EP1356079A2 - Modulierung des srebp-wegs mit hisrs als zielmolekül - Google Patents

Modulierung des srebp-wegs mit hisrs als zielmolekül

Info

Publication number
EP1356079A2
EP1356079A2 EP02713405A EP02713405A EP1356079A2 EP 1356079 A2 EP1356079 A2 EP 1356079A2 EP 02713405 A EP02713405 A EP 02713405A EP 02713405 A EP02713405 A EP 02713405A EP 1356079 A2 EP1356079 A2 EP 1356079A2
Authority
EP
European Patent Office
Prior art keywords
hisrs
assay
srebp
agent
assay system
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP02713405A
Other languages
English (en)
French (fr)
Other versions
EP1356079A4 (de
Inventor
Cynthia Seidel-Dugan
Lisa C. Kadyk
Felipa A. Mapa
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Exelixis Inc
Original Assignee
Exelixis 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 Exelixis Inc filed Critical Exelixis Inc
Publication of EP1356079A2 publication Critical patent/EP1356079A2/de
Publication of EP1356079A4 publication Critical patent/EP1356079A4/de
Withdrawn legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/92Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving lipids, e.g. cholesterol, lipoproteins, or their receptors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • 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
    • 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)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/25Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving enzymes not classifiable in groups C12Q1/26 - C12Q1/66
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • SREBP sterol regulatory element binding protein
  • SREBPs are also implicated in the process of fat cell differentiation and adipose cell gene expression, particularly as transcription factors that can promote adipogenesis in a dominant fashion (reviewed by Spiegelman et al, Cell (1996) 87:377-389). SREBP function is regulated by intracellular levels of sterols or polyunsaturated fatty acids (PUFAs) (Xu J. et al, J. Biol. Chem. (1999) 274:23577- 23583).
  • PUFAs polyunsaturated fatty acids
  • SREBPs are retained as membrane-bound protein precursors that are kept inactive by virtue of being attached to the nuclear envelope and endoplasmic reticulum (ER) and therefore, excluded from the nucleus.
  • An SREBP in its membrane-bound form has large N-terminal and C-terminal segments facing the cytoplasm and a short loop projecting into the lumen of the organelle.
  • the N-terminal domain is a transcription factor of the basic-helix-loop-helix-leucine zipper (bHLH-Zip) family, and contains an "acid blob" typical of many transcriptional activators (Brown and Goldstein, Cell (1997) 89:331-340).
  • the N-terminal acid blob is followed by a basic helix-loop-hehx/leucine zipper domain (bHLH-Zip) similar to those found in many other DNA-binding transc ⁇ ptional regulators
  • SIP site 1 protease
  • S2P site 2 protease
  • SCAP SREBP Cleavage Activating Piotein
  • model organisms such as Drosophila and C elegans
  • Drosophila and C elegans provide a powerful means to analyze biochemical processes that, due to significant evolutionary conservation of genes, pathways, and cellular processes, have direct relevance to more complex vertebrate organisms
  • Identification of novel functions of genes involved in particular pathways in such model organisms can directly contribute to the understanding of the correlative pathways in mammals and of methods of modulating them (see e g , Miklos GL and Rubin GM, Cell 1996, 86 521-529)
  • Drosophila and C elegans are not susceptible to human pathologies
  • various experimental models can mimic the pathological states
  • a correlation between the pathology model and the modified expression of a Drosophila or C elegans gene can identity the association of the human orthologue with the human disease
  • a genetic screen is performed in an invertebrate model organism displaying a mutant (generally visible or selectable) phenotype due to misexpression - generally reduced, enhanced or ectopic expression - of a known gene (the "genetic entry point") Additional genes are mutated in a random or targeted manner When an additional gene mutation changes the original mutant phenotype, this gene is identified as a modifier ' that directly or indirectly interacts with the genetic entry point and its associated pathway If the genetic entry point is an ortholog of a human gene associated with a human pathology, such as lipid metabolic disorders, the screen can identify modifier genes that are candidate targets for novel therapeutics
  • Tl 1G6 1 (GenBank Identifier LGI] 7507690)
  • HisRS human histidyl tRNA synthetase
  • HisRS histidyl tRNA synthetases
  • tRNA Hli histidine tRNA
  • HisRS proteins are homodimeric enzymes that have characteristic motifs shared by class II tRNA synthetases (also including Gly, Pro, Ser and Thr tRNA synthetases) These enzymes are conserved throughout evolution, in species ranging from bacte ⁇ a to humans (Amaar, Y G and D L Bailhe (1993) Nucl Acids Res 221 4344-4347)
  • HisRS is one of several aaRS that can synthesize the intracellular signalling molecule diadenosine tetraphosphate (Freist, W et al (1999) Biol Chem 380 623-646, isselev, et al (1998) FEBS Lett 427 157-163) This molecule is abundant in pancreatic
  • HisRS has been implicated in nutrient sensing and control of translation through regulation of p70 S6K ⁇ nase
  • S6K phosphorylates the 40S iibosomal protein S6 which results in the increased initiation of translation of ⁇ bosomal mRNAs
  • HisRS has been implicated in nutrient sensing and control of translation through regulation of p70 S6K ⁇ nase
  • S6K phosphorylates the 40S iibosomal protein S6 which results in the increased initiation of translation of ⁇ bosomal mRNAs
  • the mechanism of this feedback is not understood, but it appears that accumulation of deacylated tRNAs results in a signal which inhibits S6K activity SUMMARY OF THE IN
  • the invention provides a method of identifying candidate agents for modulating the SREBP pathway, lipid metabolism, and/or adipogenesis using an assay system compnsing a HisRS polypeptide or nucleic acid, contacting the assay system with a test agent undei conditions whereby, but for the presence of the test agent, the system provides a reference activity, and detecting a test agent-biased activity of the assay system, wherein a difference between the test agent-biased activity and the reference activity identifies the test agent as a candidate agent for modulating the SREBP pathway, lipid metabolism, and/or adipogenesis
  • Candidate test agents include small molecule modulators, antibodies, and nucleic acid modulators such as antisense ohgomers and PMOs, among others
  • the assay system comprises cultured cells or a non-human animal expressing HisRS, and the assay system detects an agent-biased change in the SREBP pathway, lipid metabolism, and/or adipogenesis
  • candidate HisRS-modulating agents are identified in cell- free or cell-based assays, and a second assay system that detects an agent-biased change in an activity associated with the SREBP pathway, lipid metabolism, and/or adipogenesis is used to confirm the SREBP pathway modulating activity of the candidate agent
