight, and gender of the subject, time and frequency of administration, drug combmat ⁇ on(s), reaction sensitivities, and response to therapy
  • Long-acting pharmaceutical 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 ⁇ g to 100,000 ⁇ g, 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
  • antibodies which specifically bind SYNT may be used for the diagnosis of disorders characte ⁇ zed by expression of SYNT, or in assays to monitor patients being treated with SYNT or agonists, antagonists, or inhibitors of SYNT
  • Antibodies useful for diagnostic purposes may be prepared in the same manner as desc ⁇ bed above for therapeutics Diagnostic assays for SYNT include methods which utilize the antibody and a label to detect SYNT m human body fluids or m 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 va ⁇ ety of reporter molecules, several of which are descnbed above, are known in the art and may be used
  • a variety of protocols for measunng SYNT including ELISAs, RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of SYNT expression
  • Normal or standard values for SYNT expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, for example, human subjects, with antibody to SYNT under conditions suitable for complex formation
  • the amount of standard complex formation may be quantitated by va ⁇ ous methods, such as photometnc means Quantities of SYNT expressed in subject, control, and disease samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease.
  • the polynucleotides encoding SYNT may be used for diagnostic purposes.
  • the polynucleotides which may be used include ohgonucleotide 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 SYNT may be correlated with disease
  • the diagnostic assay may be used to determine absence, presence, and excess expression of SYNT, and to monitor regulation of SYNT levels during therapeutic intervention
  • hybndization with PCR probes which are capable of detecting polynucleotide sequences, including genomic sequences, encoding SYNT or closely related molecules may be used to identify nucleic acid sequences which encode SYNT.
  • 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 hybndization or amplification will determine whether the probe identifies only naturally occurring sequences encoding SYNT, allehc vanants, 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 SYNT encoding sequences
  • the hybridization probes of the subject invention may be DNA or RNA and may be denved from the sequence of SEQ ID NO- 16-30 or from genomic sequences including promoters, enhancers, and introns of the SYNT gene
  • Means for producing specific hybndization probes for DNAs encoding SYNT include the cloning of polynucleotide sequences encoding SYNT or SYNT denvatives 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 appropnate RNA polymerases and the appropriate labeled nucleotides
  • Hybndization probes may be labeled by a va ⁇ ety of reporter groups, for example, by radionuchdes such as 32 P or
  • Polynucleotide sequences encoding SYNT may be used for the diagnosis of disorders associated with expression of SYNT
  • disorders include, but are not limited to, an immune disorder such as inflammation, actinic keratosis, acquired immunodeficiency syndrome (AIDS), Addison 's disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, arte ⁇ osclerosis, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, bronchitis, bursitis, cholecystitis, cirrhosis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes melhtus, emphysema, erythroblastosis fetahs, erythema nodosum, atrophic gast ⁇ tis, glomerulonephntis, Goodpasture's syndrome, gout,
  • the nucleotide sequences encoding SYNT may be useful m assays that detect the presence of associated disorders, particularly those mentioned above
  • the nucleotide sequences encoding SYNT may be labeled by standard methods and added to a fluid or tissue sample from a patient under conditions suitable for the formation of hybndization complexes After a suitable incubation penod, the sample is washed and the signal is quantified and compared with a standard value If the amount of signal m the patient sample is significantly altered in companson to a control sample then the presence of altered levels of nucleotide sequences encoding SYNT m 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 tnals, 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
  • SYNT 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 SYNT, under conditions suitable for hybndization or amplification. Standard hybndization may be quantified by companng the values obtained from normal subjects with values from an expenment in which a known amount of a substantially punfied 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
  • hybridization assays may be repeated on a regular basis to determine 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 penod ranging from several days to months
  • the presence of an abnormal amount of transcnpt (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 p ⁇ or 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 ohgonucleotides designed from the sequences encoding SYNT may involve the use of PCR.
  • Ohgomers may be chemically synthesized, generated enzymatically, or produced m vitro
  • Ohgomers will preferably contain a fragment of a polynucleotide encoding SYNT, or a fragment of a polynucleotide complementary to the polynucleotide encoding SYNT, and will be employed under optimized conditions for identification of a specific gene or condition.
  • Ohgomers may also be employed under less stnngent conditions for detection or quantification of closely related DNA or RNA sequences
  • ohgonucleotide pnmers derived from the polynucleotide sequences encoding SYNT may be used to detect single nucleotide polymorphisms (SNPs).
  • SNPs are substitutions, insertions and deletions that are a frequent cause of inhe ⁇ ted or acquired genetic disease in humans
  • Methods of SNP detection include, but are not limited to, single-stranded conformation polymorphism (SSCP) and fluorescent SSCP (fSSCP) methods
  • SSCP single-stranded conformation polymorphism
  • fSSCP fluorescent SSCP
  • ohgonucleotide p ⁇ mers denved from the polynucleotide sequences encoding SYNT are used to amplify DNA using the polymerase chain reaction (PCR)
  • PCR polymerase chain reaction
  • the DNA may be denved, for example, from diseased or normal tissue, biopsy samples, bodily fluids, and the like SNPs in the DNA cause differences
  • Methods which may also be used to quantify the expression of SYNT include radiolabelmg or biotinylatmg nucleotides, coamphfication of a control nucleic acid, and interpolating results from standard curves (See, e g , Melby, P C et al ( 1993) J Immunol.
  • ohgonucleotides or longer fragments denved from any of the polynucleotide sequences descnbed herein may be used as elements on a microarray
  • the microarray can be used in transcnpt imaging techniques which monitor the relative expression levels of large numbers of genes simultaneously as descnbed in Seilhamer, J J et al , "Comparative Gene Transcnpt Analysis," U S Patent No 5,840,484, incorporated herein by reference
  • the microarray may also be used to identify genetic va ⁇ ants, 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 appropnate and effective treatment regimen for that patient For example, therapeutic agents which are highly effective and display
  • antibodies specific for SYNT, or SYNT or fragments thereof may be used as elements on a microarray
  • the microarray may be used to monitor or measure protein-protem interactions, drug-target interactions, and gene expression profiles, as descnbed 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.
  • a transcnpt image may be generated by hybridizing the polynucleotides of the present invention or their complements to the totality of transc ⁇ pts or reverse transc ⁇ pts of a particular tissue or cell type.
  • the hybndization takes place in high-throughput format, wherein the polynucleotides of the present invention or their complements compnse a subset of a plurality of elements on a microarray
  • the resultant transcnpt image would provide a profile of gene activity.
  • Transcript images may be generated using transc ⁇ pts isolated from tissues, cell lines, biopsies, or other biological samples
  • the transcnpt image may thus reflect gene expression in vivo, as in the case of a tissue or biopsy sample, or vitro, as m the case of a cell line
  • Transcnpt images which profile the expression of the polynucleotides of the present invention may also be used m conjunction with in vitro model systems and prechnical evaluation of pharmaceuticals, as well as toxicological testing of indust ⁇ al and naturally-occurring environmental compounds All compounds induce charactenstic gene expression patterns, frequently termed molecular fingerprints or toxicant signatures, which are indicative of mechanisms of action and toxicity (Nuwaysir, E.F. et al.
  • 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 hyb ⁇ dized with one or more probes specific to the polynucleotides of the present invention, so that transcnpt levels corresponding to the polynucleotides of the present invention may be quantified
  • the transcnpt levels in the treated biological sample are compared with levels in an untreated biological sample Differences in the transcnpt levels between the two samples are indicative of a toxic response caused by the test compound in the treated sample
  • proteome refers to the global pattern of protein expression in a particular tissue or cell type
  • 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
  • the separation is achieved using two-dimensional gel electrophoresis, in which proteins from a sample are separated by isoelectnc focusing in the first dimension, and then according to molecular weight by sodium dodecyl sulfate slab gel electrophoresis in the second dimension (Sterner 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
  • a proteomic profile may also be generated using antibodies specific for SYNT to quantify the levels of SYNT expression
  • 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- 1 1 1 , Mendoze, L G et al (1999) Biotechniques 27 778-788)
  • Detection may be performed by a va ⁇ ety of methods known in the art, for example, by reacting the proteins in the sample with a thiol- or ammo-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 transcnpt level There is a poor correlation between transcnpt and protein abundances for some proteins in some tissues (Anderson,
  • proteome toxicant signatures may be useful in the analysis of compounds which do not significantly affect the transcnpt image, but which alter the proteomic profile.
  • 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
  • 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 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.
  • 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 m the art.
  • methods known m 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 W095/251 1 16; Shalon, D et al ( 1995) PCT application WO95/35505; 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 desc ⁇ bed in DNA Microarrays. A Practical Approach. M. Schena, ed. (1999) Oxford University Press, London, hereby expressly incorporated by reference.
  • nucleic acid sequences encoding SYNT may be used to generate hybndization probes useful in mapping the naturally occumng genomic sequence
  • Either coding or noncodmg sequences may be used, and in some instances, noncodmg sequences may be preferable over coding sequences.
  • sequences of the invention 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), bactenal artificial chromosomes (BACs), bactenal PI constructions, or single chromosome cDNA libraries (See, e g , Har ⁇ ngton, J J et al ( 1997) Nat Genet 15 345-355 Pnce, C M ( 1993) Blood Rev 7 127-134, and Trask, B J ( 1991) Trends Genet 7 149-154 )
  • the nucleic acid sequences of the invention may be used to develop genetic linkage maps, for example, which correlate the mhentance of a disease state with the inheritance of
  • Fluorescent in situ hybndization may be correlated with other physical and genetic map data (See, e g , Heinz-Ul ⁇ ch, et al ( 1995) in Meyers, supra, pp 965-968 ) Examples of genetic map data can be found m va ⁇ ous scientific journals or at the Online Mendehan Inhe ⁇ tance m Man (OMIM) World Wide Web site Correlation between the location of the gene encoding SYNT 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
  • SYNT in another embodiment, SYNT, its catalytic or immunogenic fragments, or ohgopeptides thereof can be used for screening hbranes of compounds in any of a vanety 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 mtracellularly
  • the formation of binding complexes between SYNT 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 WO84/03564 )
  • This method large numbers of different small test compounds are synthesized on a solid substrate
  • the test compounds are reacted with SYNT, or fragments thereof, and washed Bound SYNT is then detected by methods well known in the art Punfied SYNT can also be coated directly onto plates for use in the aforementioned drug screening techniques
  • non-neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support
  • the nucleotide sequences which encode SYNT may be used in any molecular
  • RNA was purchased from Clontech or isolated from tissues described in Table 4 SoVne tissues were homogenized and lysed in guamdinium 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 centnfuged over CsCl 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 hbranes, poly(A+) RNA was isolated using ohgo d(T)-coupled paramagnetic particles (Promega), OLIGOTEX latex particles (QIAGEN, Chatsworth CA), or an OLIGOTEX mRNA
  • RNA was provided with RNA and constructed the corresponding cDNA hbranes
  • cDNA was synthesized and cDNA hbranes 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 transc ⁇ ption was initiated using ohgo d(T) or random p ⁇ mers
  • Synthetic ohgonucleotide adapters were ligated to double stranded cDNA, and the cDNA was digested with the appropnate restriction enzyme or enzymes. For most hbranes.
  • cDNA was size-selected (300- 1000 bp) using SEPHACRYL S 1000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (Amersham Pharmacia Biotech) or preparative agarose gel electrophoresis.
  • cDNAs were ligated into compatible rest ⁇ ction enzyme sites of the polyhnker of a suitable plasmid, e.g., PBLUESCRIPT plasmid (Stratagene), PSPORTl plasmid (Life Technologies), pcDNA2.1 plasmid (Invitrogen, Carlsbad CA), or pINCY plasmid (Incyte Genomics, Palo Alto CA)
  • a suitable plasmid e.g., PBLUESCRIPT plasmid (Stratagene), PSPORTl plasmid (Life Technologies), pcDNA2.1 plasmid (Invitrogen, Carlsbad CA), or pINCY plasmid (Incyte Genomics,
  • Plasmids obtained as desc ⁇ bed in Example I were recovered from host cells by in vivo excision using the UNIZAP vector system (Stratagene) or by cell lysis. Plasmids were punfied using at least one of the following: a Magic or WIZARD Minipreps DNA purification system (Promega); an AGTC Mi prep punfication kit (Edge Biosystems, Gaithersburg MD); and QIAWELL 8 Plasmid, QIAWELL 8 Plus Plasmid, QIAWELL 8 Ultra Plasmid punfication systems or the R E.A.L. PREP 96 plasmid punfication kit from QIAGEN. Following precipitation, plasmids were resuspended in 0.1 ml of distilled water and stored, with or without lyophihzation, at 4°C.
  • 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 fluoromet ⁇ cally using PICOGREEN dye (Molecular Probes, Eugene OR) and a FLUOROSKAN II fluorescence scanner (Labsystems Oy, Helsinki, Finland)
  • Incyte cDNA recovered in plasmids as descnbed in Example II were sequenced as follows Sequencing reactions were processed using standard methods or high-throughput instrumentation such as the ABI CATALYST 800 (PE 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 (PE Biosystems).
  • Electrophoretic separation of cDNA sequencing reactions and detection of labeled polynucleotides were earned out using the MEGABACE 1000 DNA sequencing system (Molecular Dynamics); the ABI PRISM 373 or 377 sequencing system (PE 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 VI
  • the polynucleotide sequences derived from cDNA sequencing were assembled and analyzed using a combination of software programs which utilize algonthms well known to those skilled in the art Table 5 summa ⁇ zes the tools, programs, and algonthms used and provides applicable descriptions, references, and threshold parameters
  • Table 5 summa ⁇ zes the tools, programs, and algonthms used and provides applicable descriptions, references, and threshold parameters
  • the first column of Table 5 shows the tools, programs, and algonthms used
  • the second column provides bnef descriptions thereof
  • the third column presents appropnate references, all of which are incorporated by reference herein in their entirety
  • 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, the greater the homology between two sequences)
  • Sequences were analyzed using MACDNASIS PRO software (Hitachi Software Engineenng, South San Francisco CA) and LASERGENE software (DNASTAR)
  • the polynucleotide sequences were validated by removing vector, linker, and polyA sequences and by masking ambiguous bases, using algorithms and programs based on BLAST, dynamic programing, and dinucleotide nearest neighbor analysis
  • the sequences were then que ⁇ ed against a selection of public databases such as the GenBank pnmate, rodent, mammalian, vertebrate, and eukaryote databases, and BLOCKS, PRINTS, DOMO, PRODOM, and PFAM to acquire annotation using programs based on BLAST, FASTA, and BLIMPS
