WO2000009556A1 - Human lysophospholipase gene (cbfblh05) - Google Patents

Human lysophospholipase gene (cbfblh05) Download PDF

Info

Publication number
WO2000009556A1
WO2000009556A1 PCT/CN1998/000164 CN9800164W WO0009556A1 WO 2000009556 A1 WO2000009556 A1 WO 2000009556A1 CN 9800164 W CN9800164 W CN 9800164W WO 0009556 A1 WO0009556 A1 WO 0009556A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
identity
seq
subject
amino acid
Prior art date
Application number
PCT/CN1998/000164
Other languages
French (fr)
Inventor
Juan Zhou
Mao Mao
Min Ye
Qinghua Zhang
Original Assignee
Shanghai Second Medical University
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 Shanghai Second Medical University filed Critical Shanghai Second Medical University
Priority to PCT/CN1998/000164 priority Critical patent/WO2000009556A1/en
Priority to CN98810060.6A priority patent/CN1275131A/en
Publication of WO2000009556A1 publication Critical patent/WO2000009556A1/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
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/18Carboxylic ester hydrolases (3.1.1)
    • 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

Abstract

The CBFBLH05 polypeptides and polynucleotides and methods for producing such polypeptides by recombinant techniques are disclosed. Also disclosed are methods for utilising CBFBLH05 polypeptides and polynucleotides in therapy, and diagnostic assays for such.