  • the second assay detects an agent-biased change in an activity associated with SREBP pathway
  • Preferred second assay systems are carried out in cultured cells
  • the invention further provides a method of modulating the SREBP pathway in a mammalian cell comprising contacting the cell with an agent that specifically binds a HisRS polypeptide or nucleic acid
  • the agent is administered to a mammalian animal predetermined to have a pathology associated with the SREBP pathway
  • Preferred agents include small molecule modulators, nucleic acid modulators, or antibodies
  • wild-type worms were mutagenized with EMS, allowed to self-fertilize for two generations to generate worms homozygous for newly induced mutations.
  • the resulting homozygotes were placed on SREBP RNAi feeding plates.
  • C. elegans containing suppressor mutations were identified as having intestines that were more darkly pigmented that the pale intestines of wild type C. elegans on SREBP RNAi feeding plates.
  • the modifier gene's identity was discovered through genetic mapping, positional cloning and phenotypic rescue of mutants.
  • Tl 1G6.1 (GI 7507690), an ortholog of the human HisRS gene, as a suppressor of the C. elegans SREBP phenotype, and hence a member of the SREBP pathway.
  • HisRS genes i.e., nucleic acids and polypeptides
  • treatment involves decreasing signaling through the SREBP pathway in order to treat pathologies related to metabolic syndrome.
  • the invention provides in vitro and in vivo methods of assessing HisRS function, and methods of modulating (generally inhibiting or agonizing) HisRS activity, which are useful for further elucidating the SREBP pathway and for developing diagnostic and therapeutic modalities for pathologies associated with the SREBP pathway.
  • pathologies associated with the SREBP signaling pathway encompass pathologies where the SREBP pathway contributes to maintaining the healthy state, as well as pathologies whose course may be altered by modulation of the SREBP pathway.
  • cDNA Human HisRS nucleic acid (cDNA) and protein sequences are provided in SEQ ED NOs: 1 and 2, respectively.
  • HisRS polypeptide refers to a full-length HisRS protein or a fragment or derivative thereof that is “functionally active,” meaning that the HisRS protein derivative or fragment exhibits one or more functional activities associated with a full- length, wild-type HisRS protein.
  • a fragment or derivative may have antigenicity such that it can be used in immunoassays, for immunization, for generation of inhibitory antibodies, etc, as discussed further below.
  • a functionally active HisRS fragment or derivative displays one or more biological activities associated with HisRS proteins such as enzymatic activity, signaling activity, ability to bind natural cellular substrates, etc.
  • Preferred HisRS polypeptides display enzymatic activity.
  • a functionally active HisRS polypeptide is a HisRS derivative capable of rescuing defective endogenous HisRS activity, such as in cell based or animal assays; the rescuing derivative may be from the same or a different species.
  • the fragments preferably comprise a HisRS domain, such as a C- or N-terminal or catalytic domain, among others, and preferably comprise at least 10, preferably at least 20, more preferably at least 25, and most preferably at least 50 contiguous amino acids of a HisRS protein Functional domains can be identified using the PFAM program (Bateman A et al , 1999 Nucleic Acids Res 27 260- 262, website at pfam wustl edu)
  • HisRS nucleic acid refers to a DNA or RNA molecule that encodes a HisRS polypeptide
  • the HisRS polypeptide or nucleic acid or fragment thereof is from a human, but it can be an ortholog or derivative thereof with at least 70%, preferably with at least 80%, preferably 85%, still more preferably 90%, and most preferably at least 95% sequence identity with human HisRS
  • percent (%) sequence identity with respect to a specified subject sequence, or a specified portion thereof, is defined as the percentage of nucleotides or ammo acids in the candidate derivative sequence identical with the nucleotides or amino acids in the subject sequence (or specified portion thereof), after aligning the sequences and introducing gaps, if necessary to achieve the maximum percent sequence identity, as generated by the program WU-BLAST-2 0al9 (Altschul er / , J Mol Biol (1997) 215 403-410, http //blast wustl edu/blast/README
  • Derivative nucleic acid molecules of the subject nucleic acid molecules include sequences that hybridize to the nucleic acid sequence of SEQ ID NO 1.
  • the stringency of hybndization can be controlled by temperature, ionic strength, pH, and the presence of denatui ing agents such as foimamide during hybndization and washing Conditions loutinely used are set out in readily available procedure texts (e g , Current Protocol in Moleculai Biology, Vol 1 , Chap 2 10, John Wiley & Sons, Publishers (1994), Sambrook et al , Molecular Cloning, Cold Spring Harbor (1989)).
  • a nucleic acid molecule of the invention is capable of hybridizing to a nucleic acid molecule containing the nucleotide sequence of SEQ ID NO: l under stringent hybridization conditions that comprise' prehyb ⁇ dization of filters containing nucleic acid for 8 hours to overnight at 65° C in a solution comprising 6X single strength citrate (SSC) (IX SSC is 0 15 M NaCl, 0 015 M Na citrate, pH 7.0), 5X Denhardt's solution, 0 05% sodium pyrophosphate and 100 ⁇ g/ml herring sperm DNA; hybridization for 18-20 hours at 65° C in a solution containing 6X SSC, IX Denhardt's solution, 100 ⁇ g/ml yeast tRNA and 0 05% sodium pyrophosphate, and washing of filters at 65° C for 1 h in a solution containing 0.2X SSC and 0.1% SDS (sodium dodecyl sulfate).
  • SSC
  • moderately stringent hybridization conditions comprise: pretreatment of filters containing nucleic acid for 6 h at 40° C in a solution containing 35% formamide, 5X SSC, 50 mM T ⁇ s-HCl (pH 7.5), 5 mM EDTA, 0.1% PVP, 0.1% Ficoll, 1% BSA, and 500 ⁇ g/ml denatured salmon sperm DNA; hybridization for 18-20 h at 40° C in a solution containing 35% formamide, 5X SSC, 50 mM T ⁇ s-HCl (pH 7.5), 5 mM EDTA, 0.02% PVP, 002% Ficoll, 0.2% BSA, 100 ⁇ g/ml salmon sperm DNA, and 10% (wt/vol) dextran sulfate, followed by washing twice for 1 hour at 55° C in a solution containing 2X SSC and 0 1% SDS.
  • low stringency conditions can be used that comprise: incubation for 8 hours to overnight at 37° C in a solution comprising 20% formamide, 5 x SSC, 50 mM sodium phosphate (pH 7 6), 5X Denhardt's solution, 10% dextran sulfate, and 20 ⁇ g/ml denatured sheared salmon sperm DNA; hybridization in the same buffer for 18 to 20 hours, and washing of filters in 1 x SSC at about 37° C for 1 hour.
  • the HisRS is an ortholog of human HisRS.
  • Methods of identifying the human orthologs of these genes are known in the art. Normally, orthologs in difteient species retain the same function, due to presence of one or more protein motifs and/or 3-d ⁇ mens ⁇ onal structures. Orthologs are generally identified by sequence homology analysis, such as BLAST analysis, usually using protein bait sequences.