  • the sequences were assembled into full length polynucleotide sequences using programs based on Phred, Phrap, and Consed, and were screened for open reading frames using programs based on GeneMark, BLAST, and FASTA
  • the full length polynucleotide sequences were translated to de ⁇ ve the corresponding full length ammo acid sequences, and these full length sequences were subsequently analyzed by query
  • Northern analysis is a laboratory technique used to detect the presence of a transcnpt of a gene and involves the hybndization 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, supra, ch 7, Ausubel, 1995, supra, ch. 4 and 16.)
  • the product score takes into account both the degree of similanty between two sequences and the 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 score of +5 for every base that matches in a high-sco ⁇ ng 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
  • the results of northern analyses are reported as a percentage distnbution of hbranes in which the transcnpt encoding SYNT occurred.
  • Analysis involved the catego ⁇ zation of cDNA hbranes by organ/tissue and disease.
  • the organ/tissue categones included cardiovascular, dermatologic, developmental, endoc ⁇ ne, gastrointestinal, hematopoietic/immune, musculoskeletal, nervous, reproductive, and urologic.
  • the disease/condition categones included cancer, inflammation, trauma, cell proliferation, neurological, and pooled.
  • the full length nucleic acid sequences of SEQ ID NO: 16-30 were produced by extension of an appropnate fragment of the full length molecule using ohgonucleotide pnmers designed from this fragment.
  • One pnmer was synthesized to initiate 5' extension of the known fragment, and the other primer, to initiate 3' extension of the known fragment
  • the initial pnmers 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 p ⁇ mer-pnmer dime ⁇ zations was avoided
  • 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 p ⁇ mer, reaction buffer containing Mg 2 *, (NH 4 ) 2 S0 4 , and ⁇ -mercaptoethanol, Taq DNA polymerase (Amersham Pharmacia Biotech), ELONGASE enzyme (Life Technologies), and Pfu DNA polymerase (Stratagene), with the following parameters for p ⁇ mer pair PCI A and PCI B: Step 1. 94°C, 3 mm; Step 2: 94°C, 15 sec; Step 3. 60°C, 1 mm; Step 4.
  • Step 7 storage at 4°C
  • the parameters for pnmer pair T7 and SK+ were as follows: Step 1 94°C, 3 mm, Step 2 94°C, 15 sec, Step 3 57 °C, 1 min; Step 4: 68°C, 2 mm; 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% (v/v) PICOGREEN; Molecular Probes, Eugene OR) dissolved in IX TE and 0.5 ⁇ l of undiluted PCR product into each well of an opaque fluonmeter 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 ⁇ l to 10 ⁇ l aliquot of the reaction mixture was analyzed by electrophoresis on a 1 % agarose mini-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 pnor to rehgation into pUC 18 vector (Amersham Pharmacia Biotech).
  • CviJI cholera virus endonuclease Molecular Biology Research, Madison WI
  • sonicated or sheared pnor to rehgation into pUC 18 vector Amersham Pharmacia Biotech.
  • 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 rehgated using T4 hgase (New England Biolabs, Beverly MA) into pUC 18 vector (Amersham Pharmacia Biotech), treated with Pfu DNA polymerase (Stratagene) to fill-in rest ⁇ ction 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 LB/2x carb liquid media
  • Step 1 94°C, 3 m ; Step 2: 94°C 15 sec; Step 3 60°C, 1 min; Step 4. 72°C, 2 m , Step 5- steps 2, 3, and 4 repeated 29 times; Step 6- 72°C, 5 mm; Step 7- storage at 4°C DNA was quantified by PICOGREEN reagent (Molecular Probes) as described above Samples with low DNA recovenes were reamp fied using the same conditions as described above.
  • polynucleotide sequences of SEQ ID NO: 16-30 are used to obtain 5' regulatory sequences using the procedure above, along with ohgonucleotides designed for such extension, and an appropnate genomic library.
  • the cDNA sequences which were used to assemble SEQ ID NO: 16-30 were compared with sequences from the Incyte LIFESEQ database and public domain databases using BLAST and other implementations of the Smith- Waterman algonthm. Sequences from these databases that matched SEQ ID NO: 16-30 were assembled into clusters of contiguous and overlapping sequences using assembly algonthms such as Phrap (Table 5) 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 cluster resulted in the assignment of all sequences of that cluster, including its particular SEQ ID NO-, to that map location.
  • assembly algonthms such as Phrap (Table 5)
  • Radiation hybrid and genetic mapping data available from public resources such as the Stanford Human Genome Center (SHGC), Whitehead Institute for Genome Research (WIGR),
  • SEQ ID NO.16, SEQ ID NO: 17, SEQ ID NO- 18, SEQ ID NO:21, SEQ ID NO.24, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO-29, and SEQ ID NO'30 are desc ⁇ bed in The Invention as ranges, or intervals, of human chromosomes More than one map location is reported for SEQ ID NO.28 and SEQ ID NO:29, indicating that previously mapped sequences having similanty, but not complete identity, to SEQ ID N0 28 and SEQ ID NO:29 were assembled into their respective clusters.
  • centiM centiMorgan
  • centiM centiMorgan
  • Mb megabase
  • the cM distances are based on genetic markers mapped by Genethon which provide boundanes for radiation hybnd markers whose sequences were included in each of the clusters Diseases associated with the public and Incyte sequences located within the indicated intervals are also reported in the Invention where applicable.
  • Hybndization probes de ⁇ ved from SEQ ID NO 16-30 are employed to screen cDNAs, genomic DNAs. or mRNAs
  • ohgonucleotides consisting of about 20 base pairs
  • OLIGO 4 06 software National Biosciences
  • the labeled ohgonucleotides are substantially purified using a SEPHADEX G-25 superfine size exclusion dextran bead column (Amersham Pharmacia Biotech) An aliquot containing 10 7 counts per minute of the labeled probe is used
  • the linkage or synthesis of array elements upon a microarray can be achieved utilizing photolithography, piezoelectnc pnnting (ink-jet printing, See, e g , Baldeschweiler, supra), mechanical microspotting technologies, and denvatives 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
  • 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 appropnate number of elements (See, e g , Schena, M et al ( 1995) Science 270 467-470, Shalon, D et al ( 1996) Genome
  • ESTs Expressed Sequence Tags
  • fragments or ohgomers thereof may compnse the elements of the microarray Fragments or ohgomers suitable for hybndization can be selected using software well known in the art such as LASERGENE software (DNASTAR)
  • the array elements are hybndized 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
  • nonhybndized 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 hybndization
  • the degree of complementanty and the relative abundance of each polynucleotide which hybndizes to an element on the microarray may be assessed
  • microarray preparation and usage is descnbed in detail below Tissue or Cell Sample
  • RNA is isolated from tissue samples using the guamdinium thiocyanate method and poly(A) + RNA is punfied using the ohgo-(dT) cellulose method
  • Each poly(A) + RNA sample is reverse transc ⁇ bed using MMLV reverse-transc ⁇ ptase, 0 05 pg/ ⁇ l ohgo-(dT) primer (21mer), IX first strand buffer, 0 03 units/ ⁇ l RNase inhibitor, 500 ⁇ M dATP, 500 ⁇ M dGTP, 500 ⁇ M dTTP, 40 ⁇ M dCTP, 40 ⁇ M dCTP-Cy3 (BDS) or dCTP-Cy5 (Amersham Pharmacia Biotech)
  • the reverse transcnption 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 trans
  • Sequences of the present invention are used to generate array elements Each array element is amplified from bactenal cells containing vectors with cloned cDNA inserts PCR amplification uses p ⁇ mers 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 punfied using SEPHACRYL-400 (Amersham Pharmacia Biotech) Punfied 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% ammopropyl silane (Sigma) in 95% ethanol Coated slides are cured in a 1 10°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 ⁇ l of the array element DNA, at an average concentration of 100 ng/ ⁇ l, is loaded into the open capillary printing element by a high-speed robotic apparatus The apparatus then deposits about 5 nl of array element sample per slide
  • Microarrays are UV-crosslinked using a STRATALINKER UV-crosshnker (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
  • 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 Cy5
  • 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
  • a mixed gas multiline laser excites the two fluorophores sequentially Emitted light is split, based on wavelength, into two photomultiplier tube detectors (PMT R1477, Hamamatsu Photonics Systems, B ⁇ dgewater NJ) corresponding to the two fluorophores
  • Appropnate 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 Cy5
  • Each array is typically scanned twice, one scan per fluorophore using the appropnate 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 hybndizing species of 1 100,000
  • 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
  • A/D analog-to-digital
  • 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 g ⁇ d is supe ⁇ mposed 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 numencal value corresponding to the average intensity of the signal
  • the software used for signal analysis is the GEMTOOLS gene expression analysis program (Incyte)
  • Sequences complementary to the SYNT-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occumng SYNT
  • ohgonucleotides compnsing from about 15 to 30 base pairs is descnbed, essentially the same procedure is used with smaller or with larger sequence fragments
  • Appropnate ohgonucleotides are designed using OLIGO 4 06 software (National Biosciences) and the coding sequence of SYNT
  • a complementary o gonucleotide is designed from the most unique 5' sequence and used to prevent promoter binding to the coding sequence
  • a complementary ohgonucleotide is designed to prevent ⁇ bosomal binding to the SYNT-encod g transcnpt
  • SYNT expression and punfication of SYNT is achieved using bacterial or virus-based expression systems
  • cDNA is subcloned into an appropnate vector containing an antibiotic resistance gene and an inducible promoter that directs high levels of cDNA transcnption
  • promoters include, but are not limited to, the trp-lac (tac) hyb ⁇ d promoter and the T5 or T7 bactenophage promoter in conjunction with the lac operator regulatory element
  • Recombinant vectors are transformed into suitable bactenal hosts, e g , BL21(DE3)
  • Antibiotic resistant bacteria express SYNT upon induction with isopropyl beta-D- thiogalactopyranoside (IPTG)
  • IPTG isopropyl beta-D- thiogalactopyranoside
  • the nonessential polyhedrin gene of baculovirus is replaced with cDNA encoding SYNT 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 Spodoptera frugiperda (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.)
  • SYNT 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 iaponicum. 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 SYNT at specifically engineered sites.
  • FLAG an 8-amino acid peptide
  • 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, supra, ch. 10 and 16). Purified SYNT obtained by these methods can be used directly in the assays shown in Examples X and XIV.
  • An SYNT activity assay measures aminoacylation of tRNA in the presence of a radiolabeled substrate.
  • a cell-free extract depleted of endogenous aminoacyl-tRNA synthetase is prepared from Escherichia coli. SYNT, either biochemically purified or recombinantly produced, is added to the cell free extract. The cell-free extract is incubated with [ 14 C]-labeled amino acid under conditions favorable for translation. Incorporation of the [ 14 C]-labeled amino acid into acid-precipitable aminoacyl-tRNA is measured using a radioisotope counter. The amount of the [ 14 C]-labeled amino acid incorporated into aminoacyl tRNA is proportional to the amount of SYNT activity.
  • SYNT activity may be assayed as follows. SYNT, or biologically active fragments thereof, are labeled with i25 I Bolton-Hunter reagent. (See, e.g., Bolton et al. ( 1973) Biochem. J. 133:529.) Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled SYNT, washed, and any wells with labeled SYNT complex are assayed. Data obtained using different concentrations of SYNT are used to calculate values for the number, affinity, and association of SYNT with the candidate molecules.
  • SYNT function is assessed by expressing the sequences encoding SYNT at physiologically elevated levels in mammalian cell culture systems cDNA is subcloned into a mammalian expression vector containing a strong promoter that d ⁇ ves high levels of cDNA expression
  • Vectors of choice include pCMV SPORT plasmid (Life Technologies) and pCR3 1 plasmid (Invitrogen), 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 ⁇ 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 Mark
  • FCM Flow cytometry
  • FCM detects and quantifies the uptake of fluorescent molecules that diagnose events preceding or coincident with cell death
  • events include changes in nuclear DNA content as measured by staining of DNA with propidium iodide, changes in cell size and granula ⁇ ty as measured by forward light scatter and 90 degree side light scatter, down-regulation of DNA synthesis as measured by decrease in bromodeoxyu ⁇ dine 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
  • SYNT The influence of SYNT on gene expression can be assessed using highly purified populations of cells transfected with sequences encoding SYNT 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 SYNT and other genes of interest can be analyzed by northern analysis or microarray techniques XII. Production of SYNT Specific Antibodies
  • the SYNT amino acid sequence is analyzed using LASERGENE software (DNASTAR) to determine regions of high immunogenicity, and a corresponding ohgopeptide is synthesized and used to raise antibodies by means known to those of skill in the art Methods for selection of appropnate 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 1 1 )
  • ohgopeptides of about 15 residues in length are synthesized using an ABI 431 A peptide synthesizer (PE Biosystems) using FMOC chemistry and coupled to KLH (Sigma-Ald ⁇ ch, 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 ohgopeptide- KLH complex in complete Freund's adjuvant Resulting antisera are tested for antipeptide and anti- SYNT activity by, for example, binding the peptide or SYNT to a substrate, blocking with 1 % BSA, reacting with rabbit antisera, washing, and reacting with radio-iodmated goat anti-rabbit IgG XIII. Purification of Naturally Occurring SYNT Using Specific Antibodies
  • Naturally occurring or recombinant SYNT is substantially purified by immunoaffinity chromatography using antibodies specific for SYNT
  • An immunoaffinity column is constructed by covalently coupling anti-SYNT 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 SYNT are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of SYNT (e g , high ionic strength buffers in the presence of detergent)
  • the column is eluted under conditions that disrupt antibody/SYNT binding (e g , a buffer of pH 2 to pH 3, or a high concentration of a chaotrope, such as urea or thiocyanate ion), and SYNT is collected
  • SYNT or biologically active fragments thereof, are labeled with I25 I Bolton-Hunter reagent (See, e g , Bolton A E and W M Hunter (1973) Biochem J 133 529-539 )
  • I25 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 SYNT, washed, and any wells with labeled SYNT complex are assayed
  • Data obtained using different concentrations of SYNT are used to calculate values for the number, affinity, and association of SYNT with the candidate molecules
  • molecules interacting with SYNT are analyzed using the yeast two-hybnd system as desc ⁇ bed in Fields, S and O Song (1989, Nature 340 245-246), or using commercially available kits based on the two-hybnd system, such as the MATCHMAKER system (Clontech)
  • SYNT may also be used in the PATHCALLING process (CuraGen Corp , New Haven CT) which employs the yeast two-hybnd system in a high-throughput manner to determine all interactions between the proteins encoded by two large hbranes of genes (Nandabalan, K et al (2000) U S Patent No 6,057, 101 )
  • ABI FACTURA A program that removes vector sequences and PE Biosystems, Foster City, CA. masks ambiguous bases in nucleic acid sequences.
  • ABI/PARACEL FDF A Fast Data Finder useful in comparing and PE Biosystems, Foster City, CA; Mismatch ⁇ 50% annotating amino acid or nucleic acid sequences. Paracel Inc., Pasadena, CA.
  • ABI AutoAssembler A program that assembles nucleic acid sequences. PE Biosystems, Foster City, CA.
  • fastx score 100 or greater
  • Phred A base-calling algorithm that examines automated Ewing, B. et al. (1998) Genome Res. sequencer traces with high sensitivity and 8: 175-185; Ewing, B. and P. Green probability. (1998) Genome Res. 8: 186-194.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pulmonology (AREA)
  • Reproductive Health (AREA)
  • Endocrinology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Urology & Nephrology (AREA)
  • Diabetes (AREA)
  • Gynecology & Obstetrics (AREA)
  • Dermatology (AREA)
  • Communicable Diseases (AREA)
  • Psychiatry (AREA)
  • Pregnancy & Childbirth (AREA)
  • Psychology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Vascular Medicine (AREA)