Description

Human Lysophospholipase Gene (CBFBLH05)
Field of the Invention
This invention relates to newly identified polypeptides and polynucleotides encodmg such polypeptides, to their use in therapy and in identifying compounds which may be agonists, antagonists and /or inhibitors which are potentially useful m therapy, and to production of such polypeptides and polynucleotides
Background of the Invention The drug discovery process is currently undergoing a fundamental revolution as it embraces
'functional genomics', that is, high throughput genome- or gene-based biology This approach is rapidly superseding earlier approaches based on 'positional cloning' A phenotype, that is a biological function or genetic disease, would be identified and this would then be tracked back to the responsible gene, based on its genetic map position Functional genomics relies heavily on the various tools of bioinformatics to identify gene sequences of potential interest from the many molecular biology databases now available There is a continuing need to identify and characterise further genes and their related polypeptides/proteins, as targets for drug discovery
Summary of the Invention
The present invention relates to CBFBLH05, m particular CBFBLH05 polypeptides and CBFBLH05 polynucleotides, recombinant materials and methods for their production In another aspect, the invention relates to methods for using such polypeptides and polynucleotides, including the treatment of hver diseases, cancer, autoimmune diseases, and lαdney disorders, hereinafter referred to as "the Diseases", amongst others In a further aspect, the mvention relates to methods for identifying agonists and antagomsts/inhibitors using the mateπals provided by the mvention, and treatmg conditions associated with CBFBLH05 imbalance with the identified compounds In a still further aspect, the invention relates to diagnostic assays for detecting diseases associated with inappropriate CBFBLH05 activity or levels
Description of the Invention
In a first aspect, the present invention relates to CBFBLH05 polypeptides Such peptides include isolated polypeptides comprising an ammo acid sequence which has at least 70% identity , preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, most preferably at least 97-99% identity, to that of SEQ ID NO 2 over the entire length of SEQ ID NO 2 Such polypeptides mclude those comprising the ammo acid of SEQ ID NO 2
Further peptides of the present mvention mclude isolated polypeptides m which the amino acid sequence has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, most preferably at least 97-99% identity, to the ammo acid sequence of SEQ ID NO 2 over the entire length of SEQ ID NO 2 Such polypeptides mclude the polypeptide of SEQ ID NO 2
Further peptides of the present mvention mclude isolated polypeptides encoded by a polynucleotide compπsmg the sequence contained m SEQ ID NO 1
Polypeptides of the present invention are of interest because human lysophosphohpase is cloned from umbilical cord blood hematopoietic progenitor CD34+ cells, and it has two expression forms a short form and a long form In the CD34+ cell, the short form is predominant These properties are hereinafter referred to as "CBFBLH05 activity" or "CBFBLH05 polypeptide activity" or "biological activity of CBFBLH05" Also mcluded amongst these activities are antigemc and irnmunogemc activities of said CBFBLH05 polypeptides, in particular the antigemc and lmmunogenic activities of the polypeptide of SEQ ID NO 2 Preferably, a polypeptide of the present mvention exhibits at least one biological activity of CBFBLH05
The polypeptides of the present mvention may be m the form of the "mature" protem or may be a part of a larger protem such as a fusion protem It is often advantageous to include an additional ammo acid sequence which contains secretory or leader sequences, pro-sequences, sequences which aid in purification such as multiple histidine residues, or an additional sequence for stability during recombinant production
The present invention also includes mclude variants of the aforementioned polypeptides, that is polypeptides that vary from the referents by conservative ammo acid substitutions, whereby a residue is substituted by another with like characteristics Typical such substitutions are among Ala, Val, Leu and lie, among Ser and Thr, among the acidic residues Asp and Glu, among Asn and Gin, and among the basic residues Lys and Arg, or aromatic residues Phe and Tyr Particularly preferred are variants in which several, 5-10, 1-5, 1-3, 1-2 or 1 ammo acids are substituted, deleted, or added m any combination Polypeptides of the present invention can be prepared in any suitable manner Such polypeptides mclude isolated naturally occurring polypeptides, recombinantly produced polypeptides, synthetically produced polypeptides, or polypeptides produced by a combination of these methods Means for preparing such polypeptides are well understood in the art In a further aspect, the present mvention relates to CBFBLH05 polynucleotides Such polynucleotides mclude isolated polynucleotides comprising a nucleotide sequence encoding a polypeptide which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, to the ammo acid sequence of SEQ ID NO 2, over the entire length of SEQ ID NO 2 In this regard, polypeptides which have at least 97% identity are highly preferred, whilst those with at least 98-99% identity are more highly preferred, and those with at least 99% identity are most highly preferred Such polynucleotides mclude a polynucleotide comprising the nucleotide sequence contained in SEQ ID NO 1 encoding the polypeptide of SEQ ID NO 2 Further polynucleotides of the present mvention mclude isolated polynucleotides compπsmg a nucleotide sequence that has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, to a nucleotide sequence encoding a polypeptide of SEQ ID NO 2, over the entire codmg region In this regard, polynucleotides which have at least 97% identity are highly preferred, whilst those with at least 98-99% identity are more highly preferred, and those with at least 99% identity are most highly preferred