  • Sequences are assigned as a potential ortholog if the best hit sequence from the forward BLAST result retrieves the original query sequence in the reverse BLAST (Huynen MA and Bork P, Proc Natl Acad Sci (1998) 95 5849-5856; Huynen MA et al, Genome Research (2000) 10 1204-1210)
  • Programs for multiple sequence alignment such as CLUSTAL (Thompson JD et al, 1994, Nucleic Acids Res 22 4673-4680) may be used to highlight conserved regions and/or residues of orthologous proteins and to generate phylogenetic trees
  • orthologous sequences from two species generally appear closest on the tree with respect to all other sequences from these two species
  • Structural thieading or other analysis of protein folding e g., using software by PioCeryon, Biosciences, Salzburg, Austria
  • HisRS nucleic acids and polypeptides are useful for identifying and testing agents that modulate HisRS function and for other applications related to the involvement of HisRS in the SREBP pathway
  • HisRS nucleic acids may be obtained using any available method For instance, techniques for isolating cDNA or genomic DNA sequences of interest by screening DNA libraries or by using polymerase chain reaction (PCR) are well known in the art
  • HisRS polypeptides In geneial, the intended use for the polypeptide will dictate the particulars of expression, production, and purification methods For instance, production of polypeptides for use in screening for modulating agents may require methods that preserve specific biological activities of these proteins, whereas production of polypeptides for antibody generation may lequire structuial integrity of particular epitopes Expression of polypeptides to be purified for screening or antibody production may require the addition of specific tags (t e , generation of fusion proteins) Overexpression of a HisRS polypeptide for cell-based assays used to assess HisRS function, such as involvement in lipid metabolism, may lequire expression in eukaryotic cell lines capable of these cellular activities Techniques foi the expression, production, and purification of proteins are well known in the art, any suitable means therefor may be used (e g , Higgins SJ and Hames BD (eds ) Protein Expression A Practical Approach, Oxford University Press Inc , New York 1999, Stanbury PF
  • the nucleotide sequence encoding a HisRS polypeptide can be inserted into any appropriate vector for expression of the inserted protein-coding sequence
  • the necessary transc ⁇ ptional and translational signals can derive from the native HisRS gene and/or its flanking regions or can be heterologous
  • a variety of host-vector expression systems may be utilized, such as mammalian cell systems infected with virus (e g vaccinia virus, adenovirus, etc ), insect cell systems infected with virus (e g baculovirus), microorganisms such as yeast containing yeast vectors, or bacte ⁇ a tiansfoimed with bacte ⁇ ophage, plasmid, or cosmid DNA
  • a host cell strain that modulates the expiession of, modifies, and/oi specifically processes the gene product may be used
  • the HisRS polypeptide may be optionally expressed as a fusion or chime ⁇ c product, joined via a peptide bond to a heterologous
  • a HisRS polypeptide can be isolated and purified using standard methods (e g ion exchange, affinity, and gel exclusion chromatography, cent ⁇ fugation, differential solubility, electrophoresis)
  • native HisRS proteins can be purified from natural sources, by standard methods (e g immunoaffinity purification)
  • a protein Once a protein is obtained, it may be quantified and its activity measured by appropriate methods, such as immunoassay, bioassay, or other measurements of physical properties, such as crystallography
  • the methods of this invention may also use cells that have been engineered for altered expression (mis-expression) of HisRS or other genes associated with the SREBP pathway, lipid metabolism, and/or adipogenesis
  • mis-expression encompasses ectopic expression, over-expression, under-expression, and non-expression (e ⁇ > by gene knock-out oi blocking expiession that would otherwise normally occur)
  • the methods of this invention may use non-human animals that have been genetically modified to alter expression of HisRS and/or other genes known to be involved in adipogenesis, lipid metabolism, and/or the SREBP pathway
  • Preferred genetically modified animals are mammals, particularly mice or rats
  • Preferred non-mammalian species include Zebrafish, C elegans, and Drosophila
  • the altered HisRS or othei gene expiession results in a detectable phenotype, such as modified lipid profile as compaied to control animals having normal expression of the altered gene
  • the genetically modified animals can be used to further elucidate the SREBP pathway, in animal models of pathologies associate with adipogenesis, lipid metabolism, and/or the SREBP pathway, and for in vivo testing of candidate therapeutic agents, as described below
  • Preferred genetically modified animals are transgemc, at least a portion of their cells hai bo ⁇ ng non-native nucleic acid that is present either as a stable genomic insertion oi as an extia-chi omosomal element, which is typically mosaic
  • Preferred transgemc animals have geim-hne insertions that are stably transmitted to all cells of progeny animals
  • Non-native nucleic acid is introduced into host animals by any expedient method. Methods of making transgenic non-human animals are well-known in the art (for mice see Brinster et al., Proc. Nat. Acad. Sci. USA 1985, 82:4438-42; U.S. Pat. Nos.
  • Homozygous or heterozygous alterations in the genomes of transgenic animals may result in mis-expression of native genes, including ectopic expression, over- expression (e.g. by multiple gene copies), under-expression, and non-expression (e.g. by gene knock-out or blocking expression that would otherwise normally occur).
  • a "knock-out" animal is generated, typically using homologous recombination, in which an alteration in an endogenous gene causes a decrease in that gene's function, preferably such that gene expression is undetectable or insignificant.
  • the invention provides methods to identify agents that interact with and/or modulate the function of HisRS and/or the SREBP pathway. Such agents are useful in a variety of diagnostic and therapeutic applications associated with the SREBP pathway, as well as in further analysis of the HisRS protein and its contribution to the SREBP pathway. Accordingly, the invention also provides methods for modulating the SREBP pathway comprising the step of specifically modulating HisRS activity by administering a HisRS-interacting or -modulating agent.
  • HisRS-modulating agents inhibit or enhance HisRS activity or otherwise affect normal HisRS function, including transcription, protein expiession, protein localization, and cellular or extra-cellular activity.
  • the candidate SREBP pathway- modulating agent specifically modulates the function of the HisRS.