Abstract

The invention provides human synthetases (SYNT) and polynucleotides which identify and encode SYNT. 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 expression of SYNT.

Description

HUMAN SYNTHETASES
TECHNICAL FIELD
This invention relates to nucleic acid and ammo acid sequences of synthetases and to the use of these sequences in the diagnosis, treatment, and prevention of immune, neuronal, and reproductive disorders, and cell pro ferative disorders including cancer
BACKGROUND OF THE INVENTION
A large number of cellular biosynthetic intermediary metabolism processes involve lntermolecular transfer of carbon atom-containing substrates (carbon substrates) Examples of such reactions include the tncarboxyhc acid cycle, synthesis of fatty acids and long-chain phospholipids, synthesis of alcohols and aldehydes, synthesis of intermediary metabolites, and reactions involved in the amino acid degradation pathways Many of these reactions are catalyzed by synthetases (also called hgases), which catalyze the formation of a bond between two substrate molecules Some of these reactions require input of energy, usually in the form of conversion of ATP to either ADP or AMP and pyrophosphate Synthetases are named for the products of the reaction they catalyze and are involved in such processes as metabolism and the synthesis of macromolecules Ligases forming carbon-oxygen bonds
Proteins make up more than half of the total dry mass of a cell The synthesis of proteins is central to cell maintenance, growth, and development Synthesis occurs on πbosomes and depends on the cooperative interaction of several classes of RNA molecules The process begins with transcription of the genetic code contained within the DNA to form messenger RNA (mRNA) The mRNA moves in steps through a πbosome and the nucleotide sequence of the mRNA is translated into a corresponding sequence of amino acids to construct a distinct protein chain
The am oacyl-transfer RNA (tRNA) synthetases are important RNA-associated enzymes with roles in translation Protein biosynthesis depends on each amino acid forming a linkage with the appropnate tRNA The aminoacyl-tRNA synthetases are responsible for the activation and correct attachment of an amino acid with its cognate tRNA The 20 aminoacyl-tRNA synthetase enzymes can be divided into two structural classes, and each class is charactenzed by a distinctive topology of the catalytic domain Class I enzymes contain a catalytic domain based on the nucleotide-binding Rossman 'fold' Class II enzymes contain a central catalytic domain, which consists of a seven-stranded antiparallel β-sheet motif, as well as N- and C- terminal regulatory domains Class II enzymes are separated into two groups based on the heterodimenc or homodimenc structure of the enzyme, the latter group is further subdivided by the structure of the N- and C-terminal regulatory domains (Hartlein, M and Cusack, S ( 1995) J Mol Evol 40 519-530) Autoantibodies against aminoacyl-tRNAs are generated by patients with dermatomyositis and polymyositis, and correlate strongly with complicating interstitial lung disease (ILD) These antibodies appear to be generated in response to viral infection, and coxsackie virus has been used to induce expeπmental viral myositis m animals (Fπedman, A W et al (1996) Semin Arthritis Rheum 26 459-467) A synthetase homolog has been shown to be expressed in chronic myeloid leukemia (CML) A phenylalanine-tRNA synthetase homolog has been found to be tumor-selective and expressed in a cell cycle stage- and differentiation-dependent fashion in an acute-phase human CML cell line (Sen, S et al ( 1997) Proc Natl Acad Sci USA 94 6164-6169) Ligases forming carbon-sulfur bonds (Acid-thiol ligases)
In many cases, a carbon substrate is denved from a small molecule containing at least two carbon atoms The carbon substrate is often covalently bound to a larger molecule which acts as a carbon substrate earner molecule within the cell In the biosynthetic mechanisms descnbed above, the earner molecule is coenzyme A Coenzyme A (CoA) is structurally related to denvatives of the nucleotide ADP and consists of 4'-ρhosphopantetheιne linked via a phosphodiester bond to the alpha phosphate group of adenosine 3',5'-bιsphosphate The terminal thiol group of 4'-phosphopantetheme acts as the site for carbon substrate bond formation The predominant carbon substrates which utilize CoA as a earner molecule dunng biosynthesis and intermediary metabolism in the cell are acetyl, succinyl, and propionyl moieties, collectively referred to as acyl groups Other carbon substrates include enoyl lipid, which acts as a fatty acid oxidation intermediate, and carnitine, which acts as an acetyl-CoA flux regulator/ mitochondnal acyl group transfer protein Acyl-CoA and acetyl-CoA are synthesized in the cell by acyl-CoA synthetase and acetyl-CoA synthetase, respectively
Activation of fatty acids is mediated by at least three forms of acyl-CoA synthetase activity 0 acetyl-CoA synthetase, which activates acetate and several other low molecular weight carboxylic acids and is found in muscle mitochondna and the cytosol of other tissues, n) medium-chain acyl-CoA synthetase, which activates fatty acids containing between four and eleven carbon atoms (predominantly from dietary sources), and is present only in liver mitochondna, and in) acyl CoA synthetase, which is specific for long chain fatty acids with between six and twenty carbon atoms, and is found m microsomes and the mitochondna Proteins associated with acyl-CoA synthetase activity have been identified from many sources including bactena, yeast, plants, mouse, and man The activity of acyl-CoA synthetase may be modulated by phosphorylation of the enzyme by cAMP-dependent protein kinase The COL4A5 (collagen, type IV, alpha-5) chromosomal region found deleted in 2 patients with Alport syndrome, el ptocytosis, and mental retardation (Piccini, M et al (1998) Genomics 47 350-358) is contiguous with the region containing long-chain acyl-CoA synthetase 4 (FACL4) Therefore, it has been suggested (Piccini supra) that the absence of FACL4 may be involved in the development of mental retardation and other phenotypes associated with Alport syndrome in these patients Ligases forming carbon-nitrogen bonds
A key representative of the amide synthases is the enzyme glutamine synthetase (glutamate- ammonia hgase) that catalyzes the animation of glutamic acid to glutamme by ammonia using the energy of ATP hydrolysis Glutamine is the primary source for the amino group in vaπous amide transfer reactions involved in de novo pyrimidine nucleotide synthesis and in puπne and pyπmidine πbonucleotide interconversions, as well as the conversion of aspartate to asparagine Overexpression of glutamine synthetase has been observed in pπmary liver cancer (Chnsta, L et al ( 1994) Gastroent 106 1312- 1320) Cyclo-hgases and other carbon-nitrogen ligases compnse vaπous enzymes and enzyme complexes that participate in the de novo pathways to puπne and pyπmidine biosynthesis Because these pathways are cπtical to the synthesis of nucleotides for replication of both RNA and DNA, many of these enzymes have been the targets of clinical agents for the treatment of cell prohferative disorders such as cancer and infectious diseases Puπne biosynthesis occurs de novo from the amino acids glycine and glutamine, and other small molecules Three of the key reactions in this process are catalyzed by a tπfunctional enzyme composed of glycinamide-πbonucleotide synthetase (GARS), aminoimidazole πbonucleotide synthetase (AIRS), and glycinamide πbonucleotide transformylase (GART) Together these three enzymes combine πbosylamine phosphate with glycine to yield phosphoπbosyl aminoimidazole, a precursor to both adenylate and guanylate nucleotides This tπfunctional protein has been implicated in the pathology of Downs syndrome (Ainu, J et al ( 1990) Nucleic Acid Res 18 6665-6672) Adenylosuccinate synthetase catalyzes a later step in purine biosynthesis that converts inosinic acid to adenylosuccinate, a key step on the path to ATP synthesis This enzyme is also similar to another carbon-nitrogen gase, argininosuccinate synthetase, that catalyzes a similar reaction the urea cycle (Powell, S M et al (1992) FEBS Lett 303 4-10)
Like the de novo biosynthesis of puπnes, de novo synthesis of the pyπmidine nucleotides uπdylate and cytidylate also aπses from a common precursor, in this instance the nucleotide orotidylate deπved from orotate and phosphoπbosyl pyrophosphate (PPRP) Again a tπfunctional enzyme compnsing three carbon-nitrogen ligases plays a key role in the process In this case the enzymes aspartate transcarbamylase (ATCase), carbamyl phosphate synthetase II, and dihydroorotase (DHOase) are encoded by a single gene called CAD Together these three enzymes combine the initial reactants in pyπmidine biosynthesis, glutamine, CO, and ATP to form dihydroorotate, the precursor to orotate and orotidylate (Iwahana, H et al ( 1996) Biochem Biophys Res Commun 219 249-255) Further steps then lead to the synthesis of undine nucleotides from orotidylate Cytidine nucleotides are denved from uπdιne-5'-tπphosphate (UTP) by the amidation of UTP using glutamine as the amino donor and the enzyme CTP synthetase Regulatory mutations in the human CTP synthetase are believed to confer multi-drug resistance to agents widely used in cancer therapy (Yamauchi, M et al. ( 1990) EMBO J. 9:2095-2099) Ligases forming carbon-carbon bonds
Ligases in this group are represented by the carboxylases acetyl-CoA carboxylase and pyruvate carboxylase Acetyl-CoA carboxylase is a complex which includes a biotin carboxyl earner protein, biotm carboxylase, and a carboxyl transferase made up of two alpha and two beta subunits. This complex catalyzes the carboxylation of Acetyl-CoA from CO, and t O using the energy of ATP hydrolysis (PRINTS document PRO 1069). Acetyl-CoA carboxylase is the rate-limiting step in the biogenesis of long-chain fatty acids Two isoforms of Acetyl-CoA carboxylase, types I and types II, are expressed in humans in a tissue-specific manner (Ha, J. et al. (1994) Eur. J. Biochem. 219:297- 306). Pyruvate carboxylase is a nuclear-encoded mitochondπal enzyme that catalyzes the conversion of pyruvate to oxaloacetate, a key intermediate in the citπc acid cycle. Ligases forming phosphoπc ester bonds
Ligases in this group are represented by the DNA ligases involved in both DNA replication and repair DNA ligases seal phosphodiester bonds between two adjacent nucleotides in a DNA chain using the energy from ATP hydrolysis to first activate the free 5'-phosphate of one nucleotide and then react it with the 3'-OH group of the adjacent nucleotide. This reseahng reaction is used in both DNA replication to join small DNA fragments called "Okazaki" fragments that are transiently formed in the process of replicating new DNA, and in DNA repair. DNA repair is the process by which accidental base changes, such as those produced by oxidative damage, hydrolytic attack, or uncontrolled methylation of DNA, are corrected before replication or transcπption of the DNA can occur. Bloom's syndrome is an inheπted human disease in which individuals are partially deficient in DNA hgation and consequently have an increased incidence of cancer (Alberts, B et al. ( 1994) The Molecular Biology of the Cell, Garland Publishing Inc., New York, NY, p. 247) The discovery of new synthetases and the polynucleotides encoding them satisfies a need in the art by providing new compositions which are useful m the diagnosis, prevention, and treatment of immune, neuronal, and reproductive disorders, and cell prohferative disorders including cancer.
SUMMARY OF THE INVENTION The invention features punfied polypeptides, human synthetases, referred to collectively as
"SYNT" and individually as "SYNT-1 ," "SYNT-2," "SYNT-3," "SYNT-4,""SYNT-5," "SYNT-6," "SYNT-7," "SYNT-8," "SYNT-9,""SYNT-10," "SYNT-1 1 ," "SYNT- 12," "SYNT- 13," "SYNT- 14," and "SYNT- 15 " In one aspect, the invention provides an isolated polypepttde compnsing an amino acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, b) a naturally occumng amino acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO.1-15, and d) an immunogenic fragment of an ammo acid sequence selected from the group consisting of SEQ ID NO: 1-15. In one alternative, the invention provides an isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 1-15. The invention further provides an isolated polynucleotide encoding a polypeptide compnsing an amino acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, b) a naturally occumng amino acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, and d) an immunogenic fragment of an am o acid sequence selected from the group consisting of SEQ ID NO: 1-15. In one alternative, the polynucleotide encodes a polypeptide selected from the group consisting of SEQ ID NO- 1-15 In another alternative, the polynucleotide is selected from the group consisting of SEQ ID NOT 6-30.
Additionally, the invention provides a recombinant polynucleotide compnsing a promoter sequence operably linked to a polynucleotide encoding a polypeptide compnsing an amino acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO: l-15, b) a naturally occumng amino acid sequence having at least 90% sequence identity to an ammo acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO: 1- 15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15. In one alternative, the invention provides a cell transformed with the recombinant polynucleotide. In another alternative, the invention provides a transgenic organism compnsing the recombinant polynucleotide
The invention also provides a method for producing a polypeptide compnsing an am o acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15, b) a naturally occurring amino acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOT-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO- 1-15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO: 1-15. The method compnses a) cultuπng a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide compnsing a promoter sequence operably linked to a polynucleotide encoding the polypeptide, and b) recoveπng the polypeptide so expressed.
Additionally, the invention provides an isolated antibody which specifically binds to a polypeptide compnsing an amino acid sequence selected from the group consisting of a) an ammo acid sequence selected from the group consisting of SEQ ID NO: 1-15, b) a naturally occumng ammo acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, and d) an immunogenic fragment of an ammo acid sequence selected from the group consisting of SEQ ID NO 1-15 The invention further provides an isolated polynucleotide compnsing a polynucleotide sequence selected from the group consisting of a) a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, b) a naturally occu ng polynucleotide sequence having at least 90% sequence identity to a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, c) a polynucleotide sequence complementary to a), d) a polynucleotide sequence complementary to 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 comprising a polynucleotide sequence selected from the group consisting of a) a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, b) a naturally occumng polynucleotide sequence having at least 90% sequence identity to a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, c) a polynucleotide sequence complementary to a), d) a polynucleotide sequence complementary to b), and e) an RNA equivalent of a)-d) The method comprises a) hybndizing the sample with a probe compnsing at least 20 contiguous nucleotides compnsing a sequence complementary to said target polynucleotide in the sample, and which probe specifically hybndizes to said target polynucleotide, under conditions whereby a hybndization complex is formed between said probe and said target polynucleotide or fragments thereof, and b) detecting the presence or absence of said hybndization complex, and optionally, if present, the amount thereof In one alternative, the probe compnses 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 compnsing a polynucleotide sequence selected from the group consisting of a) a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, b) a naturally occumng polynucleotide sequence having at least 90% sequence identity to a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, c) a polynucleotide sequence complementary to a), d) a polynucleotide sequence complementary to b), and e) an RNA equivalent of a)-d) The method compnses 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 pharmaceutical composition compnsing an effective amount of a polypeptide compnsing an amino acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, b) a naturally occumng am o acid sequence having at least 90% sequence identity to an ammo acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, and a pharmaceutically acceptable excipient In one embodiment, the pharmaceutical composition compnses an amino acid sequence selected from the group consisting of SEQ ID NO 1-15 The invention additionally provides a method of treating a disease or condition associated with decreased expression of functional SYNT, compnsing administering to a patient in need of such treatment the pharmaceutical composition
The invention also provides a method for screening a compound for effectiveness as an agonist of a polypeptide compnsing an amino acid sequence selected from the group consisting of a) an ammo acid sequence selected from the group consisting of SEQ ID NO 1-15, b) a naturally occurring amino acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15 The method compnses a) exposing a sample compnsing the polypeptide to a compound, and b) detecting agonist activity in the sample In one alternative, the invention provides a pharmaceutical 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 SYNT, compnsing admimstenng to a patient in need of such treatment the pharmaceutical composition
Additionally, the invention provides a method for screening a compound for effectiveness as an antagonist of a polypeptide compnsing an ammo acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, b) a naturally occumng ammo acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15 The method compnses a) exposing a sample compnsing the polypeptide to a compound, and b) detecting antagonist activity in the sample In one alternative, the invention provides a pharmaceutical composition compnsing 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 SYNT, compnsing admimstenng to a patient in need of such treatment the pharmaceutical composition The invention further provides a method of screening for a compound that specifically binds to a polypeptide compnsing an amino acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, b) a naturally occumng ammo acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1-15 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 comprising an ammo acid sequence selected from the group consisting of a) an amino acid sequence selected from the group consisting of SEQ ID NO 1- 15, b) a naturally occumng amino acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO 1-15, c) a biologically active fragment of an ammo acid sequence selected from the group consisting of SEQ ID NO 1- 15, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO 1- 15 The method compnses 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) companng 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 altenng expression of a target polynucleotide, wherein said target polynucleotide compnses a sequence selected from the group consisting of SEQ ID NO 16-30, the method compnsing a) exposing a sample compnsing 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 compnsing a) treating a biological sample containing nucleic acids with the test compound, b) hybndizing the nucleic acids of the treated biological sample with a probe compnsing at least 20 contiguous nucleotides of a polynucleotide compnsing a polynucleotide sequence selected from the group consisting of I) a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, n) a naturally occumng polynucleotide sequence having at least 90% sequence identity to a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, in) a polynucleotide sequence complementary to I), iv) a polynucleotide sequence complementary to n), and v) an RNA equivalent of ι)-ιv) Hybndization occurs under conditions whereby a specific hybndization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide compnsing a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, n) a naturally occumng polynucleotide sequence having at least 90% sequence identity to a polynucleotide sequence selected from the group consisting of SEQ ID NO 16-30, in) a polynucleotide sequence complementary to l), iv) a polynucleotide sequence complementary to n), and v) an RNA equivalent of ι)-ιv) Alternatively, the target polynucleotide compnses a fragment of the above polynucleotide sequence, c) quantifying the amount of hybndization complex, and d) companng the amount of hybndization complex in the treated biological sample with the amount of hybndization complex in an untreated biological sample, wherein a difference in the amount of hybndization complex in the treated biological sample is indicative of toxicity of the test compound
BRIEF DESCRIPTION OF THE TABLES Table 1 shows polypeptide and nucleotide sequence identification numbers (SEQ ID NOs), clone identification numbers (clone IDs), cDNA hbranes, and cDNA fragments used to assemble full- length sequences encoding SYNT
Table 2 shows features of each polypeptide sequence, including potential motifs, homologous sequences, and methods, algorithms, and searchable databases used for analysis of SYNT Table 3 shows selected fragments of each nucleic acid sequence, the tissue-specific expression patterns of each nucleic acid sequence as determined by northern analysis, diseases, disorders, or conditions associated with these tissues, and the vector into which each cDNA was cloned
Table 4 describes the tissues used to construct the cDNA hbranes from which cDNA clones encoding SYNT were isolated
Table 5 shows the tools, programs, and algonthms 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, matenals and methods descπbed, as these may vary It is also to be understood that the terminology used herein is for the purpose of descnbing 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 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, matenals, and methods similar or equivalent to those descnbed herein can be used to practice or test the present invention, the preferred machines, matenals and methods are now descnbed All publications mentioned herein are cited for the purpose of descnbing and disclosing the cell lines, protocols, reagents and vectors which are reported in the publications and which might be used m 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
"SYNT" refers to the ammo acid sequences of substantially puπfied SYNT obtained from any species, particularly a mammalian species, including bovine, ovine, porcine, munne, 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 SYNT Agonists may include proteins, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of SYNT either by directly interacting with SYNT or by acting on components of the biological pathway in which SYNT participates
An "allehc vaπant" is an alternative form of the gene encoding SYNT Allehc vaπants 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 allehc vanants of its naturally occumng form Common mutational changes which give nse to allehc vaπants 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 SYNT include those sequences with deletions, insertions, or substitutions of different nucleotides, resulting in a polypeptide the same as SYNT or a polypeptide with at least one functional characteπstic of SYNT Included within this definition are polymorphisms which may or may not be readily detectable using a particular oligonucleotide probe of the polynucleotide encoding SYNT, and improper or unexpected hybndization to allehc vanants, with a locus other than the normal chromosomal locus for the polynucleotide sequence encoding SYNT 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 SYNT Deliberate amino acid substitutions may be made on the basis of similaπty in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues, as long as the biological or immunological activity of SYNT is retained For example, negatively charged amino acids may include aspartic acid and glutamic acid, and positively charged am o acids may include lysine and argimne. Amino acids with uncharged polar side chains having similar hydrophilicity values may include: asparagme and glutamine; and senne 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 "ammo acid" and "amino acid sequence" refer to an ohgopeptide, peptide, polypeptide, or protein sequence, or a fragment of any of these, and to naturally occumng or synthetic molecules. Where "amino acid sequence" is recited to refer to a sequence of a naturally occumng protein molecule, "ammo 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 earned 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 SYNT Antagonists may include proteins such as antibodies, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of SYNT either by directly interacting with SYNT or by acting on components of the biological pathway in which SYNT 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 SYNT polypeptides can be prepared using intact polypeptides or using fragments containing small peptides of interest as the immunizing antigen. The polypeptide or ohgopeptide 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 earner protein if desired. Commonly used earners that are chemically coupled to peptides include bovine serum albumin, thyroglobu n, and keyhole limpet hemocyanin (KLH). 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 lmmunogen used to elicit the immune response) for binding to an antibody
The term "antisense" refers to any composition capable of base-paiπng with the "sense" (coding) strand of a specific nucleic acid sequence. Antisense compositions may include DNA, RNA; peptide nucleic acid (PNA); ohgonucleotides having modified backbone linkages such as phosphorothioates, methylphosphonates, or benzylphosphonates; o gonucleotides having modified sugar groups such as 2 -methoxyethyl sugars or 2'-methoxyethoxy sugars; or ohgonucleotides 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 occumng nucleic acid sequence produced by the cell to form duplexes which block either transcπption 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 occumng molecule. Likewise, "immunologically active" or "immunogenic" refers to the capability of the natural, recombinant, or synthetic SYNT, or of any ohgopeptide thereof, to induce a specific immune response in appropnate animals or cells and to bind with specific antibodies.
"Complementary" descnbes 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 compπse a dry formulation or an aqueous solution. Compositions compnsing polynucleotide sequences encoding SYNT or fragments of SYNT may be employed as hybndization probes. The probes may be stored in freeze-dned form and may be associated with a stabilizing agent such as a carbohydrate In hybπdizations, 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 XL-PCR kit (PE 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 WI) 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 onginal 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 onginal amino acid in a protein and which are regarded as conservative amino acid substitutions Ongmal Residue Conservative Substitution
Ala Gly, Ser
Figure imgf000014_0001
Cys Ala, Ser
Figure imgf000014_0002
He Leu, Val
Leu He, Val Lys Arg, Gin, Glu
Met Leu, He
Phe His, Met, Leu, Trp, Tyr
Ser Cys, Thr
Thr Ser, Val
Figure imgf000014_0003
Conservative ammo 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 ammo acid residues or nucleotides The term "denvative" refers to a chemically modified polynucleotide or polypeptide
Chemical modifications of a polynucleotide sequence can include, for example, replacement of hydrogen by an alkyl, acyl, hydroxyl, or amino group A denvative polynucleotide encodes a polypeptide which retains at least one biological or immunological function of the natural molecule A denvative 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 deπved
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
A "fragment" is a unique portion of SYNT or the polynucleotide encoding SYNT which is identical in sequence to but shorter in length than the parent sequence A fragment may compπse up to the entire length of the defined sequence, minus one nucleotide/amino acid residue For example, a fragment may compπse from 5 to 1000 contiguous nucleotides or amino acid residues A fragment used as a probe, pπmer, 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 compπse 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 NO 16-30 compnses a region of unique polynucleotide sequence that specifically identifies SEQ ID NO 16-30, for example, as distinct from any other sequence in the genome from which the fragment was obtained A fragment of SEQ ID NO 16-30 is useful, for example, in hybndization and amplification technologies and in analogous methods that distinguish SEQ ID NO 16-30 from related polynucleotide sequences The precise length of a fragment of SEQ ID NO 16-30 and the region of SEQ ID NO 16-30 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 1- 15 is encoded by a fragment of SEQ ID NO 16-30 A fragment of SEQ ID NO 1-15 compnses a region of unique amino acid sequence that specifically identifies SEQ ID NO 1-15 For example, a fragment of SEQ ID NO 1- 15 is useful as an immunogenic peptide for the development of antibodies that specifically recognize SEQ ID NO 1-15 The precise length of a fragment of SEQ ID NO 1- 15 and the region of SEQ ID NO 1 - 15 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 sinulanty 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 algonthm Such an algoπthm 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 compaπson of the two sequences
Percent identity between polynucleotide sequences may be determined using the default parameters of the CLUSTAL V algonthm 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 descnbed 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, wιndow=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 similanty" between aligned polynucleotide sequences Alternatively, a suite of commonly used and freely available sequence compaπson algonthms is provided by the National Center for Biotechnology Information (NCBI) 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.mh gov/BLAST/ The BLAST software suite includes vanous sequence analysis programs including "blastn," that is used to align a known polynucleotide sequence with other polynucleotide sequences from a vaπety of databases. Also available is a tool called "BLAST 2 Sequences" that is used for direct pairwise companson 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 2.0.12 (Apπl-21-2000) set at default parameters. Such default parameters may be, for example-
Matrix: BLOSUM62
Reward for match: J Penalty for mismatch: -2
Open Gap 5 and Extension Gap: 2 penalties
Gap x drop-off- 50
Expect 10
Filter: on
Percent identity may be measured over the length of an entire defined 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 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 algonthm. 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 algonthm as incorporated into the MEGALIGN version 3.12e sequence alignment program (descπbed and referenced above). For pairwise alignments of polypeptide sequences using CLUSTAL V, the default parameters are set as follows: Ktuple=l , gap penalty=3, wιndow=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 similanty" between aligned polypeptide sequence pairs.
Alternatively the NCBI BLAST software suite may be used. For example, for a pairwise companson of two polypeptide sequences, one may use the "BLAST 2 Sequences" tool Version 2.0.12 (Apr-21-2000) with blastp set at default parameters. Such default parameters may be, for example.
Matrix. BLOSUM62
Open Gap. 11 and Extension Gap: 1 penalties Gap x drop-off: 50
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 descπbe 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 onginal binding ability "Hybndization" refers to the process by which a polynucleotide strand anneals with a complementary strand through base painng under defined hybndization conditions Specific hybndization is an indication that two nucleic acid sequences share a high degree of complementarity Specific hybndization complexes form under permissive annealing conditions and remain hybridized after the "washing" step(s) The washing step(s) is particularly important in determining the stnngency of the hybndization process with more stnngent conditions allowing less non-specific binding, 1 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 hybndization experiments, whereas wash conditions may be vaned among expenments to achieve the desired stnngency, and therefore hybndization 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 μg/ml sheared, denatured salmon sperm DNA
Generally, stnngency of hybndization is expressed, in part, with reference to the temperature under which the wash step is earned out Such wash temperatures are typically selected to be about 5°C to 20°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH The Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe An equation for calculating Tm and conditions for nucleic acid hybndization are well known and can be found in Sambrook, J et al , 1989, Molecular Cloning A Laboratory Manual, 2nd ed , vol 1-3, Cold Spring Harbor Press, Plainview NY, specifically see volume 2, chapter 9
High stnngency conditions for hybndization 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 hybndization Such blocking reagents include, for instance, sheared and denatured salmon sperm DNA at about 100-200 μg/ml 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 hybπdizations Useful vanations on these wash conditions will be readily apparent to those of ordinary skill in the art Hybndization, particularly under high stnngency conditions, may be suggestive of evolutionary similanty between the nucleotides Such similanty is strongly indicative of a similar role for the nucleotides and their encoded polypeptides
The term "hybndization 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 , C0t or R^ analysis) or formed between one nucleic acid sequence present m solution and another nucleic acid sequence immobilized on a solid support (e g , paper, membranes, filters, chips, pins or glass slides, or any other appropnate 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 vaπous factors, e g., cytokines, chemokines, and other signaling molecules, which may affect cellular and systemic defense systems
An "immunogenic fragment" is a polypeptide or ohgopeptide fragment of SYNT 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 ohgopeptide fragment of SYNT 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 SYNT For example, modulation may cause an increase or a decrease in protein activity, binding characteristics, or any other biological, functional, or immunological properties of SYNT The phrases "nucleic acid" and "nucleic acid sequence" refer to a nucleotide, ohgonucleotide, polynucleotide, or any fragment thereof These phrases also refer to DNA or RNA of genomic or synthetic ongin 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-hke or RNA-hke mateπal
"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 transcnption 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 compnses an ohgonucleotide of at least about 5 nucleotides in length linked to a peptide backbone of ammo acid residues ending m lysine. The terminal lysme confers solubility to the composition PNAs preferentially bind complementary single stranded DNA or RNA and stop transcnpt elongation, and may be pegylated to extend their hfespan in the cell
"Post-translational modification" of an SYNT may involve hpidation, 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 SYNT
"Probe" refers to nucleic acid sequences encoding SYNT, their complements, or fragments thereof, which are used to detect identical, allehc or related nucleic acid sequences Probes are isolated ohgonucleotides or polynucleotides attached to a detectable label or reporter molecule Typical labels include radioactive isotopes, hgands, chemiluminescent agents, and enzymes
"Pπmers" are short nucleic acids, usually DNA ohgonucleotides, which may be annealed to a target polynucleotide by complementary base-painng 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 pπmers as used in the present invention typically compπse at least 15 contiguous nucleotides of a known sequence. In order to enhance specificity, longer probes and pnmers may also be employed, such as probes and pπmers that compπse 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 pπmers 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 prepaπng and using probes and pnmers are descnbed in the references, for example Sambrook, J. et al., 1989, Molecular Cloning: A Laboratory Manual, 2nd ed., vol 1-3, Cold Spring Harbor Press, Plamview NY; Ausubel, F.M. et al., 1987, Current Protocols m Molecular Biology, 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 pπmer pairs can be deπved from a known sequence, for example, by using computer programs intended for that purpose such as Pπmer (Version 0.5, 1991, Whitehead Institute for Biomedical Research, Cambndge MA).
O gonucleotides for use as pπmers are selected using software known in the art for such purpose. For example, OLIGO 4 06 software is useful for the selection of PCR pnmer pairs of up to 100 nucleotides each, and for the analysis of ohgonucleotides 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. For example, the PπmOU pnmer 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 pnmers from megabase sequences and is thus useful for designing pnmers on a genome-wide scope. The Pπmer3 primer selection program (available to the public from the Whitehead Institute/MIT Center for Genome Research, Cambndge MA) allows the user to input a "mispπming library," m which sequences to avoid as pπmer binding sites are user-specified. Pπmer3 is useful, in particular, for the selection of o gonucleotides for microarrays. (The source code for the latter two pπmer selection programs may also be obtained from their respective sources and modified to meet the user's specific needs ) The PπmeGen program (available to the public from the UK Human Genome Mapping Project Resource Centre, Cambndge UK) designs pπmers based on multiple sequence alignments, thereby allowing selection of pπmers that hybndize 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 ohgonucleotides and polynucleotide fragments identified by any of the above selection methods are useful in hybridization technologies, for example, as PCR or sequencing pπmers, microarray elements, or specific probes to identify fully or partially complementary polynucleotides in a sample of nucleic acids Methods of o gonucleotide selection are not limited to those described above A "recombinant nucleic acid" is a sequence that is not naturally occurring 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 engineeπng techniques such as those described m 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 denved 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, am o acid, or antibody Reporter molecules include radionuc des, enzymes, fluorescent, chemiluminescent, or chromogemc 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 nbose instead of deoxyπbose
The term "sample" is used in its broadest sense A sample suspected of containing nucleic acids encoding SYNT, or fragments thereof, or SYNT itself, may compπse 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 puπfied" 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 ngid or semi-ngid support including membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, tubing, plates, polymers, microparticles and capillanes The substrate can have a vaπety 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" descπbes a process by which exogenous DNA is introduced into a recipient cell Transformation may occur under natural or artificial conditions according to vaπous 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, bactenophage or viral infection, electroporation, heat shock, hpofection, 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 peπods of time A "transgenic organism," as used herein, is any organism, including but not limited to animals and plants, m 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 m 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 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 bactena, cyanobacteπa, 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 transferπng the DNA of the present invention into such organisms are widely known and provided in references such as Sambrook et al ( 1989), supra
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 2 0 9 (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 95% or at least 98% or greater sequence identity over a certain defined length A vanant may be descnbed as, for example, an "allehc" (as defined above), "splice," "species," or "polymorphic" vanant A splice vanant 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 duπng mRNA processing The corresponding polypeptide may possess additional functional domains or lack domains that are present m the reference molecule Species vaπants are polynucleotide sequences that vary from one species to another The resulting polypeptides generally will have significant am o 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 vaπants also may encompass "single nucleotide polymorphisms" (SNPs) in which the polynucleotide sequence vanes 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 "vanant" 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 2 0 9 (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 95%, or at least 98% 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 synthetases (SYNT), the polynucleotides encoding SYNT, and the use of these compositions for the diagnosis, treatment, or prevention of immune, neuronal, and reproductive disorders, and cell prohferative disorders including cancer Table 1 lists the Incyte clones used to assemble full length nucleotide sequences encoding
SYNT Columns 1 and 2 show the sequence identification numbers (SEQ ID NOs) of the polypeptide and nucleotide sequences, respectively Column 3 shows the clone IDs of the Incyte clones in which nucleic acids encoding each SYNT were identified, and column 4 shows the cDNA hbranes from which these clones were isolated Column 5 shows Incyte clones and their corresponding cDNA libraries Clones for which cDNA libraries are not indicated were deπved from pooled cDNA hbranes In some cases, GenBank sequence identifiers are also shown in column 5. The Incyte clones and GenBank cDNA sequences, where indicated, in column 5 were used to assemble the consensus nucleotide sequence of each SYNT and are useful as fragments in hybridization technologies.
The columns of Table 2 show various properties of each of the polypeptides of the invention, column 1 references the SEQ ID NO; column 2 shows the number of amino acid residues in each polypeptide, column 3 shows potential phosphorylation sites, column 4 shows potential glycosylation sites; column 5 shows the amino acid residues compnsing signature sequences and motifs; column 6 shows homologous sequences as identified by BLAST analysis, and column 7 shows analytical methods and in some cases, searchable databases to which the analytical methods were applied The methods of column 7 were used to characteπze each polypeptide through sequence homology and protein motifs.
The columns of Table 3 show the tissue-specificity and diseases, disorders, or conditions associated with nucleotide sequences encoding SYNT. The first column of Table 3 lists the nucleotide SEQ ID NOs. Column 2 lists fragments of the nucleotide sequences of column 1 These fragments are useful, for example, in hybndization or amplification technologies to identify SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO-22, SEQ ID N0 23, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO.29, and SEQ ID NO 30 and to distinguish between SEQ ID NO: 16, SEQ ID NO.17, SEQ ID NO: 18, SEQ ID NO- 19 SEQ ID NO:20, SEQ ID NO:21 , SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO.29, and SEQ ID NO-30 and related polynucleotide sequences. The polypeptides encoded by these fragments are useful, for example, as immunogenic peptides
Column 3 lists tissue categories which express SYNT as a fraction of total tissues expressing SYNT Column 4 lists diseases, disorders, or conditions associated with those tissues expressing SYNT as a fraction of total tissues expressing SYNT. Column 5 lists the vectors used to subclone each cDNA library The columns of Table 4 show descnptions of the tissues used to construct the cDNA hbranes from which cDNA clones encoding SYNT were isolated. Column 1 references the nucleotide SEQ ID NOs, column 2 shows the cDNA hbranes from which these clones were isolated, and column 3 shows the tissue origins and other descriptive information relevant to the cDNA hbranes m column 2. SEQ ID NO 16 maps to chromosome 5 within the interval from 147 10 to 150.00 centiMorgans SEQ ID NO: 17 maps to chromosome 10 within the interval from 137.60 to 139.20 centiMorgans This interval also contains gene MXI1, a member of the MYC family. SEQ ID NO 18 maps to chromosome 2 within the interval from 228.80 to 230.10 centiMorgans This interval also contains a gene for a proto-oncogene encoding a tyrosine-protem kinase SEQ ID NO 21 maps to chromosome 5 within the interval from 172.6 to 184 7 centiMorgans SEQ ID NO'24 maps to chromosome 2 within the interval from 1 18 0 to 127.4 centiMorgans. SEQ ID NO 26 maps to chromosome 3 within the interval from 157 4 to 162.0 centiMorgans. SEQ ID NO 27 maps to chromosome 12 within the interval from 97 1 to 1 16.6 centiMorgans. SEQ ID NO 28 maps to chromosome 4 within the interval from 77 3 to 99.2 centiMorgans and to chromosome 5 within the intervals from 79.2 to 92.3 centiMorgans, from 1 16.3 to 127.9 centiMorgans, and from 157.6 to 163 0 centiMorgans SEQ ID NO:29 maps to chromosome 1 within the interval from 242.5 to 258.7 centiMorgans and to chromosome 19 within the interval from 69.9 to 104.9 centiMorgans. SEQ ID NO:30 maps to chromosome 1 within the interval from 57.2 to 57.5 centiMorgans.
The invention also encompasses SYNT variants. A preferred SYNT vanant is one which has at least about 80%, or alternatively at least about 90%, or even at least about 95% ammo acid sequence identity to the SYNT ammo acid sequence, and which contains at least one functional or structural charactenstic of SYNT
The invention also encompasses polynucleotides which encode SYNT. In a particular embodiment, the invention encompasses a polynucleotide sequence compnsing a sequence selected from the group consisting of SEQ ID NO: 16-30, which encodes SYNT. The polynucleotide sequences of SEQ ID NO: 16-30, 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 πbose instead of deoxynbose.
The invention also encompasses a vanant of a polynucleotide sequence encoding SYNT. In particular, such a vanant polynucleotide sequence will have at least about 70%, or alternatively at least about 85%, or even at least about 95% polynucleotide sequence identity to the polynucleotide sequence encoding SYNT. A particular aspect of the invention encompasses a variant of a polynucleotide sequence compnsing a sequence selected from the group consisting of SEQ ID NO: 16-30 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 NO: 16-30. Any one of the polynucleotide vaπants descπbed above can encode an amino acid sequence which contains at least one functional or structural characteπstic of SYNT.
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 SYNT, some beaπng minimal similarity to the polynucleotide sequences of any known and naturally occurring gene, may be produced. Thus, the invention contemplates each and every possible vaπation of polynucleotide sequence that could be made by selecting combinations based on possible codon choices. These combinations are made in accordance with the standard tnplet genetic code as applied to the polynucleotide sequence of naturally occumng SYNT, and all such vanations are to be considered as being specifically disclosed
Although nucleotide sequences which encode SYNT and its vanants are generally capable of hybndizing to the nucleotide sequence of the naturally occumng SYNT under appropnately selected conditions of stπngency, it may be advantageous to produce nucleotide sequences encoding SYNT or its deπvatives possessing a substantially different codon usage, e.g., inclusion of non-naturally occumng 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 alteπng the nucleotide sequence encoding SYNT and its deπvatives without altenng the encoded ammo acid sequences include the production of RNA transcπpts having more desirable properties, such as a greater half-life, than transcnpts produced from the naturally occumng sequence
The invention also encompasses production of DNA sequences which encode SYNT and SYNT denvatives, 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 SYNT 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 ID NO: 16-30 and fragments thereof under various conditions of stnngency. (See, e.g., Wahl, G.M. and SL. Berger ( 1987) Methods Enzymol. 152:399-407; Kimmel, A.R. ( 1987) Methods Enzymol. 152:507-51 1 ) 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 (PE Biosystems, Foster City CA), 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 NV), PTC200 thermal cycler (MJ Research, Watertown MA) and ABI CATALYST 800 thermal cycler (PE Biosystems). Sequencing is then earned out using either the ABI 373 or 377 DNA sequencing system (PE 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 vanety of algonthms which are well known in the art. (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 Biology and Biotechnology. Wiley VCH, New York NY, pp 856-853 )
The nucleic acid sequences encoding SYNT 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 pnmers to amplify unknown sequence from genomic DNA within a cloning vector (See, e g , Sarkar, G ( 1993) PCR Methods Apphc 2 318-322 ) Another method, inverse PCR, uses primers that extend in divergent directions to amplify unknown sequence from a circulaπzed template The template is denved from restnction fragments compnsing a known genomic locus and surrounding sequences (See, e g , Tngha, 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 Apphc 1 1 1 1-1 19 ) In this method, multiple restnction enzyme digestions and hgations 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 retπeve 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 pnmers, and PROMOTERFINDER hbranes (Clontech, Palo Alto CA) to walk genomic DNA This procedure avoids the need to screen hbranes and is useful in finding intron/exon junctions For all PCR-based methods, primers may be designed using commercially available software, such as OLIGO 4 06 Pnmer Analysis software (National Biosciences, Plymouth MN) or another appropnate 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 hbranes that have been size-selected to include larger cDNAs In addition, random-pπmed hbranes, which often include sequences containing the 5' regions of genes, are preferable for situations in which an ohgo d(T) library does not yield a full-length cDNA Genomic hbranes may be useful for extension of sequence into 5' non-transcnbed 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 Output light intensity may be converted to electπcal signal using appropnate software (e g , GENOTYPER and SEQUENCE NAVIGATOR, PE 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 SYNT may be cloned in recombinant DNA molecules that direct expression of SYNT, or fragments or functional equivalents thereof, in appropnate 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 SYNT
The nucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter SYNT-encoding sequences for a vanety 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 ohgonucleotides may be used to engineer the nucleotide sequences. For example, ohgonucleotide- mediated site-directed mutagenesis may be used to introduce mutations that create new restπction sites, alter glycosylation patterns, change codon preference, produce splice vanants, and so forth