  • the phrases "specific modulating agent”, “specifically modulates”, etc., are used herein to refer to modulating agents that directly bind to the HisRS polypeptide or nucleic acid, and preferably inhibit, enhance, or otherwise alter the function of the HisRS.
  • the term also encompasses modulating agents that alter the interaction of HisRS with a binding partner or substrate (e.g. by binding to a binding partner of a HisRS, or to a protein/binding partner complex, and inhibiting function).
  • Preferred HisRS-modulating agents include small molecule chemical agents, HisRS-interacting proteins, including antibodies and other biotherapeutics, and nucleic acid modulators, including antisense oligomers and RNA.
  • the modulating agents may be formulated in pharmaceutical compositions, for example, as compositions that may comprise other active ingredients, as in combination therapy, and/or suitable carriers or excipients. Techniques for formulation and administration of the compounds may be found in "Remington's Pharmaceutical Sciences” Mack Publishing Co., Easton, PA, 19 l edition.
  • Chemical agents referred to in the art as "small molecule” compounds are typically organic, non-peptide molecules, having a molecular weight less than 10,000, preferably less than 5,000, more preferably less than 1,000, and most preferably less than 500.
  • This class of modulators includes chemically synthesized molecules, for instance, compounds from combinatorial chemical libraries. Synthetic compounds may be rationally designed or identified based on known or inferred properties of the HisRS protein or may be identified by screening compound libraries. Alternative appropriate modulators of this class are natural products, particularly secondary metabolites from organisms such as plants or fungi, which can also be identified by screening compound libraries for HisRS-modulating activity. Methods for generating and obtaining compounds are well known in the art (Schreiber SL, Science (2000) 151 : 1964-1969; Radmann J and Gunther J, Science (2000) 151 : 1947-1948).
  • Small molecule modulators identified from screening assays, as described below, can be used as lead compounds from which candidate clinical compounds may be designed, optimized, and synthesized. Such clinical compounds may have utility in treating pathologies associated with the SREBP pathway.
  • the activity of candidate small molecule modulating agents may be improved several-fold through iterative secondary functional validation, as further described below, structure determination, and candidate modulator modification and testing.
  • candidate clinical compounds are generated with specific regard to clinical and pharmacological properties.
  • the reagents may be derivatized and re-screened using in vitro and in vivo assays to optimize activity and minimize toxicity for pharmaceutical development. Protein Modulators
  • a HisRS-interacting protein may be endogenous, i e one that normally interacts genetically or biochemically with a HisRS, such as a member of the HisRS pathway that modulates HisRS expression, localization, and/or activity HisRS-modulators include dominant negative forms of HisRS-interacting proteins and of HisRS proteins themselves Yeast two-hybrid and vanant screens offer preferred methods for identifying endogenous HisRS-inteiacting (Finley, R L et al (1996) in DNA Cloning-Expression Systems A Practical Approach, eds Glover D & Hames B D (Oxford University Press, Oxford, England), pp 169-203, Fashema SF et al , Gene (2000) 250 1-14, Drees BL Curr Opin Chem Biol (1999) 3 64-70, Vidal M and Legrain P Nucleic Acids Res (1999) 27 919-29, and U S Pat No 5,928,868) Mass spectrometry offers alternative preferred methods for the elucidation
  • the HisRS-interacting protein is an antibody Antibodies that specifically bind HisRS polypeptides can be generated using known methods
  • the antibody is specific to a mammalian HisRS polypeptide, and moie pieferably, a human HisRS Antibodies may be polyclonal, monoclonal (mAbs), humanized oi chimeiic antibodies, single chain antibodies, Fab fragments, F(ab') sub 2 fiagments, fragments produced by a FAb expression library, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above Monoclonal antibodies with affinities of 10 8 M ' preferably 10 9 M ' to 10 10 M ', or stronger can be made by standard procedures as described (Harlow E and Lane D, 1988, Antibodies A Laboratory Manual, CSH Laboratory Press, (Harlow E and Lane D, 1999 Using Antibodies 1 A Laboiatoiy Manual, CSH Laboratory Press, Goding (1986) Monoclonal antibodies with affin
  • the subject polypeptides are covalently coupled to the keyhole limpet hemocyanin (KLH) camei , and the conjugate is emulsified in Freund's complete adjuvant, which enhances the immune lesponse
  • KLH keyhole limpet hemocyanin
  • An appiop ⁇ ate immune system such as a laboratory rabbit or mouse is immunized according to conventional protocols
  • Chimeiic antibodies specific to HisRS polypeptides can be made that contain diffeient portions from different animal species
  • a human immunoglobulin constant region may be linked to a variable region of a murine mAb, such that the antibody derives its biological activity from the human antibody, and its binding specificity from the murine fragment.
  • Chimeric antibodies are produced by splicing together genes that encode the appropriate regions from each species (Morrison et al., Proc. Natl. Acad. Sci. (1984) 81 :6851-6855; Neuberger et al., Nature (1984) 312:604-608; Takeda et al., Nature (1985) 31 :452-454).
  • Humanized antibodies which are a form of chimeric antibodies, can be generated by grafting complementary-determining regions (CDRs) (Carlos, T. M., J. M. Harlan. 1994. Blood 84:2068-2101) of mouse antibodies into a background of human framework regions and constant regions by recombinant DNA technology (Riechmann LM, et al., 1988 Nature 323: 323-327). Humanized antibodies contain -10% murine sequences and -90% human sequences, and thus further reduce or eliminate immunogenicity, while retaining the antibody specificities (Co MS, and Queen C. 1991 Nature 351 : 501-501 ; Morrison SL. 1992 Ann. Rev. Immun. 10:239-265). Humanized antibodies and methods of their production are well-known in the art (US PAT NO: 5,530,101; US PAT NO:5,585,089; US PAT NO:5,693J62, and US PAT NO:6, 180,370).
  • CDRs complementary-determining regions
  • HisRS-specific single chain antibodies which are recombinant, single chain polypeptides formed by linking the heavy and light chain fragments of the Fv regions via an amino acid bridge, can be produced (U.S. Pat. No. 4,946,778; Bird, Science (1988) 242:423-426; Huston et al., Proc. Natl. Acad. Sci. USA (1988) 85:5879-5883; and Ward et al., Nature (1989) 334:544-546).
  • T-cell antigen receptors are included within the scope of antibody modulators (Harlow and Lane, 1988, supra).
  • polypeptides and antibodies of the present invention may be used with or without modification. Frequently, the polypeptides and antibodies will be labeled by joining, either covalently or non-covalently, a substance that provides for a detectable signal, or that is toxic to cells that express the targeted protein (Menard S, et al., Int J. Biol Markers (1989) 4: 131-134).