The nucleotides of the present invention may be subjected to DNA shuffling techniques such as MOLECULARBREEDING (Maxygen Inc., Santa Clara CA: descπbed in U.S Patent Number 5,837,458, Chang, C -C. et al ( 1999) Nat. Biotechnol. 17:793-797; Chπstians, F.C. et al. (1999) Nat. Biotechnol. 17.259-264; and Crameπ, A. et al. ( 1996) Nat. Biotechnol 14-315-319) to alter or improve the biological properties of SYNT, 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 vaπants is produced using PCR-mediated recombination of gene fragments. The library is then subjected to selection or screening procedures that identify those gene vanants with the desired properties. These preferred vanants 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 occumng genes in a directed and controllable manner. In another embodiment, sequences encoding SYNT 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, SYNT itself or a fragment thereof may be synthesized using chemical methods. For example, peptide synthesis can be performed using vanous 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 431 A peptide synthesizer (PE Biosystems) Additionally, the amino acid sequence of SYNT, or any part thereof, may be altered duπng direct synthesis and/or combined with sequences from other proteins, or any part thereof, to produce a vanant polypeptide or a polypeptide having a sequence of a naturally occumng polypeptide The peptide may be substantially punfied by preparative high performance liquid chromatography (See, e.g , Chiez, R M and F Z. Regmer ( 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 SYNT, the nucleotide sequences encoding SYNT or deπvatives thereof may be inserted into an appropnate expression vector, I e., a vector which contains the necessary elements for transcnptional 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 SYNT Such elements may vary in their strength and specificity Specific initiation signals may also be used to achieve more efficient translation of sequences encoding SYNT. Such signals include the ATG initiation codon and adjacent sequences, e.g the Kozak sequence. In cases where sequences encoding SYNT and its initiation codon and upstream regulatory sequences are inserted into the appropriate expression vector, no additional transcnptional 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 vanous oπgins, 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 SYNT and appropnate transcnptional 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 Spnng Harbor Press, Plainview NY, ch 4, 8, and 16-17, Ausubel, F M. et al. ( 1995) Current Protocols in Molecular Biology. John Wiley & Sons, New York NY, ch 9, 13, and 16.)
A vaπety of expression vector/host systems may be utilized to contain and express sequences encoding SYNT These include, but are not limited to, microorganisms such as bactena transformed with recombinant bactenophage, 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 bacteπal 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; Bitter, G.A. et al. ( 1987) Methods Enzymol. 153:516-544; Scorer, CA. et al. ( 1994) Bio/Technology 12: 181-184; 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; Takamatsu, N. ( 1987) EMBO J. 6:307-31 1 ; Coruzzi, G et al. ( 1984) EMBO J. 3: 1671-1680; Broghe, R. et al. ( 1984) Science 224:838-843; Winter, J et al. ( 1991) Results Probl. Cell Differ. 17:85- 105; The McGraw Hill Yearbook of Science and Technology ( 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 denved from retroviruses, adenoviruses, or herpes or vaccinia viruses, or from vaπous bactenal 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, I.M. and N. Somia (1997) Nature 389:239-242.) The invention is not limited by the host cell employed.
In bactenal systems, a number of cloning and expression vectors may be selected depending upon the use intended for polynucleotide sequences encoding SYNT. For example, routine cloning, subcloning, and propagation of polynucleotide sequences encoding SYNT can be achieved using a multifunctional E. coli vector such as PBLUESCRIPT (Stratagene, La Jolla CA) or PSPORT1 plasmid (Life Technologies). Ligation of sequences encoding SYNT into the vector's multiple cloning site disrupts the lac gene, allowing a coloπmetπc screening procedure for identification of transformed bactena 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 SYNT are needed, e.g. for the production of antibodies, vectors which direct high level expression of SYNT may be used. For example, vectors containing the strong, inducible T5 or T7 bactenophage promoter may be used.
Yeast expression systems may be used for production of SYNT. 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 pastons. 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, supra; Bitter, supra: and Scorer, supra.) Plant systems may also be used for expression of SYNT. Transcnption of sequences encoding SYNT may be dπven 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 6:307-31 1). Alternatively, plant promoters such as the small subunit of RUBISCO or heat shock promoters may be used. (See, e.g., Coruzzi, supra; Brog e, supra; and Winter, supra.) These constructs can be introduced into plant cells by direct DNA transformation or pathogen-mediated transfection. (See, e.g., The McGraw Hill Yearbook of Science and Technology ( 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 SYNT may be gated into an adenovirus transcnption/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential El or E3 region of the viral genome may be used to obtain infective virus which expresses SYNT in host cells. (See, e.g., Logan, J. and T. Shenk ( 1984) Proc Natl Acad. Sci. USA 81 :3655-3659.) In addition, transcnption 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, polycatio c 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 SYNT in cell lines is preferred. For example, sequences encoding SYNT can be transformed into cell lines using expression vectors which may contain viral ongins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells may be allowed to grow for about 1 to 2 days ennched 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 appropnate 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 phosphonbosyltransferase genes, for use in tk and apr cells, respectively (See, e.g., Wigler, M. et al. (1977) Cell 1 1 :223-232, Lowy, I. et al. (1980) Cell 22.817-823 ) Also, antimetabohte, antibiotic, or herbicide resistance can be used as the basis for selection. For example, dhfr confers resistance to methotrexate, neo confers resistance to the aminoglycosides neomycm and G-418; and als and pat confer resistance to chlorsulfuron and phosphinotncin 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., anthocyamns, green fluorescent proteins (GFP; Clontech), β glucuromdase and its substrate β-glucuronide, or luciferase and its substrate lucifeπn 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, CA. ( 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 SYNT is inserted within a marker gene sequence, transformed cells containing sequences encoding SYNT can be identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a sequence encoding SYNT 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 SYNT and that express
SYNT 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 hybndizations, 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 SYNT using either 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-interfeπng epitopes on SYNT 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 Laboratory Manual. APS Press, St. Paul MN, Sect. IV; Cohgan, J.E. et al. (1997) Current Protocols in Immunology. Greene Pub. Associates and Wiley-Interscience, New York NY; and Pound, J.D. (1998) Immunochemical Protocols. Humana Press, Totowa NJ.) A wide vaπety of labels and conjugation techniques are known by those skilled in the art and may be used in vaπous nucleic acid and amino acid assays. Means for producing labeled hybndization or PCR probes for detecting sequences related to polynucleotides encoding SYNT include oligolabehng, nick translation, end-labeling, or PCR amplification using a labeled nucleotide Alternatively, the sequences encoding SYNT, 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 appropnate RNA polymerase such as T7, T3, or SP6 and labeled nucleotides These procedures may be conducted using a vanety 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 radionuchdes, enzymes, fluorescent, chemiluminescent, or chromogenic agents, as well as substrates, cofactors, inhibitors, magnetic particles, and the like
Host cells transformed with nucleotide sequences encoding SYNT 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 SYNT may be designed to contain signal sequences which direct secretion of SYNT 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, phosphorylation, hpidation, 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 charactenstic mechanisms for post-translational activities (e.g., CHO, HeLa, MDCK, HEK293, and WI38) 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 SYNT may be hgated to a heterologous sequence resulting in translation of a fusion protein in any of the aforementioned host systems. For example, a chimeric SYNT protein containing a heterologous moiety that can be recognized by a commercially available antibody may facilitate the screening of peptide hbranes for inhibitors of SYNT activity. Heterologous protein and peptide moieties may also facilitate purification of fusion proteins using commercially available affinity matnces. Such moieties include, but are not limited to, glutathione S-transferase (GST), maltose binding protein (MBP), thioredoxin (Trx), calmodulin binding peptide (CBP), 6-Hιs, FLAG, c-myc, and hemagglutimn (HA) GST, MBP, Trx, CBP, and 6-Hιs enable punfication of their cognate fusion proteins on immobilized glutathione, maltose, phenylarsine oxide, calmodulin, and metal-chelate resins, respectively. FLAG, c-myc, and hemagglutimn (HA) enable immunoaffinity punfication 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 SYNT encoding sequence and the heterologous protein sequence, so that SYNT may be cleaved away from the heterologous moiety following purification Methods for fusion protein expression and punfication are discussed in Ausubel ( 1995, supra, ch. 10). A vaπety of commercially available kits may also be used to facilitate expression and punfication of fusion proteins.
In a further embodiment of the invention, synthesis of radiolabeled SYNT may be achieved in vitro using the TNT rabbit reticulocyte lysate or wheat germ extract system (Promega). These systems couple transcnption and translation of protein-coding sequences operably associated with the T7, T3, or SP6 promoters. Translation takes place in the presence of a radiolabeled amino acid precursor, for example, 35S-methιonιne.
SYNT of the present invention or fragments thereof may be used to screen for compounds that specifically bind to SYNT. At least one and up to a plurality of test compounds may be screened for specific binding to SYNT. Examples of test compounds include antibodies, o gonucleotides, proteins (e.g., receptors), or small molecules
In one embodiment, the compound thus identified is closely related to the natural ligand of SYNT, e.g , a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a natural binding partner (See, Cohgan, J.E. et al. ( 1991) Current Protocols in Immunology 1(2) Chapter 5.) Similarly, the compound can be closely related to the natural receptor to which SYNT 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 appropnate cells which express SYNT, either as a secreted protein or on the cell membrane Preferred cells include cells from mammals, yeast, Drosophila, or E. coli. Cells expressing SYNT or cell membrane fractions which contain SYNT are then contacted with a test compound and binding, stimulation, or inhibition of activity of either SYNT 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 compπse the steps of combining at least one test compound with SYNT, either in solution or affixed to a solid support, and detecting the binding of SYNT 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 earned out using cell-free preparations, chemical hbranes, or natural product mixtures, and the test compound(s) may be free in solution or affixed to a solid support.
SYNT of the present invention or fragments thereof may be used to screen for compounds that modulate the activity of SYNT. Such compounds may include agonists, antagonists, or partial or inverse agonists. In one embodiment, an assay is performed under conditions permissive for SYNT activity, wherein SYNT is combined with at least one test compound, and the activity of SYNT in the presence of a test compound is compared with the activity of SYNT in the absence of the test compound. A change in the activity of SYNT in the presence of the test compound is indicative of a compound that modulates the activity of SYNT Alternatively, a test compound is combined with an in vitro or cell-free system compnsing SYNT under conditions suitable for SYNT activity, and the assay is performed In either of these assays, a test compound which modulates the activity of SYNT 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 SYNT 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 No 5, 175,383 and U S Patent No 5,767,337 ) For example, mouse ES cells, such as the mouse 129/SvJ cell line, are deπved from the early mouse embryo and grown m 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 SYNT 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 1 145-1 147)
Polynucleotides encoding SYNT 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 SYNT 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 descnbed 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 SYNT, e g , by secreting SYNT 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 similanty, e g , in the context of sequences and motifs, exists between regions of SYNT and human synthetases In addition, the expression of SYNT is closely associated with hematopoietic/immune, cancerous, proliferating, inflamed, immune, nervous, gastrointestinal and reproductive tissues Therefore, SYNT appears to play a role in an immune disorder such as inflammation, actinic keratosis, acquired immunodeficiency syndrome (AIDS), Addison s disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, arteriosclerosis, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, bronchitis, bursitis, cholecystitis, cirrhosis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes melhtus, emphysema, erythroblastosis feta s, erythema nodosum, atrophic gastπtis, glomerulonephntis, Goodpasture's syndrome, gout, Graves' disease, Hashimoto's thyroiditis, paroxysmal nocturnal hemoglobinuπa, hepatitis, hypereosmophilia, irritable bowel syndrome, episodic lymphopenia with lymphocytotoxins, mixed connective tissue disease (MCTD), multiple sclerosis, myasthenia gravis, myocardial or peπcardial inflammation, myelofibrosis, osteoarthπtis, osteoporosis, pancreatitis, polycythemia vera, polymyositis, psoπasis, Reiter's syndrome, rheumatoid arthπtis, scleroderma, Sjogren's syndrome, systemic anaphylaxis, systemic lupus erythematosus, systemic sclerosis, pπmary thrombocythemia, thrombocytopenic purpura, ulcerative colitis, uveitis, Werner syndrome, complications of cancer, hemodialysis, and extracorporeal circulation, trauma, viral, bacterial, fungal, parasitic, protozoal, and helminthic infections, and hematopoietic cancer including lymphoma, leukemia, and myeloma; a neuronal disorder, such as akathesia, Alzheimer's disease, amnesia, amyotrophic lateral sclerosis, bipolar disorder, catatonia, cerebral neoplasms, dementia, depression, diabetic neuropathy, Down's syndrome, tardive dyskinesia, dystonias, epilepsy, Huntington's disease, peripheral neuropathy, multiple sclerosis, neurofibromatosis, Parkinson's disease, paranoid psychoses, postherpetic neuralgia, schizophrenia, and Tourette's disorder; a reproductive disorder, such as a disorder of prolactin production, infertility, including tubal disease, ovulatory defects, and endometnosis, a disruption of the estrous cycle, a disruption of the menstrual cycle, polycystic ovary syndrome, ovaπan hyperstimulation syndrome, an endometnal or ovaπan tumor, a uterine fibroid, autoimmune disorders, an ectopic pregnancy, and teratogenesis; cancer of the breast, fibrocystic breast disease, and galactorrhea; a disruption of spermatogenesis, abnormal sperm physiology, cancer of the testis, cancer of the prostate, benign prostatic hyperplasia, prostatitis, Peyronie's disease, impotence, carcinoma of the male breast, and gynecomastia; and a cell prohferative disorder, such as actinic keratosis, arteπosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCTD), myelofibrosis, paroxysmal nocturnal hemoglobinuπa, polycythemia vera, psonasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, a cancer 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
In another embodiment, a vector capable of expressing SYNT or a fragment or denvative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of SYNT including, but not limited to, those described above In a further embodiment, a pharmaceutical composition comprising a substantially punfied
SYNT in conjunction with a suitable pharmaceutical earner may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of SYNT including, but not limited to, those provided above
In still another embodiment, an agonist which modulates the activity of SYNT may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of SYNT including, but not limited to, those listed above
In a further embodiment, an antagonist of SYNT may be administered to a subject to treat or prevent a disorder associated with increased expression or activity of SYNT Examples of such disorders include, but are not limited to, those immune, neuronal, reproductive, and cell prohferative disorders descnbed above In one aspect, an antibody which specifically binds SYNT may be used directly as an antagonist or indirectly as a targeting or delivery mechanism for bnnging a pharmaceutical agent to cells or tissues which express SYNT
In an additional embodiment, a vector expressing the complement of the polynucleotide encoding SYNT may be administered to a subject to treat or prevent a disorder associated with increased expression or activity of SYNT including, but not limited to, those descnbed above
In other embodiments, any of the proteins, antagonists, antibodies, agonists, complementary sequences, or vectors of the invention may be administered m combination with other appropriate therapeutic agents Selection of the appropnate 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 vanous disorders descπbed 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 SYNT may be produced using methods which are generally known in the art In particular, punfied SYNT may be used to produce antibodies or to screen hbranes of pharmaceutical agents to identify those which specifically bind SYNT Antibodies to SYNT may also be generated using methods that are well known in the art Such antibodies may include, but are not limited to, polyclonal, monoclonal, chimenc, 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, vaπous hosts including goats, rabbits, rats, mice, humans, and others may be immunized by injection with SYNT or with any fragment or ohgopeptide 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 m humans, BCG (bacilli Calmette-Guenn) and Corynebacteπum parvum are especially preferable It is preferred that the ohgopeptides, peptides, or fragments used to induce antibodies to SYNT have an ammo 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 ohgopeptides, peptides, or fragments are identical to a portion of the amino acid sequence of the natural protein Short stretches of SYNT ammo acids may be fused with those of another protein, such as KLH, and antibodies to the chimenc molecule may be produced
Monoclonal antibodies to SYNT 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 hybndoma technique, the human B-cell hybπdoma technique, and the EBV-hybπdoma 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 "chimenc 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., Momson, 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 descπbed for the production of single chain antibodies may be adapted, using methods known in the art, to produce SYNT-specific single chain antibodies Antibodies with related specificity, but of distinct ldiotypic composition, may be generated by chain shuffling from random combinatorial immunoglobulin hbranes (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 m the lymphocyte population or by screening immunoglobulin hbranes 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 SYNT 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 bndges of the F(ab')2 fragments. Alternatively, Fab expression hbranes 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 lmmunoradiometπc 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 SYNT and its specific antibody A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfeπng SYNT epitopes is generally used, but a competitive binding assay may also be employed (Pound, supra)
Various methods such as Scatchard analysis in conjunction with radioimmunoassay techniques may be used to assess the affinity of antibodies for SYNT Affinity is expressed as an association constant, Ka, which is defined as the molar concentration of SYNT-antibody complex divided by the molar concentrations of free antigen and free antibody under equihbπum conditions The Ka determined for a preparation of polyclonal antibodies, which are heterogeneous in their affinities for multiple SYNT epitopes, represents the average affinity, or avidity, of the antibodies for SYNT The Ka determined for a preparation of monoclonal antibodies, which are monospecific for a particular SYNT epitope, represents a true measure of affinity High-affinity antibody preparations with K, ranging from about 109 to 1012 L/mole are preferred for use in immunoassays in which the SYNT-antibody complex must withstand πgorous manipulations Low-affinity antibody preparations with Ka ranging from about 106 to 107 L/mole are preferred for use in immunopuπfication and similar procedures which ultimately require dissociation of SYNT, 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 requiπng precipitation of SYNT-antibody complexes Procedures for evaluating antibody specificity, titer, and avidity, and guidelines for antibody quality and usage in vanous applications, are generally available (See, e g , Catty, supra, and Cohgan et al , supra ) In another embodiment of the invention, the polynucleotides encoding SYNT, 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 ohgonucleotides) to the coding or regulatory regions of the gene encoding SYNT Such technology is well known in the art, and antisense ohgonucleotides or larger fragments can be designed from vaπous locations along the coding or control regions of sequences encoding SYNT (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 mtracellularly 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 Chn. Immunol. 102(3).469-475, and Scanlon, K.J et al. ( 1995) 9( 13).1288- 1296.) Antisense sequences can also be introduced mtracellularly 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 hposome-deπved systems, artificial viral envelopes, and other systems known m the art (See, e.g., Rossi, J.J. ( 1995) Br. Med. Bull. 51(l):217-225; Boado, R.J. et al. ( 1998) J. Pharm. Sci. 87(1 1)- 1308-1315; and Moms, M.C. et al (1997) Nucleic Acids Res 25(14):2730-2736.)
In another embodiment of the invention, polynucleotides encoding SYNT may be used for somatic or germline gene therapy. Gene therapy may be performed to (l) correct a genetic deficiency (e.g., in the cases of severe combined immunodeficiency (SCID)-Xl disease characteπzed by X- hnked inhentance (Cavazzana-Calvo, M. et al. (2000) Science 288-669-672), severe combined immunodeficiency syndrome associated with an inhented 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 VIH or Factor IX deficiencies (Crystal, R.G. ( 1995) Science 270-404-410; Verma, I.M. and Somia, N (1997) Nature 389:239-242)), (n) express a conditionally lethal gene product (e.g., in the case of cancers which result from unregulated cell proliferation), or (in) 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: 1 1395-1 1399), hepatitis B or C virus (HBV, HCV); fungal parasites, such as Candida albicans and Paracoccidioides brasihensis, and protozoan parasites such as Plasmodium falciparum and Trypanosoma cruzi). In the case where a genetic deficiency in SYNT expression or regulation causes disease, the expression of SYNT from an appropnate 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 SYNT are treated by constructing mammalian expression vectors encoding SYNT and introducing these vectors by mechanical means into SYNT-deficient cells. Mechanical transfer technologies for use with cells in vivo or ex vitro include (I) direct DNA micromjection into individual cells, (n) ballistic gold particle delivery, (in) hposome-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. Biotechnol 9:445-450). Expression vectors that may be effective for the expression of SYNT 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). SYNT may be expressed using (I) a constitutively active promoter, (e.g., from cytomegalovirus (CMV), Rous sarcoma virus (RSV), SV40 virus, thymidine kinase (TK), or β-actin genes), (n) an inducible promoter (e.g , the tetracychne-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) Curr. Opin. Biotechnol. 9:451-456), commercially available in the T-REX plasmid (Invitrogen)); the ecdysone-mducible promoter (available in the plasmids PVGRXR and PIND; Invitrogen); the FK506/rapamycιn inducible promoter; or the RU486/mιfepπstone inducible promoter (Rossi, F.M.V. and H.M. Blau, supra)), or (iii) a tissue-specific promoter or the native promoter of the endogenous gene encoding SYNT 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 expenmental 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 pnmary 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 SYNT expression are treated by constructing a retrovirus vector consisting of (1) the polynucleotide encoding SYNT under the control of an independent promoter or the retrovirus long terminal repeat (LTR) promoter, (n) appropnate RNA packaging signals, and (in) a Rev-responsive element (RRE) along with additional retrovirus cu-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 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 retrovirus 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, ML. (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 SYNT to cells which have one or more genetic abnormalities with respect to the expression of SYNT. 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 descnbed in U.S. Patent Number 5,707,618 to Armentano ("Adenovirus vectors for gene therapy"), hereby incorporated by reference. For adenoviral vectors, see also Antmozzi, P.A. et al. ( 1999) Annu. Rev. Nutr. 19:51 1-544; and Verma, I.M. 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 SYNT to target cells which have one or more genetic abnormalities with respect to the expression of SYNT The use of herpes simplex virus (HSV)-based vectors may be especially valuable for introducing SYNT 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 pπmates (Liu, X. et al. (1999) Exp. Eye Res.169:385-395). The construction of a HSV-1 virus vector has also been disclosed m 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 appropnate 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 SYNT 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 Opm. Biotech. 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 SYNT into the alphavirus genome in place of the capsid-coding region results in the production of a large number of SYNT-codmg RNAs and the synthesis of high levels of SYNT 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 vanant of Smdbis 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 SYNT into a vanety of cell types The specific transduction of a subset of cells in a population may require the sorting of cells pπor 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 deπved from the transcnption 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 tπple helix base-painng methodology Tπple helix paiπng is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcπption factors, or regulatory molecules. Recent therapeutic advances using tπplex DNA have been descnbed in the literature. (See, e.g., Gee, J.E. et al. (1994) in Huber, B.E. and B.I. Carr, Molecular and Immunologic 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 transcnpt from binding to nbosomes.