  • labels and conjugation techniques are known and are reported extensively in both the scientific and patent literature. Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, fluorescent emitting lanthanide metals, chemiluminescent moieties, bioluminescent moieties, magnetic particles, and the like (U.S.
  • the antibodies of the subject invention are typically administered parenterally, when possible at the target site, or intravenously
  • the therapeutically effective dose and dosage regimen is determined by clinical studies Typically, the amount of antibody administered is in the range of about 0 1 mg/kg -to about 10 mg/kg of patient weight
  • the antibodies are formulated in a unit dosage injectable form (e g , solution, suspension, emulsion) in association with a phaimaceutically acceptable vehicle
  • a unit dosage injectable form e g , solution, suspension, emulsion
  • a phaimaceutically acceptable vehicle Such vehicles are inherently nontoxic and non-therapeutic Examples are water, saline, Ringer's solution, dextrose solution and 5% human serum albumin
  • Nonaqueous vehicles such as fixed oils, ethyl oleate, or liposome carriers may also be used
  • the vehicle may contain minor amounts of additives, such as buffers and preservatives, which enhance isotonicity and chemical stability or otherwise enhance therapeutic
  • HisRS-modulating agents comprise nucleic acid molecules, such as antisense oligomers or double stranded RNA (dsRNA), which generally inhibit HisRS activity
  • the antisense oligomer interferes with the function of HisRS nucleic acids, such as DNA leplication, transcription, HisRS RNA translocation, translation of protein from the HisRS RNA, RNA splicing, and any catalytic activity in which the HisRS RNA participates
  • the antisense oligomer is an ohgonucleotide that is sufficiently complementaiy to a HisRS mRNA to bind to and prevent translation from the HisRS mRNA, preferably by binding to the 5' untranslated region HisRS-specific antisense oligonucleotides preferably range from at least 6 to about 200 nucleotides In some embodiments the ohgonucleotide is preferably at least 10, 15, or 20 nucleotides in length In other embodiments, the ohgonucleotide is preferably less than 50, 40, or 30 nucleotides in length
  • the ohgonucleotide can be DNA or RNA, a chimeric mixture of DNA and RNA
  • the antisense oligomer is a phosphorothioate morpho no oligomer (PMO)
  • PMOs are assembled from four different morphohno subunits, each of which containing one of four genetic bases (A, C, G, or T) linked to a six-membered morphohne ring Polymers of these subunits are joined by non-ionic phosphodiamidate inter-subunit linkages
  • Methods of producing and using PMOs and other antisense oligonucleotides are well known in the art (e g see W099/18193, Summerton J, and Weller D, Antisense Nucleic Acid Drug Dev 1997, 7 187-95, Probst JC, Methods 2000, 22 271-281, US PAT NO 5,325,033, US PAT NO 5,378,841)
  • Antisense oligomers are commonly used as research reagents, diagnostics, and therapeutics
  • antisense oligonucleotides which are able to specifically inhibit gene expression are often used to elucidate the function of particular genes (see, e g US PAT NO 6,165,790)
  • Antisense oligomers are also used, for example, to distinguish between functions of various members of a biological pathway
  • Antisense oligomers have been employed as therapeutic moieties in the treatment of disease states in animals and humans and have been demonstrated in numerous clinical trials to be safe and effective (Milhgan JF et al, 1993, J Med Chem 36 1923-1937, Tonkinson JL et al , 1996, Cancer Invest 14 54-65)
  • a HisRS-specific antisense oligomer is used in an assay to further elucidate the function of HisRS in the SREBP pathway
  • a HisRS-specific antisense oligomer is used as
  • RNAi is the process of sequence-specific, post- transc ⁇ ptional gene silencing in animals and plants, initiated by double-stranded RNA (dsRNA) that is homologous in sequence to the silenced gene
  • dsRNA double-stranded RNA
  • Methods relating to the use of RNAi to silence genes in C elegans, Drosophila, plants, and mammals are known in the art (Fire A, et al , 1998 Nature 391 806-811 , Fire, A Trends Genet 15, 358-363 ( 1999) Sharp, P A RNA interference 2001 Genes Dev 15, 485-490 (2001), Hammond, S M , et al , Nature Rev Genet 2, 110-1119 (2001), Tuschl, T Chem Biochem 2, 239-245 (2001), Hamilton, A et al , Science 286, 950-952 (1999), Hammond, S M , et al , Nature 404, 293-296 (2000), Zamore
  • an "assay system” encompasses all the components required for performing and analyzing results of an assay that detects and/or measures a particular event or events
  • primary assays are used to identify or confirm a modulator's specific biochemical or molecular effect with respect to the HisRS nucleic acid or protein
  • secondaiy assays further assess the activity of a HisRS-modulating agent identified by a primary assay and may confirm that the modulating agent affects HisRS in a manner relevant to the SREBP pathway, lipid metabolism and/or adipogenesis
  • HisRS-modulators will be directly tested in a "secondary assay," without having been identified or confirmed in a "primary assay "
  • the assay system comprises contacting a suitable assay system comprising a HisRS polypeptide or nucleic acid with a candidate agent under conditions whereby, but for the presence of the agent, the system provides a reference activity, which is based on the particular molecular event the assay system detects
  • the method further comprises detecting the same type of activity in the presence of a candidate agent (the agent-biased activity of the system' )
  • a difference between the agent-biased activity and the reference activity indicates that the candidate agent modulates HisRS activity, and hence the SREBP pathway A difference, as used herein, is statistically significant
  • the assay systems generally include positive and/or negative controls, as are well known in the art
  • the type of modulator tested generally determines the type of primary assay
  • screening assays are used to identify candidate modulators
  • Screening assays may be cell-based or may use a cell-free system that recreates or retains the relevant biochemical reaction of the target protein (reviewed in Sittampalam GS et al , Curr Opin Chem Biol (1997) 1 384-91 and accompanying references)
  • the term "cell-based” refers to assays using live cells, dead cells, or a particular cellulai fraction, such as a membrane, endoplasmic reticulum, or mitochond ⁇ al fraction
  • the term “cell free” encompasses assays using substantially purified protein (either endogenous or recombinantly produced), partially purified cellular extracts, or crude cellular extracts
  • Screening assays may detect a variety of molecular events, including protein-DNA interactions, protein-protein interactions (e g , receptor- ligand binding), transcnptional activity (e g , using a reporter gene), enzymatic
  • screening assays use fluorescence technologies, including fluorescence polarization, time-resolved fluorescence, and fluorescence lesonance energy transfer These systems offer means to monitor protein-protein or DNA- protein interactions in which the intensity of the signal emitted from dye-labeled molecules depends upon their interactions with partner molecules (e g , Selvm PR, Nat Struct Biol (2000) 7:730-4; Fernandes PB, Curr Opin Chem Biol (1998) 2:597-603; Hertzberg RP and Pope AJ, Curr Opin Chem Biol (2000) 4:445-451).
  • fluorescence technologies including fluorescence polarization, time-resolved fluorescence, and fluorescence lesonance energy transfer
  • Suitable assay formats that may be adapted to screen for HisRS modulators are known in the art.
  • a primary function of HisRS is to attach histidine to histidine-tRNA.
  • suitable assays may detect aminoacylation activity of this enzyme (e.g., Yan W et al., 1996, Biochemistry 35:6559-68).
  • Preferred screening assays are high throughput or ultra high throughput and thus provide automated, cost-effective means of screening compound libraries for lead compounds (Fernandes PB, 1998, supra; Sundberg SA, Curr Opin Biotechnol 2000, 11 :47-53).
  • Cell-based screening assays usually require systems for recombinant expression of HisRS and any auxiliary proteins demanded by the particular assay.
  • Cell-free assays often use recombinantly produced purified or substantially purified proteins.
  • Appropriate methods for generating recombinant proteins produce sufficient quantities of proteins that retain their relevant biological activities and are of sufficient purity to optimize activity and assure assay reproducibility.
  • Yeast two-hybrid and variant screens, and mass spectrometry provide preferred methods for determining protein-protein interactions and elucidation of protein complexes.
  • the binding specificity of the interacting protein to the HisRS protein may be assayed by various known methods, including binding equilibrium constants (usually at least about 10 7 M " ', preferably at least about 10 8 M "1 , more preferably at least about 10 9 M "1 ), and immunogenic properties.
  • binding may be assayed by, respectively, substrate and ligand processing.
  • the screening assay may measure a candidate agent's ability to specifically bind to or modulate activity of a HisRS polypeptide, a fusion protein thereof, or to cells or membranes bearing the polypeptide or fusion protein.
  • the HisRS polypeptide can be full length or a fragment thereof that retains functional HisRS activity.
  • the HisRS polypeptide may be fused to another polypeptide, such as a peptide tag for detection or anchoring, or to another tag.
  • the HisRS polypeptide is preferably human HisRS, or is an ortholog or derivative thereof as described above.
  • the screening assay detects candidate agent-based modulation of HisRS interaction with a binding target, such as an endogenous or exogenous protein or other substrate that has HisRS -specific binding activity, and can be used to assess normal HisRS gene function.
  • a binding target such as an endogenous or exogenous protein or other substrate that has HisRS -specific binding activity
  • Certain screening assays may also be used to test antibody and nucleic acid modulators; for nucleic acid modulators, appropriate assay systems involve HisRS mRNA expression.
  • appropriate primary assays are binding assays that test the antibody's affinity to and specificity for the HisRS protein. Methods for testing antibody affinity and specificity are well known in the art (Harlow and Lane, 1988, 1999, supra).
  • the enzyme-linked immunosorbant assay (ELISA) is a preferred methods for detecting HisRS-specific antibodies; others include FACS assays, radioimmunoassays, and fluorescent assays.
  • primary assays may test the ability of the nucleic acid modulator to inhibit HisRS gene expression, preferably mRNA expression.
  • expression analysis comprises comparing HisRS expression in like populations of cells (e.g., two pools of cells that endogenously or recombinantly express HisRS) in the presence and absence of the nucleic acid modulator. Methods for analyzing mRNA and protein expression are well known in the art.
  • Northern blotting For instance, Northern blotting, slot blotting, ribonuclease protection, quantitative RT-PCR (e.g., using the TaqMan®, PE Applied Biosystems), or microarray analysis may be used to confirm that HisRS mRNA expression is reduced in cells treated with the nucleic acid modulator (e.g., Current Protocols in Molecular Biology (1994) Ausubel FM et al, eds., John Wiley & Sons, Inc., chapter 4; Freeman WM et al, Biotechniques (1999) 26: 112-125; Kallioniemi OP, Ann Med 2001, 33: 142-147; Blohm DH and Guiseppi-Elie, ACurr Opin Biotechnol 2001, 12:41-47).
  • the nucleic acid modulator e.g., Current Protocols in Molecular Biology (1994) Ausubel FM et al, eds., John Wiley & Sons, Inc., chapter 4
  • Protein expression may also be monitored. Proteins are most commonly detected with specific antibodies or antisera directed against either the HisRS protein or specific peptides. A variety of means including Western blotting, ELISA, or in situ detection, are available (Harlow E and Lane D, 1988 and 1999, supra).
  • Secondary assays may be used to further assess the activity of a HisRS-modulating agent identified by any of the above methods to confirm that the modulating agent affects HisRS in a manner relevant to the SREBP pathway.
  • HisRS-modulating agents encompass candidate clinical compounds or other agents derived from previously identified modulating agent.
  • Secondary assays can also be used to test the activity of a modulator on a particular genetic or biochemical pathway or to test the specificity of the modulator's interaction with HisRS.
  • Secondary assays generally compare like populations of cells or animals (e.g., two pools of cells or animals that endogenously or recombinantly express HisRS) in the presence and absence of the candidate modulator.
  • such assays test whether treatment of cells or animals with a candidate HisRS-modulating agent results in changes in the SREBP pathway, lipid metabolism, and/or adipogenesis, in comparison to untreated (or mock- or placebo-treated) cells or animals.
  • Certain assays use sensitized genetic backgrounds, used herein to describe cells or animals engineered for altered expression of genes in the SREBP or interacting pathways, or other pathways associated with lipid metabolism and/or adipogenesis
  • Cell based assays may use a variety of mammalian cell types capable of SREBP signaling, including HEK-293 cells, CHO cells, primary hepatocytes, or hepatocytic cell lines such as McA-RH7777 (DeBose-Boyd et al., 2001, PNAS 98: 1477-1482) or HEPG2 (Kotzka et al., 2000J. Lipid Res. 41:99-108). Cell based assays may detect endogenous SREBP pathway activity or may rely on recombinant expression of SREBP pathway components.
  • Cell based assays typically use culture condition that permit SREBP signaling, such as low cholesterol or low PUFA conditions, or glucose- or insulin- stimulation