Ribozymes, enzymatic RNA molecules, may also be used to catalyze the specific cleavage of RNA The mechanism of πbozyme action involves sequence-specific hybndization of the nbozyme molecule to complementary target RNA, followed by endonucleolytic cleavage. For example, engineered hammerhead motif nbozyme molecules may specifically and efficiently catalyze endonucleolytic cleavage of sequences encoding SYNT
Specific nbozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for nbozyme cleavage sites, including the following sequences GUA, GUU, and GUC Once identified, short RNA sequences of between 15 and 20 πbonucleotides, 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 hybndization with complementary o gonucleotides using nbonuclease protection assays
Complementary πbonucleic 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 ohgonucleotides such as solid phase phosphoramidite chemical synthesis Alternatively, RNA molecules may be generated by in vitro and in vivo transcnption of DNA sequences encoding SYNT Such DNA sequences may be incorporated into a wide vanety 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 mosine, queosine, and wybutosme, as well as acetyl-, methyl-, thio-, and similarly modified forms of adenine, cytidine, guanine, thymine, and undine 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 m altenng expression of a polynucleotide encoding SYNT Compounds which may be effective in altenng expression of a specific polynucleotide may include, but are not limited to, ohgonucleotides, antisense ohgonucleotides, tnple helix-forming ohgonucleotides, transcπption factors and other polypeptide transcnptional 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 SYNT expression or activity, a compound which specifically inhibits expression of the polynucleotide encoding SYNT may be therapeutically useful, and in the treament of disorders associated with decreased SYNT expression or activity, a compound which specifically promotes expression of the polynucleotide encoding SYNT may be therapeutically useful At least one, and up to a plurality, of test compounds may be screened for effectiveness in altenng 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 altenng polynucleotide expression; selection from an existing, commercially-available or propnetary library of naturally-occurnng 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 combinatonally or randomly A sample compnsing a polynucleotide encoding SYNT is exposed to at least one test compound thus obtained The sample may compnse, for example, an intact or permeabihzed cell, or an in vitro cell-free or reconstituted biochemical system. Alterations in the expression of a polynucleotide encoding SYNT are assayed by any method commonly known in the art Typically, the expression of a specific nucleotide is detected by hybndization with a probe having a nucleotide sequence complementary to the sequence of the polynucleotide encoding SYNT The amount of hybndization may be quantified, thus forming the basis for a compaπson 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 altenng the expression of the polynucleotide. A screen for a compound effective in altenng expression of a specific polynucleotide can be earned out, for example, using a Schizosaccharomyces 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 Commun. 268 8- 13). A particular embodiment of the present invention involves screening a combinatoπal library of ohgonucleotides (such as deoxynbonucleotides, πbonucleotides, peptide nucleic acids, and modified ohgonucleotides) 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. Biotechnol 15-462-466 ) Any of the therapeutic methods descnbed 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 pharmaceutical composition which generally compnses an active ingredient formulated with a pharmaceutically acceptable excipient Excipients may include, for example, sugars, starches, celluloses, gums, and proteins Vaπous formulations are commonly known and are thoroughly discussed in the latest edition of Remington's Pharmaceutical Sciences (Maack Publishing, Easton PA). Such pharmaceutical compositions may consist of SYNT, antibodies to SYNT, and mimetics, agonists, antagonists, or inhibitors of SYNT
The pharmaceutical compositions utilized in this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, lntra-arteπal, intramedullary, intrathecal, intraventπcular, pulmonary, transdermal, subcutaneous, intrapentoneal, intranasal, enteral, topical, sublingual, or rectal means.
Pharmaceutical compositions for pulmonary administration may be prepared in liquid or dry powder form. These compositions are generally aerosolized immediately pnor 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.
Pharmaceutical compositions suitable for use in the invention include compositions wherein the active ingredients are contained m 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 pharmaceutical compositions may be prepared for direct intracellular delivery of macromolecules compnsing SYNT or fragments thereof. For example, liposome preparations containing a cell-impermeable macromolecule may promote cell fusion and intracellular delivery of the macromolecule. Alternatively, SYNT 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 SYNT or fragments thereof, antibodies of SYNT, and agonists, antagonists or inhibitors of SYNT, which ameliorates the symptoms or condition. Therapeutic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or with expeπmental animals, such as by calculating the ED50 (the dose therapeutically effective in 50% of the population) or LD50 (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 LD50/ED,0 ratio Pharmaceutical 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 ED50 with little or no toxicity The dosage vanes 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 seventy of the disease state, the general health of the subject, the age, weight, and gender of the subject, time and frequency of administration, drug combmatιon(s), reaction sensitivities, and response to therapy Long-acting pharmaceutical 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 μg to 100,000 μg, 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 SYNT may be used for the diagnosis of disorders characteπzed by expression of SYNT, or in assays to monitor patients being treated with SYNT or agonists, antagonists, or inhibitors of SYNT Antibodies useful for diagnostic purposes may be prepared in the same manner as descπbed above for therapeutics Diagnostic assays for SYNT include methods which utilize the antibody and a label to detect SYNT m human body fluids or m 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 vaπety of reporter molecules, several of which are descnbed above, are known in the art and may be used
A variety of protocols for measunng SYNT, including ELISAs, RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of SYNT expression Normal or standard values for SYNT expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, for example, human subjects, with antibody to SYNT under conditions suitable for complex formation The amount of standard complex formation may be quantitated by vaπous methods, such as photometnc means Quantities of SYNT expressed in subject, control, and disease samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease.
In another embodiment of the invention, the polynucleotides encoding SYNT may be used for diagnostic purposes. The polynucleotides which may be used include ohgonucleotide 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 SYNT may be correlated with disease The diagnostic assay may be used to determine absence, presence, and excess expression of SYNT, and to monitor regulation of SYNT levels during therapeutic intervention
In one aspect, hybndization with PCR probes which are capable of detecting polynucleotide sequences, including genomic sequences, encoding SYNT or closely related molecules may be used to identify nucleic acid sequences which encode SYNT. 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 hybndization or amplification will determine whether the probe identifies only naturally occurring sequences encoding SYNT, allehc vanants, 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 SYNT encoding sequences The hybridization probes of the subject invention may be DNA or RNA and may be denved from the sequence of SEQ ID NO- 16-30 or from genomic sequences including promoters, enhancers, and introns of the SYNT gene Means for producing specific hybndization probes for DNAs encoding SYNT include the cloning of polynucleotide sequences encoding SYNT or SYNT denvatives 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 appropnate RNA polymerases and the appropriate labeled nucleotides Hybndization probes may be labeled by a vaπety of reporter groups, for example, by radionuchdes such as 32P or 35S, or by enzymatic labels, such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems, and the like.
Polynucleotide sequences encoding SYNT may be used for the diagnosis of disorders associated with expression of SYNT Examples of such disorders include, but are not limited to, an immune disorder such as inflammation, actinic keratosis, acquired immunodeficiency syndrome (AIDS), Addison 's disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, arteπosclerosis, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, bronchitis, bursitis, cholecystitis, cirrhosis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes melhtus, emphysema, erythroblastosis fetahs, erythema nodosum, atrophic gastπtis, glomerulonephntis, Goodpasture's syndrome, gout, Graves' disease, Hashimoto's thyroiditis, paroxysmal nocturnal hemoglobinuna, hepatitis, hypereosmophilia, lrntable bowel syndrome, episodic lymphopema with lymphocytotoxms, mixed connective tissue disease (MCTD), multiple sclerosis, myasthema gravis, myocardial or peπcardial inflammation, myelofibrosis, osteoarthntis, osteoporosis, pancreatitis, polycythemia vera, polymyositis, psoπasis, Reiter's syndrome, rheumatoid arthπtis, scleroderma, Sjogren's syndrome, systemic anaphylaxis, systemic lupus erythematosus, systemic sclerosis, pπmary thrombocythemia, thrombocytopemc purpura, ulcerative colitis, uveitis, Werner syndrome, complications of cancer, hemodialysis, and extracorporeal circulation, trauma, viral, bactenal, fungal, parasitic, protozoal, and helminthic infections, and hematopoietic cancer including lymphoma, leukemia, and myeloma, a neuronal disorder, such as akathesia, Alzheimer's disease, amnesia, amyotrophic lateral sclerosis, bipolar disorder, catatonia, cerebral neoplasms, dementia, depression, diabetic neuropathy, Down s syndrome, tardive dyskinesia, dystonias, epilepsy, Huntington's disease, peπpheral neuropathy, multiple sclerosis, neurofibromatosis, Parkinson's disease, paranoid psychoses, postherpetic neuralgia, schizophrenia, and Tourette's disorder, a reproductive disorder, such as a disorder of prolactm production, infertility, including tubal disease, ovulatory defects, and endometπosis, a disruption of the estrous cycle, a disruption of the menstrual cycle, polycystic ovary syndrome, ovarian hyperstimulation syndrome, an endometrial or ovaπan tumor, a utenne fibroid, autoimmune disorders, an ectopic pregnancy, and teratogenesis, cancer of the breast, fibrocystic breast disease, and galactorrhea, a disruption of spermatogenesis, abnormal sperm physiology, cancer of the testis, cancer of the prostate, benign prostatic hyperplasia, prostatitis, Peyronie's disease, impotence, carcinoma of the male breast, and gynecomastia, and a cell prohferative disorder, such as actinic keratosis, artenosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCTD), myelofibrosis, paroxysmal nocturnal hemoglobinuna, polycythemia vera, psoriasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, a cancer 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, skm, spleen, testis, thymus, thyroid, and uterus The polynucleotide sequences encoding SYNT may be used in Southern or northern analysis, dot blot, or other membrane-based technologies, in PCR technologies, in dipstick, pin, and multiformat ELISA-hke assays, and in microarrays utilizing fluids or tissues from patients to detect altered SYNT expression Such qualitative or quantitative methods are well known in the art
In a particular aspect, the nucleotide sequences encoding SYNT may be useful m assays that detect the presence of associated disorders, particularly those mentioned above The nucleotide sequences encoding SYNT may be labeled by standard methods and added to a fluid or tissue sample from a patient under conditions suitable for the formation of hybndization complexes After a suitable incubation penod, the sample is washed and the signal is quantified and compared with a standard value If the amount of signal m the patient sample is significantly altered in companson to a control sample then the presence of altered levels of nucleotide sequences encoding SYNT m 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 tnals, 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
SYNT, 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 SYNT, under conditions suitable for hybndization or amplification. Standard hybndization may be quantified by companng the values obtained from normal subjects with values from an expenment in which a known amount of a substantially punfied 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 determine 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 penod ranging from several days to months
With respect to cancer, the presence of an abnormal amount of transcnpt (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 pπor 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 ohgonucleotides designed from the sequences encoding SYNT may involve the use of PCR. These ohgomers may be chemically synthesized, generated enzymatically, or produced m vitro Ohgomers will preferably contain a fragment of a polynucleotide encoding SYNT, or a fragment of a polynucleotide complementary to the polynucleotide encoding SYNT, and will be employed under optimized conditions for identification of a specific gene or condition. Ohgomers may also be employed under less stnngent conditions for detection or quantification of closely related DNA or RNA sequences
In a particular aspect, ohgonucleotide pnmers derived from the polynucleotide sequences encoding SYNT may be used to detect single nucleotide polymorphisms (SNPs). SNPs are substitutions, insertions and deletions that are a frequent cause of inheπted or acquired genetic disease in humans Methods of SNP detection include, but are not limited to, single-stranded conformation polymorphism (SSCP) and fluorescent SSCP (fSSCP) methods In SSCP, ohgonucleotide pπmers denved from the polynucleotide sequences encoding SYNT are used to amplify DNA using the polymerase chain reaction (PCR) The DNA may be denved, 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 detectable using gel electrophoresis in non-denatunng gels In fSCCP, the ohgonucleotide pπmers are fluorescently labeled, which allows detection of the amp mers in high- throughput equipment such as DNA sequencing machines Additionally, sequence database analysis methods, termed in sihco SNP (isSNP), are capable of identifying polymorphisms by companng the sequence of individual overlapping DNA fragments which assemble into a common consensus sequence These computer-based methods filter out sequence vanations 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 charactenzed 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 SYNT include radiolabelmg or biotinylatmg nucleotides, coamphfication 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 ohgomer or polynucleotide of interest is presented m various dilutions and a spectrophotometπc or coloπmetπc response gives rapid quantitation
In further embodiments, ohgonucleotides or longer fragments denved from any of the polynucleotide sequences descnbed herein may be used as elements on a microarray The microarray can be used in transcnpt imaging techniques which monitor the relative expression levels of large numbers of genes simultaneously as descnbed in Seilhamer, J J et al , "Comparative Gene Transcnpt Analysis," U S Patent No 5,840,484, incorporated herein by reference The microarray may also be used to identify genetic vaπants, 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 appropnate 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, antibodies specific for SYNT, or SYNT or fragments thereof may be used as elements on a microarray The microarray may be used to monitor or measure protein-protem interactions, drug-target interactions, and gene expression profiles, as descnbed 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 transcnpt image may be generated by hybridizing the polynucleotides of the present invention or their complements to the totality of transcπpts or reverse transcπpts of a particular tissue or cell type. In one embodiment, the hybndization takes place in high-throughput format, wherein the polynucleotides of the present invention or their complements compnse a subset of a plurality of elements on a microarray The resultant transcnpt image would provide a profile of gene activity.
Transcript images may be generated using transcπpts isolated from tissues, cell lines, biopsies, or other biological samples The transcnpt image may thus reflect gene expression in vivo, as in the case of a tissue or biopsy sample, or vitro, as m the case of a cell line Transcnpt images which profile the expression of the polynucleotides of the present invention may also be used m conjunction with in vitro model systems and prechnical evaluation of pharmaceuticals, as well as toxicological testing of industπal and naturally-occurring environmental compounds All compounds induce charactenstic 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; Sterner, S. and N.L. Anderson (2000) Toxicol Lett. 1 12-1 13:467-471, 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 fingerpπnts 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 compaπson 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 hybπdized with one or more probes specific to the polynucleotides of the present invention, so that transcnpt levels corresponding to the polynucleotides of the present invention may be quantified The transcnpt levels in the treated biological sample are compared with levels in an untreated biological sample Differences in the transcnpt 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 isoelectnc focusing in the first dimension, and then according to molecular weight by sodium dodecyl sulfate slab gel electrophoresis in the second dimension (Sterner 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 companng its partial sequence, preferably of at least 5 contiguous am o 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 SYNT to quantify the levels of SYNT 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- 1 1 1 , Mendoze, L G et al (1999) Biotechniques 27 778-788) Detection may be performed by a vaπety of methods known in the art, for example, by reacting the proteins in the sample with a thiol- or ammo-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 transcnpt level There is a poor correlation between transcnpt 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 transcnpt 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 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 m 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 W095/251 1 16; Shalon, D et al ( 1995) PCT application WO95/35505; 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 descπbed 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 SYNT may be used to generate hybndization probes useful in mapping the naturally occumng genomic sequence Either coding or noncodmg sequences may be used, and in some instances, noncodmg sequences may be preferable over coding sequences. For example, conservation of a coding sequence among members of a multi-gene family may potentially cause undesired cross hybridization duπng 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), bactenal artificial chromosomes (BACs), bactenal PI constructions, or single chromosome cDNA libraries (See, e g , Harπngton, J J et al ( 1997) Nat Genet 15 345-355 Pnce, 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 mhentance of a disease state with the inheritance of a particular chromosome region or restriction fragment length polymorphism (RFLP) (See, e g , Lander, E S and D Botstein ( 1986) Proc Natl Acad Sci USA 83 7353-7357 )
Fluorescent in situ hybndization (FISH) may be correlated with other physical and genetic map data (See, e g , Heinz-Ulπch, et al ( 1995) in Meyers, supra, pp 965-968 ) Examples of genetic map data can be found m vaπous scientific journals or at the Online Mendehan Inheπtance m Man (OMIM) World Wide Web site Correlation between the location of the gene encoding SYNT 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 hybndization 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 1 lq22-23, 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, earner, or affected individuals
In another embodiment of the invention, SYNT, its catalytic or immunogenic fragments, or ohgopeptides thereof can be used for screening hbranes of compounds in any of a vanety 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 mtracellularly The formation of binding complexes between SYNT 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 WO84/03564 ) In this method, large numbers of different small test compounds are synthesized on a solid substrate The test compounds are reacted with SYNT, or fragments thereof, and washed Bound SYNT is then detected by methods well known in the art Punfied SYNT 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 SYNT specifically compete with a test compound for binding SYNT In this manner, antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants with SYNT In additional embodiments, the nucleotide sequences which encode SYNT 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 further 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 preferred specific 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, in particular U S Ser No 60/144,992 and U S Ser No 60/168,858 are hereby expressly incorporated by reference
EXAMPLES I. Construction of cDNA Libraries
RNA was purchased from Clontech or isolated from tissues described in Table 4 SoVne tissues were homogenized and lysed in guamdinium 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 centnfuged over CsCl 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 hbranes, poly(A+) RNA was isolated using ohgo d(T)-coupled paramagnetic particles (Promega), OLIGOTEX latex particles (QIAGEN, Chatsworth CA), or an OLIGOTEX mRNA punfication kit (QIAGEN) Alternatively, RNA was isolated directly from tissue lysates using other RNA isolation kits, e g , the POLY(A)PURE mRNA punfication kit (Ambion, Austin TX)
In some cases, Stratagene was provided with RNA and constructed the corresponding cDNA hbranes Otherwise, cDNA was synthesized and cDNA hbranes 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 transcπption was initiated using ohgo d(T) or random pπmers Synthetic ohgonucleotide adapters were ligated to double stranded cDNA, and the cDNA was digested with the appropnate restriction enzyme or enzymes. For most hbranes. the cDNA was size-selected (300- 1000 bp) using SEPHACRYL S 1000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (Amersham Pharmacia Biotech) or preparative agarose gel electrophoresis. cDNAs were ligated into compatible restπction enzyme sites of the polyhnker of a suitable plasmid, e.g., PBLUESCRIPT plasmid (Stratagene), PSPORTl plasmid (Life Technologies), pcDNA2.1 plasmid (Invitrogen, Carlsbad CA), or pINCY plasmid (Incyte Genomics, Palo Alto CA) Recombinant plasmids were transformed into competent E. coli cells including XLl-Blue, XLl-BlueMRF, or SOLR from Stratagene or DH5α, DH10B, or ElectroMAX DH10B from Life Technologies.