  • SREBP signaling such as low cholesterol or low PUFA conditions
  • glucose- or insulin- stimulation are typically added to the cell media but may also be injected into cells or delivered by any other efficacious means.
  • SREBP pathway activity is assessed by measu ⁇ ng expression of SREBP transcriptional targets.
  • Many transcriptional targets are known (e.g., Osborne TF, 2001, J Biol Chem 275:32379-32382; Horton JD et al, 1998, J Clin Invest 101:2331- 2339, Shimano H et al, 1997, J Clin Invest 100:2115-2124; Shimomura I et al, 1999, J Biol Chem 274 30028-30032). Any available means for expression analysis, as pieviously described, may be used. Typically, mRNA expression is detected. In a preferred application, Taqman analysis is used to directly measure mRNA expression.
  • transgenic reporter construct comprising sequences encoding a reporter gene (such as luciferase, GFP or other fluorescent proteins, beta-galactosidase, etc.) under control of regulatory sequences (e.g., enhancer/promoter regions) of an SREBP transcriptional target gene.
  • a reporter gene such as luciferase, GFP or other fluorescent proteins, beta-galactosidase, etc.
  • regulatory sequences e.g., enhancer/promoter regions
  • assays monitor SREBP processing events, such as cleavage of the membrane-bound form of SREBP, or nuclear translocation or nuclear accumulation of the activated form of SREBP. These events can be monitored directly by monitoring levels of membrane bound and cleaved forms of the protein. Typically, cells are fractionated, and protein levels in nuclear and membrane fractions are measured using immunohistochemistry Alternatively, SREBP cleavage can be monitored indirectly using specific reporters for SREBP cleavage. In one example, a fusion construct comprising sequences encoding the signal peptide and soluble catalytic domain of alkaline phosphatase (AP) linked to the C-terminal (regulatory) domain of SREBP is introduced into cells.
  • AP alkaline phosphatase
  • SREBP cleavage is monitored as secretion of AP, which is detected using a standard alkaline phosphatase assay (Sakai J., et al., 1998, Mol. Cell 2:505-514).
  • a fusion construct is generated in which the transcriptional activator domain of SREBP is replaced with another transcriptional activator domain, such as yeast GAL4.
  • the substituted domain which is preferably from a different species, specifically activates transcription of a reporter gene under the control of responsive regulatory sequences, such as UAS if GAL4 is used.
  • assays measure candidate modulators' effects on the functional output of SREBP signaling, such as lipid accumulation and lipid metabolism.
  • lipid accumulation is measured by staining fixed cells with Oil Red O (Foretz et al., 1999, PNAS 96: 12737-12742).
  • lipid synthesis is monitored by measuring C14 acetate incorporation into either cholesterol or fatty acids (Pai, J-T et al., 1998, J. Biol. Chem. 273:26138-26148).
  • a variety of non-human animal models of lipid metabolic disorders may be used to test candidate HisRS modulators. Such models typically use genetically modified animals that have been engineered to mis-express (e.g., over-express or lack expression in) genes involved in lipid metabolism, adipogenesis, and/or the SREBP pathway. Additionally, particular feeding conditions, and/or administration or certain biologically active compounds, may contribute to or create animal models of lipid and/or metabolic disorders. Assays generally required systemic delivery of the candidate modulators, such as by oral administration, injection (intravenous, subcutaneous, intraperitoneous), bolus administration, etc.
  • mis-express e.g., over-express or lack expression in
  • Assays generally required systemic delivery of the candidate modulators, such as by oral administration, injection (intravenous, subcutaneous, intraperitoneous), bolus administration, etc.
  • assays use mouse models of diabetes and/or insulin resistance.
  • Mice carrying knockouts of genes in the leptin pathway such as ob (leptin) or db (leptin receptor), or the insulin signaling pathway, such as the insulin receptor (InR) or insulin receptor substrate (IRS)
  • the insulin receptor InR
  • insulin receptor substrate insulin receptor substrate
  • Certain susceptible wild type mice such as C57BL/6, exhibit similar symptoms when fed a high fat diet (Linton and Fazio, 2001, Current Opinion in Lipidology 12:489-495). Accordingly, appropriate assays using these models test whether administration of a candidate modulator alters, preferably decreases lipid accumulation in the liver. Lipid levels in plasma and adipose tissue may also be tested.
  • SREBP pathway activity may be tested by examining changes in the transcription of SREBP target genes in the liver.
  • target genes are associated with fatty acid metabolism and include acetyl CoA carboxylase, fatty acid synthase, ATP citrate lyase, glycerol -3-phosphate acyltransferase, glucose-6-phosphate dehydrogenase, malic enzyme, and stearoyl-CoA desaturase-l,etc (Shimomuia I et al, 1999, supra)
  • Other target genes are associated with cholesterol metabolism and include HMG-CoA synthase, HMG-CoA reductase, squalene synthase, hpoprotein hpase, the low-density lipoprotein receptor (LDLR), etc (Horton JD et al, 1998, supra)
  • assays use mouse models of hpoprotein biology and cardiovascular disease
  • mouse knockouts of apolipoprotein E (apoE) display elevated plasma cholesterol and spontaneous arterial lesions (Zhang SH, 1992, Science 258 468-471 )
  • Transgenic mice over-expressing cholesterol ester transfer protein (CETP) also display increased plasma lipid levels (specifically, very-low-density hpoprotein [VLDL] and low-density hpoprotein [LDL] cholesterol levels) and plaque formation in arteries (Maiotti KR et al , 1993, Nature 364 73-75)
  • Assays using these models may test whether administration of candidate modulators alters plasma lipid levels, such as by decreasing levels of the pro-atherogenic LDL and VLDL, increasing HDL, or by deci easing oveiall lipid (including trigycende) levels
  • histological analysis of ai tenal morphology and lesion formation (i e , lesion number and size) may indicate whether a
  • mice with knockouts in both leptin and LDL receptor genes display hypercholesterolemia, hypert ⁇ glyce ⁇ demia and arterial lesions and provide a model for the lelationship between impaired fuel metabolism, increased plasma remnant hpoproteins, diabetes, and atherosclerosis (Hasty AH et al, 2001, supra )
  • HisRS is implicated in the SREBP pathway
  • Any method for assessing HisRS expression in a sample may be used
  • Such methods may, for example, utilize reagents such as the HisRS oligonucleotides and antibodies directed against HisRS, as described above for (1) the detection of the presence of HisRS gene mutations, or the detection of either over- or under-expression of HisRS mRNA relative to the non-disordei state, (2) the detection of either an over- or an under-abundance of HisRS gene product lelative to the non-disorder state, and (3) the detection of perturbations or abnormalities in a biological pathway mediated by HisRS