II. Isolation of cDNA Clones Plasmids obtained as descπbed in Example I were recovered from host cells by in vivo excision using the UNIZAP vector system (Stratagene) or by cell lysis. Plasmids were punfied using at least one of the following: a Magic or WIZARD Minipreps DNA purification system (Promega); an AGTC Mi prep punfication kit (Edge Biosystems, Gaithersburg MD); and QIAWELL 8 Plasmid, QIAWELL 8 Plus Plasmid, QIAWELL 8 Ultra Plasmid punfication systems or the R E.A.L. PREP 96 plasmid punfication kit from QIAGEN. Following precipitation, plasmids were resuspended in 0.1 ml of distilled water and stored, with or without lyophihzation, 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 fluorometπcally 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 descnbed in Example II were sequenced as follows Sequencing reactions were processed using standard methods or high-throughput instrumentation such as the ABI CATALYST 800 (PE 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 (PE Biosystems). Electrophoretic separation of cDNA sequencing reactions and detection of labeled polynucleotides were earned out using the MEGABACE 1000 DNA sequencing system (Molecular Dynamics); the ABI PRISM 373 or 377 sequencing system (PE 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 VI
The polynucleotide sequences derived from cDNA sequencing were assembled and analyzed using a combination of software programs which utilize algonthms well known to those skilled in the art Table 5 summaπzes the tools, programs, and algonthms used and provides applicable descriptions, references, and threshold parameters The first column of Table 5 shows the tools, programs, and algonthms used, the second column provides bnef descriptions thereof, the third column presents appropnate 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, the greater the homology between two sequences) Sequences were analyzed using MACDNASIS PRO software (Hitachi Software Engineenng, South San Francisco CA) and LASERGENE software (DNASTAR) Polynucleotide and polypeptide sequence alignments were generated using the default parameters specified by the clustal algonthm as incorporated into the MEGALIGN multisequence alignment program (DNASTAR), which also calculates the percent identity between aligned sequences
The polynucleotide sequences were validated by removing vector, linker, and polyA sequences and by masking ambiguous bases, using algorithms and programs based on BLAST, dynamic programing, and dinucleotide nearest neighbor analysis The sequences were then queπed against a selection of public databases such as the GenBank pnmate, rodent, mammalian, vertebrate, and eukaryote databases, and BLOCKS, PRINTS, DOMO, PRODOM, and PFAM to acquire annotation using programs based on BLAST, FASTA, and BLIMPS The sequences were assembled into full length polynucleotide sequences using programs based on Phred, Phrap, and Consed, and were screened for open reading frames using programs based on GeneMark, BLAST, and FASTA The full length polynucleotide sequences were translated to deπve the corresponding full length ammo acid sequences, and these full length sequences were subsequently analyzed by querying against databases such as the GenBank databases (described above), SwissProt, BLOCKS, PRINTS, DOMO, PRODOM, Prosite, and Hidden Markov Model (HMM)-based protein family databases such as PFAM HMM is a probabilistic approach which analyzes consensus pnmary structures of gene families (See, e g , Eddy, S R ( 1996) Curr Opin Struct Biol 6 361-365 ) The programs descnbed above for the assembly and analysis of full length polynucleotide and amino acid sequences were also used to identify polynucleotide sequence fragments from SEQ ID NO 16-30 Fragments from about 20 to about 4000 nucleotides which are useful m hybndization and amplification technologies were described in The Invention section above IV. Analysis of Polynucleotide Expression Northern analysis is a laboratory technique used to detect the presence of a transcnpt of a gene and involves the hybndization 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, supra, 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 categoπzed as exact or similar. The basis of the search is the product score, which is defined as:
BLAST Score x Percent Identity
5 x minimum { length(Seq 1), length(Seq. 2) }
The product score takes into account both the degree of similanty between two sequences and the 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 score of +5 for every base that matches in a high-scoπng 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
The results of northern analyses are reported as a percentage distnbution of hbranes in which the transcnpt encoding SYNT occurred. Analysis involved the categoπzation of cDNA hbranes by organ/tissue and disease. The organ/tissue categones included cardiovascular, dermatologic, developmental, endocπne, gastrointestinal, hematopoietic/immune, musculoskeletal, nervous, reproductive, and urologic. The disease/condition categones included cancer, inflammation, trauma, cell proliferation, neurological, and pooled. For each category, the number of hbranes expressing the sequence of interest was counted and divided by the total number of hbranes across all categones Percentage values of tissue-specific and disease- or condition-specific expression are reported in Table 3 V. Extension of SYNT Encoding Polynucleotides
The full length nucleic acid sequences of SEQ ID NO: 16-30 were produced by extension of an appropnate fragment of the full length molecule using ohgonucleotide pnmers designed from this fragment. One pnmer was synthesized to initiate 5' extension of the known fragment, and the other primer, to initiate 3' extension of the known fragment The initial pnmers 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 pπmer-pnmer dimeπzations 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 pπmers 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 pπmer, reaction buffer containing Mg2*, (NH4)2S04, and β-mercaptoethanol, Taq DNA polymerase (Amersham Pharmacia Biotech), ELONGASE enzyme (Life Technologies), and Pfu DNA polymerase (Stratagene), with the following parameters for pπmer pair PCI A and PCI B: Step 1. 94°C, 3 mm; Step 2: 94°C, 15 sec; Step 3. 60°C, 1 mm; Step 4. 68 °C, 2 mm; Step 5 Steps 2, 3, and 4 repeated 20 times; Step 6- 68°C, 5 mm, Step 7. storage at 4°C In the alternative, the parameters for pnmer pair T7 and SK+ were as follows: Step 1 94°C, 3 mm, Step 2 94°C, 15 sec, Step 3 57 °C, 1 min; Step 4: 68°C, 2 mm; 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 μl PICOGREEN quantitation reagent (0.25% (v/v) PICOGREEN; Molecular Probes, Eugene OR) dissolved in IX TE and 0.5 μl of undiluted PCR product into each well of an opaque fluonmeter 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 μl to 10 μl aliquot of the reaction mixture was analyzed by electrophoresis on a 1 % agarose mini-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 pnor to rehgation 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 rehgated using T4 hgase (New England Biolabs, Beverly MA) into pUC 18 vector (Amersham Pharmacia Biotech), treated with Pfu DNA polymerase (Stratagene) to fill-in restπction 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 LB/2x 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 m ; Step 2: 94°C 15 sec; Step 3 60°C, 1 min; Step 4. 72°C, 2 m , Step 5- steps 2, 3, and 4 repeated 29 times; Step 6- 72°C, 5 mm; Step 7- storage at 4°C DNA was quantified by PICOGREEN reagent (Molecular Probes) as described above Samples with low DNA recovenes were reamp fied using the same conditions as described above. Samples were diluted with 20% dimethysulfoxide ( 1 2, v/v), and sequenced using DYENAMIC energy transfer sequencing pπmers and the DYENAMIC DIRECT kit (Amersham Pharmacia Biotech) or the ABI PRISM BIGDYE Terminator cycle sequencing ready reaction kit (PE Biosystems)
In like manner, the polynucleotide sequences of SEQ ID NO: 16-30 are used to obtain 5' regulatory sequences using the procedure above, along with ohgonucleotides designed for such extension, and an appropnate genomic library. V. Chromosomal Mapping of SNYT Encoding Polynucleotides
The cDNA sequences which were used to assemble SEQ ID NO: 16-30 were compared with sequences from the Incyte LIFESEQ database and public domain databases using BLAST and other implementations of the Smith- Waterman algonthm. Sequences from these databases that matched SEQ ID NO: 16-30 were assembled into clusters of contiguous and overlapping sequences using assembly algonthms such as Phrap (Table 5) 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 cluster resulted in the assignment of all sequences of that cluster, including its particular SEQ ID NO-, to that map location.
The genetic map locations of SEQ ID NO.16, SEQ ID NO: 17, SEQ ID NO- 18, SEQ ID NO:21, SEQ ID NO.24, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO-29, and SEQ ID NO'30 are descπbed in The Invention as ranges, or intervals, of human chromosomes More than one map location is reported for SEQ ID NO.28 and SEQ ID NO:29, indicating that previously mapped sequences having similanty, but not complete identity, to SEQ ID N0 28 and SEQ ID NO:29 were assembled into their respective clusters. The map 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 boundanes for radiation hybnd markers whose sequences were included in each of the clusters Diseases associated with the public and Incyte sequences located within the indicated intervals are also reported in the Invention where applicable. Human genome maps and other resources available to the public, such as the NCBI "GeneMap'99" World Wide Web site which can be accessed at http://www.ncbi.nlm.nih.gov/genemap. can be employed to determine if previously identified disease genes map within or in proximity to the intervals indicated above VI. Labeling and Use of Individual Hybridization Probes
Hybndization probes deπved from SEQ ID NO 16-30 are employed to screen cDNAs, genomic DNAs. or mRNAs Although the labeling of ohgonucleotides, consisting of about 20 base pairs, is specifically descnbed, essentially the same procedure is used with larger nucleotide fragments Ohgonucleotides are designed using state-of-the-art software such as OLIGO 4 06 software (National Biosciences) and labeled by combining 50 pmol of each ohgomer, 250 μCi of [γ-32P] adenosine tπphosphate (Amersham Pharmacia Biotech), and T4 polynucleotide kinase (DuPont NEN, Boston MA) The labeled ohgonucleotides are substantially purified using a SEPHADEX G-25 superfine size exclusion dextran bead column (Amersham Pharmacia Biotech) An aliquot containing 107 counts per minute of the labeled probe is used in a typical membrane-based hybndization analysis of human genomic DNA digested with one of the following endonucleases Ase I, Bgl II, Eco Rl. 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 earned 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 Hybndization patterns are visualized using autoradiography or an alternative imaging means and compared VII. Microarrays
The linkage or synthesis of array elements upon a microarray can be achieved utilizing photolithography, piezoelectnc pnnting (ink-jet printing, See, e g , Baldeschweiler, supra), mechanical microspotting technologies, and denvatives 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 appropnate number of elements (See, e g , Schena, M et al ( 1995) Science 270 467-470, Shalon, 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 ohgomers thereof may compnse the elements of the microarray Fragments or ohgomers suitable for hybndization can be selected using software well known in the art such as LASERGENE software (DNASTAR) The array elements are hybndized 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, nonhybndized 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 hybndization The degree of complementanty and the relative abundance of each polynucleotide which hybndizes to an element on the microarray may be assessed In one embodiment, microarray preparation and usage is descnbed in detail below Tissue or Cell Sample Preparation
Total RNA is isolated from tissue samples using the guamdinium thiocyanate method and poly(A)+ RNA is punfied using the ohgo-(dT) cellulose method Each poly(A)+ RNA sample is reverse transcπbed using MMLV reverse-transcπptase, 0 05 pg/μl ohgo-(dT) primer (21mer), IX first strand buffer, 0 03 units/μl RNase inhibitor, 500 μM dATP, 500 μM dGTP, 500 μM dTTP, 40 μM dCTP, 40 μM dCTP-Cy3 (BDS) or dCTP-Cy5 (Amersham Pharmacia Biotech) The reverse transcnption 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 transcnption 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 punfied 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 μl 5X SSC/0 2% SDS Microarray Preparation
Sequences of the present invention are used to generate array elements Each array element is amplified from bactenal cells containing vectors with cloned cDNA inserts PCR amplification uses pπmers 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 μg Amplified array elements are then punfied using SEPHACRYL-400 (Amersham Pharmacia Biotech) Punfied 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% ammopropyl silane (Sigma) in 95% ethanol Coated slides are cured in a 1 10°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 μl of the array element DNA, at an average concentration of 100 ng/μl, is loaded into the open capillary printing element by a high-speed robotic apparatus The apparatus then deposits about 5 nl of array element sample per slide
Microarrays are UV-crosslinked using a STRATALINKER UV-crosshnker (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 μl of sample mixture consisting of 0 2 μg each of Cy3 and
Cy5 labeled cDNA synthesis products in 5X SSC, 0 2% SDS hybndization buffer The sample mixmre is heated to 65 °C for 5 minutes and is ahquoted onto the microarray surface and covered with an 1 8 cm2 covershp 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 μl 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 ( IX SSC, 0 1 % SDS), three times for 10 minutes each at 45 °C in a second wash buffer (0 IX SSC), and dried Detection Reporter-labeled hybndization 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 Cy5 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 R1477, Hamamatsu Photonics Systems, Bπdgewater NJ) corresponding to the two fluorophores Appropnate 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 Cy5 Each array is typically scanned twice, one scan per fluorophore using the appropnate 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 hybndizing 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 hybndized 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 hybndization 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 gπd is supeπmposed 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 numencal value corresponding to the average intensity of the signal The software used for signal analysis is the GEMTOOLS gene expression analysis program (Incyte)
VIII. Complementary Polynucleotides
Sequences complementary to the SYNT-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occumng SYNT Although use of ohgonucleotides compnsing from about 15 to 30 base pairs is descnbed, essentially the same procedure is used with smaller or with larger sequence fragments Appropnate ohgonucleotides are designed using OLIGO 4 06 software (National Biosciences) and the coding sequence of SYNT To inhibit transcπption, a complementary o gonucleotide is designed from the most unique 5' sequence and used to prevent promoter binding to the coding sequence To inhibit translation, a complementary ohgonucleotide is designed to prevent πbosomal binding to the SYNT-encod g transcnpt
IX. Expression of SYNT
Expression and punfication of SYNT is achieved using bacterial or virus-based expression systems For expression of SYNT bacteπa, cDNA is subcloned into an appropnate vector containing an antibiotic resistance gene and an inducible promoter that directs high levels of cDNA transcnption Examples of such promoters include, but are not limited to, the trp-lac (tac) hybπd promoter and the T5 or T7 bactenophage promoter in conjunction with the lac operator regulatory element Recombinant vectors are transformed into suitable bactenal hosts, e g , BL21(DE3) Antibiotic resistant bacteria express SYNT upon induction with isopropyl beta-D- thiogalactopyranoside (IPTG) Expression of SYNT in eukaryotic cells is achieved by infecting insect or mammalian cell lines with recombinant Autographica californica nuclear polyhedrosis virus (AcMNPV), commonly known as baculovirus. The nonessential polyhedrin gene of baculovirus is replaced with cDNA encoding SYNT 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 Spodoptera frugiperda (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, SYNT 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 iaponicum. 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 SYNT 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, supra, ch. 10 and 16). Purified SYNT obtained by these methods can be used directly in the assays shown in Examples X and XIV.
X. Demonstration of SYNT Activity
An SYNT activity assay measures aminoacylation of tRNA in the presence of a radiolabeled substrate. A cell-free extract depleted of endogenous aminoacyl-tRNA synthetase is prepared from Escherichia coli. SYNT, either biochemically purified or recombinantly produced, is added to the cell free extract. The cell-free extract is incubated with [14C]-labeled amino acid under conditions favorable for translation. Incorporation of the [14C]-labeled amino acid into acid-precipitable aminoacyl-tRNA is measured using a radioisotope counter. The amount of the [14C]-labeled amino acid incorporated into aminoacyl tRNA is proportional to the amount of SYNT activity. (See, for example, Ibba, M . et al . ( 1997) Science 278 : 11 19- 1 122) . Alternatively, SYNT activity may be assayed as follows. SYNT, or biologically active fragments thereof, are labeled with i25I Bolton-Hunter reagent. (See, e.g., Bolton et al. ( 1973) Biochem. J. 133:529.) Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled SYNT, washed, and any wells with labeled SYNT complex are assayed. Data obtained using different concentrations of SYNT are used to calculate values for the number, affinity, and association of SYNT with the candidate molecules.
XI. Functional Assays SYNT function is assessed by expressing the sequences encoding SYNT at physiologically elevated levels in mammalian cell culture systems cDNA is subcloned into a mammalian expression vector containing a strong promoter that dπves high levels of cDNA expression Vectors of choice include pCMV SPORT plasmid (Life Technologies) and pCR3 1 plasmid (Invitrogen), both of which contain the cytomegalovirus promoter 5-10 μg 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 μg 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 granulaπty as measured by forward light scatter and 90 degree side light scatter, down-regulation of DNA synthesis as measured by decrease in bromodeoxyuπdine 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 SYNT on gene expression can be assessed using highly purified populations of cells transfected with sequences encoding SYNT 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 SYNT and other genes of interest can be analyzed by northern analysis or microarray techniques XII. Production of SYNT Specific Antibodies
SYNT substantially punfied using polyacrylamide gel electrophoresis (PAGE, see, e g , Hamngton, M G ( 1990) Methods Enzymol 182 488-495), or other punfication techniques, is used to immunize rabbits and to produce antibodies using standard protocols
Alternatively, the SYNT amino acid sequence is analyzed using LASERGENE software (DNASTAR) to determine regions of high immunogenicity, and a corresponding ohgopeptide is synthesized and used to raise antibodies by means known to those of skill in the art Methods for selection of appropnate 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 1 1 )
Typically, ohgopeptides of about 15 residues in length are synthesized using an ABI 431 A peptide synthesizer (PE Biosystems) using FMOC chemistry and coupled to KLH (Sigma-Aldπch, 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 ohgopeptide- KLH complex in complete Freund's adjuvant Resulting antisera are tested for antipeptide and anti- SYNT activity by, for example, binding the peptide or SYNT to a substrate, blocking with 1 % BSA, reacting with rabbit antisera, washing, and reacting with radio-iodmated goat anti-rabbit IgG XIII. Purification of Naturally Occurring SYNT Using Specific Antibodies
Naturally occurring or recombinant SYNT is substantially purified by immunoaffinity chromatography using antibodies specific for SYNT An immunoaffinity column is constructed by covalently coupling anti-SYNT 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 SYNT are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of SYNT (e g , high ionic strength buffers in the presence of detergent) The column is eluted under conditions that disrupt antibody/SYNT binding (e g , a buffer of pH 2 to pH 3, or a high concentration of a chaotrope, such as urea or thiocyanate ion), and SYNT is collected
XIV. Identification of Molecules Which Interact with SYNT
SYNT, or biologically active fragments thereof, are labeled with I25I 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 SYNT, washed, and any wells with labeled SYNT complex are assayed Data obtained using different concentrations of SYNT are used to calculate values for the number, affinity, and association of SYNT with the candidate molecules
Alternatively, molecules interacting with SYNT are analyzed using the yeast two-hybnd system as descπbed in Fields, S and O Song (1989, Nature 340 245-246), or using commercially available kits based on the two-hybnd system, such as the MATCHMAKER system (Clontech)
SYNT may also be used in the PATHCALLING process (CuraGen Corp , New Haven CT) which employs the yeast two-hybnd system in a high-throughput manner to determine all interactions between the proteins encoded by two large hbranes of genes (Nandabalan, K et al (2000) U S Patent No 6,057, 101 )
Vanous modifications and variations of the descnbed 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 descnbed m 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
Table 1
O
Figure imgf000070_0001
Table 1 (cont.)
Figure imgf000071_0001
Table 2
Figure imgf000072_0001
Table 2 (cont.)
Figure imgf000073_0001
Table 2 (cont.)
Figure imgf000074_0001
Table 2 (cont.)
Figure imgf000075_0001
Table 2 (cont.)
Figure imgf000076_0001
<
Table 3
Figure imgf000077_0001
Table 3 (cont.)
Figure imgf000078_0001
Table 4
-j oo
Figure imgf000079_0001
Table 4 (cont.)
--0
SO
Figure imgf000080_0001
Table 4 (cont.)
oo
Figure imgf000081_0001
o
Table 5
Program Description Reference Parameter Threshold
ABI FACTURA A program that removes vector sequences and PE Biosystems, Foster City, CA. masks ambiguous bases in nucleic acid sequences.
ABI/PARACEL FDF A Fast Data Finder useful in comparing and PE Biosystems, Foster City, CA; Mismatch <50% annotating amino acid or nucleic acid sequences. Paracel Inc., Pasadena, CA.
ABI AutoAssembler A program that assembles nucleic acid sequences. PE Biosystems, Foster City, CA.
BLAST A Basic Local Alignment Search Tool useful in Altschul, S.F. et al. (1990) J. Mol. Biol. ESTs: Probability value= 1.0E-8 sequence similarity search for amino acid and 215:403-410; Altschul, S.F. et al. (1997) or less nucleic acid sequences. BLAST includes five Nucleic Acids Res. 25:3389-3402. Full Length sequences: Probabilit functions: blastp, blastn, blastx, tblastn, and tblastx. value= l .OE-10 or less
FASTA A Pearson and Lip an algorithm that searches for Pearson, W.R. and D.J. Lipman (1988) Proc. ESTs: fasta E value= 1.06E-6 similarity between a query sequence and a group of Natl. Acad Sci. USA 85:2444-2448; Pearson, Assembled ESTs: fasta Identity= sequences of the same type. FASTA comprises as W.R. (1990) Methods Enzymol. 183:63-98; 95% or greater and least five functions: fasta, tfasta, fastx, tfastx, and and Smith, T.F. and M.S. Waterman (1981 ) Match length=200 bases or greate ssearch. Adv. Appl. Math. 2:482-489. fastx E value=l .0E-8 or less
Full Length sequences: fastx score=100 or greater
BLIMPS A BLocks IMProved Searcher that matches a Henikoff, S. and J.G. Henikoff (1991) Nucleic Score=1000 or greater; sequence against those in BLOCKS, PRINTS, Acids Res. 19:6565-6572; Henikoff, J.G. and Ratio of Score/Strength = 0.75 or DOMO, PRODOM, and PFAM databases to search S. Henikoff (1996) Methods Enzymol. larger; and, if applicable, for gene families, sequence homology, and structural 266:88-105; and Attwood, T.K. et al. (1997) J. Probability value= 1.0E-3 or less fingerprint regions. Chem. Inf. Comput. Sci. 37:417-424.
HMMER An algorithm for searching a query sequence against Krogh, A. et al. (1994) J. Mol. Biol. Score=10-50 bits for PFAM hits, hidden Markov model (HMM)-based databases of 235: 1501 -1531 ; Sonnhammer, E.L.L. et al. depending on individual protein protein family consensus sequences, such as PFAM. (1988) Nucleic Acids Res. 26:320-322. families
Table 5 (cont.)
Program Description Reference Parameter Threshold
ProfileScan An algorithm that searches for structural and Gribskov, M. et al. (1988) CABIOS 4:61-66; Normalized quality score≥GCG- sequence motifs in protein sequences that match Gribskov, M. et al. (1989) Methods Enzymol. specified "HIGH" value for that sequence patterns defined in Prosite. 183: 146-159; Bairoch, A. et al. (1997) particular Prosite motif. Nucleic Acids Res. 25:217-221. Generally, score=l .4-2.1.
Phred A base-calling algorithm that examines automated Ewing, B. et al. (1998) Genome Res. sequencer traces with high sensitivity and 8: 175-185; Ewing, B. and P. Green probability. (1998) Genome Res. 8: 186-194.
Phrap A Phils Revised Assembly Program including Smith, T.F. and M.S. Waterman (1981) Adv. Score= 120 or greater; SWAT and CrossMatch, programs based on Appl. Math. 2:482-489; Smith, T.F. and M.S. Match length= 56 or greater efficient implementation of the Smith-Waterman Waterman (1981) J. Mol. Biol. 147: 195-197; algorithm, useful in searching sequence homology and Green, P., University of Washington, and assembling DNA sequences. Seattle, WA.
Consed A graphical tool for viewing and editing Phrap Gordon, D. et al. (1998) Genome assemblies. Res. 8: 195-202.
SPScan A weight matrix analysis program that scans protein Nielson, H. et al. (1997) Protein Engineering Score=3.5 or greater sequences for the presence of secretory signal 10: 1-6; Claverie, J.M. and S. Audic (1997) peptides. CABIOS 12:431-439.
Motifs A program that searches amino acid sequences for Bairoch, A. et al. (1997) Nucleic Acids Res. patterns that matched those defined in Prosite. 25:217-221 ; Wisconsin Package Program Manual, version 9, page M51 -59, Genetics Computer Group, Madison, WI.