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Diabetes (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Hematology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Obesity (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Endocrinology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Physics & Mathematics (AREA)
  • Child & Adolescent Psychology (AREA)
  • Emergency Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
EP02713405A 2001-01-12 2002-01-11 Modulierung des srebp-wegs mit hisrs als zielmolekül Withdrawn EP1356079A4 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US26156901P 2001-01-12 2001-01-12
US261569P 2001-01-12
PCT/US2002/001045 WO2002055663A2 (en) 2001-01-12 2002-01-11 Srebp pathway modulation through targeting hisrs

Publications (2)

Publication Number Publication Date
EP1356079A2 true EP1356079A2 (de) 2003-10-29
EP1356079A4 EP1356079A4 (de) 2005-10-19

Family

ID=22993896

Family Applications (1)

Application Number Title Priority Date Filing Date
EP02713405A Withdrawn EP1356079A4 (de) 2001-01-12 2002-01-11 Modulierung des srebp-wegs mit hisrs als zielmolekül

Country Status (4)

Country Link
EP (1) EP1356079A4 (de)
JP (1) JP2004524021A (de)
AU (1) AU2002245257A1 (de)
WO (1) WO2002055663A2 (de)

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5771142B2 (ja) 2008-06-11 2015-08-26 エータイアー ファーマ, インコーポレイテッド チロシル−tRNAシンテターゼポリペプチドの血小板新生活性
WO2009158649A1 (en) 2008-06-26 2009-12-30 Atyr Pharma, Inc. Compositions and methods comprising glycyl-trna synthetases having non-canonical biological activities
WO2010099477A2 (en) 2009-02-27 2010-09-02 Atyr Pharma, Inc. Polypeptide structural motifs associated with cell signaling activity
DK3255146T3 (da) 2009-03-16 2019-08-19 Pangu Biopharma Ltd Præparater og fremgangsmåder omfattende histidyl-tRNA-synthetasesplejsningsvarianter med ikke-kanoniske biologiske aktiviteter
US20100310576A1 (en) 2009-03-31 2010-12-09 Adams Ryan A COMPOSITIONS AND METHODS COMPRISING ASPARTYL-tRNA SYNTHETASES HAVING NON-CANONICAL BIOLOGICAL ACTIVITIES
US8828395B2 (en) 2009-12-11 2014-09-09 Atyr Pharma, Inc. Antibodies that bind tyrosyl-tRNA synthetases
CA2783731C (en) 2009-12-11 2018-03-27 Atyr Pharma, Inc. Aminoacyl trna synthetases for modulating inflammation
NZ603811A (en) 2010-07-12 2015-03-27 Atyr Pharma Inc Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of aspartyl-trna synthetases
KR20130102534A (ko) 2010-07-12 2013-09-17 에이티와이알 파마, 인코포레이티드 히스티딜­trna 합성효소의 단백질 단편에 관련된 치료적, 진단적, 및 항체 조성물의 혁신적 발견
EP2593125B1 (de) 2010-07-12 2017-11-01 aTyr Pharma, Inc. Innovative erkennung therapeutischer, diagnostischer und antikörperhaltiger zusammensetzungen in zusammenhang mit proteinfragmenten von glycyl-trna-synthetasen
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
CN103108650A (zh) 2010-08-25 2013-05-15 Atyr医药公司 与酪氨酰-tRNA合成酶的蛋白片段相关的治疗、诊断和抗体组合物的创新发现
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
CA2858613A1 (en) 2011-12-29 2013-08-08 Atyr Pharma, Inc. Aspartyl-trna synthetase-fc conjugates
KR20140123571A (ko) 2012-02-16 2014-10-22 에이티와이알 파마, 인코포레이티드 자가면역 및 염증성 질환의 치료를 위한 히스티딜­trna 신테타제
AU2014233436B2 (en) 2013-03-15 2019-12-05 Atyr Pharma, Inc. Histidyl-tRNA synthetase-Fc conjugates
WO2015027171A1 (en) * 2013-08-23 2015-02-26 Quintiles Transnational Corporation Methods for predicting toxicity in response to treatment with a drug by assessing activation of the sterol regulatory binding protein (srebp) pathway
JP2020517638A (ja) 2017-04-20 2020-06-18 エータイアー ファーマ, インコーポレイテッド 肺の炎症を治療するための組成物および方法

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000076308A1 (en) * 1999-06-14 2000-12-21 Exelixis, Inc. Animal models and methods for analysis of lipid metabolism and screening of pharmaceutical and pesticidal agents that modulate lipid metabolism

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000076308A1 (en) * 1999-06-14 2000-12-21 Exelixis, Inc. Animal models and methods for analysis of lipid metabolism and screening of pharmaceutical and pesticidal agents that modulate lipid metabolism

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of WO02055663A2 *

Also Published As

Publication number Publication date
EP1356079A4 (de) 2005-10-19
AU2002245257A1 (en) 2002-07-24
WO2002055663A2 (en) 2002-07-18
JP2004524021A (ja) 2004-08-12
WO2002055663A3 (en) 2002-09-19

Similar Documents

Publication Publication Date Title
US20040101879A1 (en) Srebp pathway modulation through targeting hisrs
WO2002055663A2 (en) Srebp pathway modulation through targeting hisrs
US6924117B2 (en) Human RRP sequences and methods of use
US20050170343A1 (en) Modulating insulin receptor signaling
US20070042371A1 (en) Mptens as modifers of the pten/igf pathway and methods of use
US20070274914A1 (en) Mptens as Modifiers of the Pten/Igf Pathway and Methods of Use
US20030138832A1 (en) Modulating insulin receptor signaling through targeting FACL
EP1860199B1 (de) Modulierte Signalisierung von Insulinrezeptoren
WO2003033656A2 (en) MSREBPs AS MODIFIERS OF THE SREBP PATHWAY AND METHODS OF USE
US20060088829A1 (en) Minrs as modifiers of insulin receptor signaling and methods of use
WO2003073063A2 (en) Modulating insulin receptor signaling through targeting helic1
WO2003066813A2 (en) MINRs AS MODIFIERS OF INSULIN RECEPTOR SIGNALING AND METHODS OF USE
US20030224406A1 (en) MBCATs as modifiers of the beta-catenin pathway and methods of use
WO2003066811A2 (en) Msrebps as modifiers of the srebp pathway and methods of use
US20050266406A1 (en) Maxs as modifiers of the axin pathway and methods of use
CA2518381A1 (en) Mracs as modifiers of the rac pathway and methods of use
CA2513668A1 (en) Dyrks as modifiers of the apc and axin pathways and methods of use
WO2003052068A2 (en) Mbcats as modifiers of the beta-catenin pathway and methods of use
WO2004048539A2 (en) Mapcaxs as modifiers of the apc and axin pathways and methods of use
WO2004083447A2 (en) Mbcats as modifiers of the beta-catenin pathway and methods of use
WO2004047761A2 (en) Mbcats as modifiers of the beta-catenin pathway and methods of use
WO2003074671A2 (en) Mbcats as modifiers of the beta-catenin pathway and methods of use
JP2007524356A (ja) RB経路のモディファイヤーとしてのFACLsおよび使用方法

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20030731

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

A4 Supplementary search report drawn up and despatched

Effective date: 20050905

RIC1 Information provided on ipc code assigned before grant

Ipc: 7A 61K 48/00 B

Ipc: 7A 61K 39/00 B

Ipc: 7C 12Q 1/25 B

Ipc: 7G 01N 33/92 A

17Q First examination report despatched

Effective date: 20051206

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20070731