Claims

What is claimed is:
1. An isolated polypeptide compnsing an amino acid sequence selected from the group consisting of: a) an amino acid sequence selected from the group consisting of SEQ ID NO.1 , SEQ ID
NO:2, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO: l l , SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO.14, b) a naturally occurring amino acid sequence having at least 90% sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO: l 1, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, c) a biologically active fragment of an amino acid sequence selected from the group consisting of SEQ ID NO: l , SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO: l 1 , SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and d) an immunogenic fragment of an amino acid sequence selected from the group consisting of SEQ ID NO.1, SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO: l 1, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14
2. An isolated polypeptide of claim 1 selected from the group consisting of SEQ ID NO: 1 , SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO: 1 1 , SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14.
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: 16, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ED NO:27, SEQ ID NO:28. SEQ ID NO.29
6. A recombinant polynucleotide compnsing 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 compnsing a recombinant polynucleotide of claim 6
9 A method for producing a polypeptide of claim 1 , the method compnsing a) cultunng a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide, and said recombinant polynucleotide compnses a promoter sequence operably linked to a polynucleotide encoding the polypeptide of claim 1 , and b) recoveπng the polypeptide so expressed.
10 An isolated antibody which specifically binds to a polypeptide of claim 1
1 1. An isolated polynucleotide compnsing a polynucleotide sequence selected from the group consisting of a) a polynucleotide sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO- 19, SEQ ID NO 20, SEQ ID NO:21 , SEQ ID NO.22, SEQ ID NO.23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO 26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, b) a naturally occumng polynucleotide sequence having at least 90% sequence identity to a polynucleotide sequence selected from the group consisting of SEQ ID NO.16, SEQ ID NO: 17, SEQ
ID NO: 19, SEQ ID NO:20, SEQ ED NO:21 , SEQ ID NO:22, SEQ ID NO.23, SEQ ID NO-24, SEQ ID NO.25, SEQ ID NO.26, SEQ ED NO.27, SEQ ID NO:28, SEQ ID NO:29, c) a polynucleotide sequence complementary to a), d) a polynucleotide sequence complementary to b), and e) an RNA equivalent of a)-d).
12 An isolated polynucleotide composing at least 60 contiguous nucleotides of a polynucleotide of claim 1 1.
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 compnsing a) hybndizing 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 hybndizes to said target polynucleotide, under conditions whereby a hybndization complex is formed between said probe and said target polynucleotide or fragments thereof, and b) detecting the presence or absence of said hybndization complex, and. optionally, if present, the amount thereof
14 A method of claim 13, wherein the probe compnses 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 1 1, 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 compnsing an effective amount of a polypeptide of claim 1 and a pharmaceutically acceptable excipient
17 A composition of claim 16, wherein the polypeptide compnses an amino acid sequence selected from the group consisting of SEQ ID NO 1 , SEQ ID NO 2, SEQ ID NO 4, SEQ ID NO 5, SEQ ID NO 6, SEQ ID NO 7, SEQ ID NO 8, SEQ ID NO 9, SEQ ID NO 10, SEQ ID NO 1 1, SEQ ID NO 12, SEQ ID NO 13, SEQ ID NO 14
18 A method for treating a disease or condition associated with decreased expression of functional SYNT, comprising admimstenng 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 compnsing 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 SYNT, compnsing admimstenng to a patient in need of such treatment a pharmaceutical composition of claim 20
22 A method for screening a compound for effectiveness as an antagonist of a polypeptide of claim 1 , the method composing a) exposing a sample compnsing 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 SYNT, comprising admi stenng 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 composing 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 compnsing 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 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 altenng expression of a target polynucleotide, wherein said target polynucleotide compnses a sequence of claim 5, the method compnsing a) exposing a sample compnsing the target polynucleotide to a compound, and b) detecting altered expression of the target polynucleotide
28 A method for assessing toxicity of a test compound, said method compnsing a) treating a biological sample containing nucleic acids with the test compound, b) hybndizing the nucleic acids of the treated biological sample with a probe comprising at least 20 contiguous nucleotides of a polynucleotide of claim 1 1 under conditions whereby a specific hybndization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide compnsing a polynucleotide sequence of a polynucleotide of claim 1 1 or fragment thereof, c) quantifying the amount of hybndization complex, and d) companng the amount of hybndization complex in the treated biological sample with the amount of hybndization complex in an untreated biological sample, wherein a difference in the amount of hybridization complex the treated biological sample is indicative of toxicity of the test compound
PCT/US2000/019980 1999-07-22 2000-07-20 Human synthetases WO2001007628A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU63647/00A AU6364700A (en) 1999-07-22 2000-07-20 Human synthetases
CA002380317A CA2380317A1 (en) 1999-07-22 2000-07-20 Human synthetases
JP2001512894A JP2003533967A (en) 1999-07-22 2000-07-20 Human synthetase
EP00950556A EP1200596A2 (en) 1999-07-22 2000-07-20 Human synthetases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US14499299P 1999-07-22 1999-07-22
US60/144,992 1999-07-22
US16885899P 1999-12-02 1999-12-02
US60/168,858 1999-12-02

Publications (2)

Publication Number Publication Date
WO2001007628A2 true WO2001007628A2 (en) 2001-02-01
WO2001007628A3 WO2001007628A3 (en) 2001-08-16

Family

ID=26842557

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/019980 WO2001007628A2 (en) 1999-07-22 2000-07-20 Human synthetases

Country Status (5)

Country Link
EP (1) EP1200596A2 (en)
JP (1) JP2003533967A (en)
AU (1) AU6364700A (en)
CA (1) CA2380317A1 (en)
WO (1) WO2001007628A2 (en)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001090324A2 (en) * 2000-05-19 2001-11-29 Millennium Pharmaceuticals, Inc. 32263, a putative human biotin transcarboxylase and uses thereof
WO2005014645A2 (en) * 2003-08-07 2005-02-17 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Pedf-r receptor and uses
WO2005118816A2 (en) * 2004-05-27 2005-12-15 Karl-Franzens-Universität Graz Protein exhibiting triglyceride hydrolase activity
WO2005115461A3 (en) * 2004-05-27 2006-05-04 Karl Franzens Uni Graz Pharmaceutical composition for modulating the activity of triglyceride hydrolase
EP2566515A2 (en) * 2010-05-03 2013-03-13 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of arginyl-trna synthetases
CN103096913A (en) * 2010-05-27 2013-05-08 Atyr医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutaminyl-tRNA synthetases
US8969301B2 (en) 2010-07-12 2015-03-03 Atyr Pharma Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of aspartyl-tRNA synthetases
US8980253B2 (en) 2010-04-26 2015-03-17 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-tRNA synthetase
US9029506B2 (en) 2010-08-25 2015-05-12 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of tyrosyl-tRNA synthetases
US9062301B2 (en) 2010-05-04 2015-06-23 Atyr Pharma, Inc. 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
US9320782B2 (en) 2010-04-28 2016-04-26 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of alanyl tRNA synthetases
US9580706B2 (en) 2010-04-27 2017-02-28 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of threonyl-tRNA synthetases
US9623093B2 (en) 2010-04-29 2017-04-18 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of asparaginyl tRNA synthetases
US9637730B2 (en) 2010-07-12 2017-05-02 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of histidyl-tRNA synthetases
US9688978B2 (en) 2011-12-29 2017-06-27 Atyr Pharma, Inc. Aspartyl-tRNA synthetase-Fc conjugates
US9687533B2 (en) 2010-05-14 2017-06-27 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-tRNA synthetases
US9714419B2 (en) 2011-08-09 2017-07-25 Atyr Pharma, Inc. PEGylated tyrosyl-tRNA synthetase polypeptides
US9790482B2 (en) 2010-05-17 2017-10-17 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of leucyl-tRNA synthetases
US9796972B2 (en) 2010-07-12 2017-10-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-tRNA synthetases
US9816084B2 (en) 2011-12-06 2017-11-14 Atyr Pharma, Inc. Aspartyl-tRNA synthetases
US9822353B2 (en) 2011-12-06 2017-11-21 Atyr Pharma, Inc. PEGylated aspartyl-tRNA synthetase polypeptides
US9896515B2 (en) 2010-04-27 2018-02-20 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl tRNA synthetases
US10093915B2 (en) 2013-03-15 2018-10-09 Atyr Pharma Inc. Histidyl-tRNA synthetase-Fc conjugates
US10179906B2 (en) 2010-05-03 2019-01-15 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-alpha-tRNA synthetases
US10189911B2 (en) 2010-04-29 2019-01-29 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Valyl-tRNA synthetases
US10563191B2 (en) 2010-10-06 2020-02-18 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related protein fragments of tryptophanyl tRNA synthetases
US11767520B2 (en) 2017-04-20 2023-09-26 Atyr Pharma, Inc. Compositions and methods for treating lung inflammation

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5265140B2 (en) * 2007-06-14 2013-08-14 学校法人日本医科大学 Ovarian cancer detection method and detection kit
EP2566496B1 (en) 2010-05-03 2018-02-28 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of methionyl-trna synthetases
EP2566516B1 (en) 2010-05-03 2019-07-03 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of seryl-trna synthetases
WO2011153277A2 (en) 2010-06-01 2011-12-08 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of lysyl-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

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997040174A1 (en) * 1996-04-22 1997-10-30 Leukosite, Inc. Mammalian hyaluronan synthases, nucleic acids, uses thereof
WO1998016648A2 (en) * 1996-10-15 1998-04-23 Isis Innovation Limited Isopenicillin n synthetase and deacetoxycephalosporin c synthetase enzymes and methods
US5759833A (en) * 1994-05-27 1998-06-02 Cubist Pharmaceuticals, Inc. Human isoleucyl-tRNA synthetase proteins, nucleic acids and tester strains comprising same

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU3395900A (en) * 1999-03-12 2000-10-04 Human Genome Sciences, Inc. Human lung cancer associated gene sequences and polypeptides

Patent Citations (3)

* 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
WO1997040174A1 (en) * 1996-04-22 1997-10-30 Leukosite, Inc. Mammalian hyaluronan synthases, nucleic acids, uses thereof
WO1998016648A2 (en) * 1996-10-15 1998-04-23 Isis Innovation Limited Isopenicillin n synthetase and deacetoxycephalosporin c synthetase enzymes and methods

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE EBI [Online] SEQUENCE AC D84223, 15 May 2000 (2000-05-15) H. MOTEGI ET AL.: "Cloning and sequence determination of a human cytoplasmic leucyl-tRNA synthetase gene." XP002152778 *
NAGASE T ET AL: "PREDICTION OF THE CODING SEQUENCES OF UNIDENTIFIED HUMAN GENES. XVITHE COMPLETE SEQUENCES OF 150 NEW CDNA CLONES FROM BRAIN WHICH CODEFOR LARGE PROTEINS IN VITRO" DNA RESEARCH,UNIVERSAL ACADEMY PRESS,JP, vol. 7, 2000, pages 65-73, XP000949814 ISSN: 1340-2838 *
See also references of EP1200596A2 *

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001090324A2 (en) * 2000-05-19 2001-11-29 Millennium Pharmaceuticals, Inc. 32263, a putative human biotin transcarboxylase and uses thereof
WO2001090324A3 (en) * 2000-05-19 2002-07-18 Millennium Pharm Inc 32263, a putative human biotin transcarboxylase and uses thereof
WO2005014645A2 (en) * 2003-08-07 2005-02-17 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Pedf-r receptor and uses
WO2005014645A3 (en) * 2003-08-07 2005-06-16 Us Gov Health & Human Serv Pedf-r receptor and uses
WO2005118816A2 (en) * 2004-05-27 2005-12-15 Karl-Franzens-Universität Graz Protein exhibiting triglyceride hydrolase activity
WO2005115461A3 (en) * 2004-05-27 2006-05-04 Karl Franzens Uni Graz Pharmaceutical composition for modulating the activity of triglyceride hydrolase
WO2005118816A3 (en) * 2004-05-27 2006-05-11 Karl Franzens Uni Graz Protein exhibiting triglyceride hydrolase activity
US8980253B2 (en) 2010-04-26 2015-03-17 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-tRNA synthetase
US10030077B2 (en) 2010-04-26 2018-07-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-tRNA synthetase
US10717786B2 (en) 2010-04-26 2020-07-21 aTye Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Cysteinyl-tRNA synthetase
US9540629B2 (en) 2010-04-26 2017-01-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Cysteinyl-tRNA synthetase
US10150958B2 (en) 2010-04-27 2018-12-11 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of threonyl-tRNA synthetases
US9580706B2 (en) 2010-04-27 2017-02-28 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of threonyl-tRNA synthetases
US9896515B2 (en) 2010-04-27 2018-02-20 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl tRNA synthetases
US10563192B2 (en) 2010-04-27 2020-02-18 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of threonyl-tRNA synthetases
US9320782B2 (en) 2010-04-28 2016-04-26 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of alanyl tRNA synthetases
US10189911B2 (en) 2010-04-29 2019-01-29 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Valyl-tRNA synthetases
US9623093B2 (en) 2010-04-29 2017-04-18 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of asparaginyl tRNA synthetases
US9068177B2 (en) 2010-04-29 2015-06-30 Atyr Pharma, Inc Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutaminyl-tRNA synthetases
US10179906B2 (en) 2010-05-03 2019-01-15 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-alpha-tRNA synthetases
AU2011248355B2 (en) * 2010-05-03 2017-01-19 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Arginyl-tRNA synthetases
EP2566515A4 (en) * 2010-05-03 2013-09-18 Atyr Pharma Inc Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of arginyl-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
EP2566515A2 (en) * 2010-05-03 2013-03-13 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of arginyl-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
US9062301B2 (en) 2010-05-04 2015-06-23 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutamyl-prolyl-tRNA synthetases
US10160814B2 (en) 2010-05-04 2018-12-25 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutamyl-prolyl-tRNA synthetases
US10220080B2 (en) 2010-05-14 2019-03-05 aTyr Pharam, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-tRNA synthetases
US9687533B2 (en) 2010-05-14 2017-06-27 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-tRNA synthetases
US9790482B2 (en) 2010-05-17 2017-10-17 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of leucyl-tRNA synthetases
US10179908B2 (en) 2010-05-17 2019-01-15 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of leucyl-tRNA synthetases
CN103096913A (en) * 2010-05-27 2013-05-08 Atyr医药公司 Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutaminyl-tRNA synthetases
US8969301B2 (en) 2010-07-12 2015-03-03 Atyr Pharma Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of aspartyl-tRNA synthetases
US10669533B2 (en) 2010-07-12 2020-06-02 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Histidyl-tRNA synthetases
US9315794B2 (en) 2010-07-12 2016-04-19 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of aspartyl-tRNA synthetases
US9637730B2 (en) 2010-07-12 2017-05-02 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of histidyl-tRNA synthetases
US9796972B2 (en) 2010-07-12 2017-10-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-tRNA synthetases
US10196629B2 (en) 2010-07-12 2019-02-05 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-tRNA synthetases
US10196628B2 (en) 2010-07-12 2019-02-05 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of histidyl-tRNA synthetases
US9029506B2 (en) 2010-08-25 2015-05-12 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of tyrosyl-tRNA synthetases
US10563191B2 (en) 2010-10-06 2020-02-18 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related protein fragments of tryptophanyl tRNA synthetases
US9714419B2 (en) 2011-08-09 2017-07-25 Atyr Pharma, Inc. PEGylated tyrosyl-tRNA synthetase polypeptides
US9816084B2 (en) 2011-12-06 2017-11-14 Atyr Pharma, Inc. Aspartyl-tRNA synthetases
US9822353B2 (en) 2011-12-06 2017-11-21 Atyr Pharma, Inc. PEGylated aspartyl-tRNA synthetase polypeptides
US9688978B2 (en) 2011-12-29 2017-06-27 Atyr Pharma, Inc. Aspartyl-tRNA synthetase-Fc conjugates
US10472618B2 (en) 2013-03-15 2019-11-12 Atyr Pharma, Inc. Histidyl-tRNA synthetase-Fc conjugates
US10093915B2 (en) 2013-03-15 2018-10-09 Atyr Pharma Inc. Histidyl-tRNA synthetase-Fc conjugates
US10711260B2 (en) 2013-03-15 2020-07-14 Atyr Pharma, Inc. Histidyl-tRNA synthetase-Fc conjugates
US11072787B2 (en) 2013-03-15 2021-07-27 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

Also Published As

Publication number Publication date
CA2380317A1 (en) 2001-02-01
AU6364700A (en) 2001-02-13
WO2001007628A3 (en) 2001-08-16
EP1200596A2 (en) 2002-05-02
JP2003533967A (en) 2003-11-18

Similar Documents

Publication Publication Date Title
WO2001007628A2 (en) Human synthetases
US20040018505A1 (en) Aminoacyl trna synthetases
CA2431493A1 (en) Aminoacyl trna synthetases
CA2408315A1 (en) Aminoacyl trna synthetases
US20050260708A1 (en) Proteases and protease inhibitors
EP1498480A2 (en) Human hydrolytic enzymes
EP1196577A2 (en) Cell cycle and proliferation proteins
US20050227277A1 (en) Apoptosis proteins
WO2001032888A2 (en) Human transferase molecules
EP1179065A2 (en) Full-length molecules expressed in human tissues
EP1244695A2 (en) Extracellular matrix and cell adhesion proteins as well as genes encoding them
EP1190050A2 (en) Human transcriptional regulator proteins
WO2001019860A2 (en) Proteins associated with cell differentiation
WO2001055358A2 (en) Phosphodiesterases
EP1206553A2 (en) Isomerase proteins
WO2002004611A2 (en) Aminoacyl trna synthetases
WO2001004308A1 (en) Human lim domain proteins
EP1124951A2 (en) Human sorting nexins
EP1477564A2 (en) Human synthetases
WO2001005969A2 (en) Electron transfer proteins
WO2001004305A2 (en) Human proteins involved in detoxification
WO2002031151A2 (en) Lipocalins
EP1203015A2 (en) Human chaperone proteins

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 10031905

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2380317

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2000950556

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2000950556

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 2000950556

Country of ref document: EP