Further polynucleotides of the present mvention mclude isolated polynucleotides compπsmg a nucleotide sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, to SEQ ID NO 1 over the entire length of SEQ ID NO 1 In this regard, polynucleotides which have at least 97% identity are highly preferred, whilst those with at least 98-99% identity are more highly preferred, and those with at least 99% identity are most highly preferred Such polynucleotides mclude a polynucleotide compπsmg the polynucleotide of SEQ ID NO 1 as well as the polynucleotide of SEQ ID NO 1
The mvention also provides polynucleotides which are complementary to all the above descπbed polynucleotides The nucleotide sequence of SEQ ID NO 1 shows homology with Rat lysophospholφase gene(H Sugimoto, J Biol Chem ,1996, 271 (13) 7705-7711 ) The nucleotide sequence of SEQ ID NO 1 is a cDNA sequence and compπses a polypeptide encoding sequence (nucleotide 6 to 647) encoding a polypeptide of 214 ammo acids, the polypeptide of SEQ ID NO 2 The nucleotide sequence encoding the polypeptide of SEQ ID NO 2 may be identical to the polypeptide encoding sequence contamed in SEQ ID NO 1 or it may be a sequence other than the one contained m SEQ ID NO 1, which, as a result of the redundancy (degeneracy) of the genetic code, also encodes the polypeptide of SEQ ID NO 2 The polypeptide of the SEQ ID NO 2 is structurally related to other proteins of the family, having homology and/or structural sirralaπty with Rat lysophosphohpase (H Sugimoto. J Biol Chem .1996. 271 (13) 7705-7711 ) Preferred polypeptides and polynucleotides of the present invention are expected to have, inter alia, similar biological functions/properties to their homologous polypeptides and polynucleotides. Furthermore, preferred polypeptides and polynucleotides of the present invention have at least one CBFBLH05 activity. Polynucleotides of the present invention may be obtained, using standard cloning and screening techniques, from a cDNA library derived from rnRNA in cells of human umbilical cord blood, using the expressed sequence tag (EST) analysis (Adams, M.D., et al. Science (1991) 252:1651-1656; Adams, M.D. et al, Nature, (1992) 355:632-634; Adams, M.D., et al, Nature (1995) 377 Supp:3-174). Polynucleotides of the invention can also be obtained from natural sources such as genomic DNA libraries or can be synthesized using well known and commercially available techniques.
When polynucleotides of the present invention are used for the recombinant production of polypeptides of the present invention, the polynucleotide may include the coding sequence for the mature polypeptide, by itself; or the coding sequence for the mature polypeptide in reading frame with other coding sequences, such as those encoding a leader or secretory sequence, a pre-, or pro- or prepro- protein sequence, or other fusion peptide portions. For example, a marker sequence which facilitates purification of the fused polypeptide can be encoded. In certain preferred embodiments of this aspect of the invention, the marker sequence is a hexa-histidine peptide, as provided in the pQE vector (Qiagen, Inc.) and described in Gentz etal., ProcNatlAcadSci USA (1989) 86:821-824, or is an HA tag. The polynucleotide may also contain non-coding 5 ' and 3 ' sequences, such as transcribed, non-translated sequences, splicing and polyadenylation signals, ribosome binding sites and sequences that stabilize mRNA.
Further embodiments of the present invention include polynucleotides encoding polypeptide variants which comprise the airiino acid sequence of SEQ ID NO:2 and in which several, for instance from 5 to 10, 1 to 5, 1 to 3, 1 to 2 or 1, arnino acid residues are substituted, deleted or added, in any combination.
Polynucleotides which are identical or sufficiently identical to a nucleotide sequence contained in SEQ ID NO: 1, may be used as hybridization probes for cDNA and genomic DNA or as primers for a nucleic acid amplification (PCR) reaction, to isolate full-length cDNAs and genomic clones encoding polypeptides of the present invention and to isolate cDNA and genomic clones of other genes (including genes encoding homologs and orthologs from species other than human) that have a high sequence similarity to SEQ ID NO: 1. Typically these nucleotide sequences are 70% identical, preferably 80% identical, more preferably 90% identical, most preferably 95% identical to that of the referent. The probes or primers will generally comprise at least 15 nucleotides, preferably, at least 30 nucleotides and may have at least 50 nucleotides. Particularly preferred probes will have between 30 and 50 nucleotides.
A polynucleotide encoding a polypeptide of the present invention, including homologs and orthologs from species other than human, may be obtained by a process which comprises the steps of screening an appropriate library under stringent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: 1 or a fragment thereof; and isolating full-length cDNA and genomic clones containing said polynucleotide sequence. Such hybridization techniques are well known to the skilled artisan. Preferred stringent hybridization conditions include overnight incubation at 42°C in a solution comprising: 50% formamide, 5xSSC (150mM NaCl, 15mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5x Denhardt's solution, 10 % dextran sulfate, and 20 microgram/ml denatured, sheared salmon sperm DNA; followed by washing the filters in 0. lx SSC at about 65°C. Thus the present invention also includes polynucleotides obtainable by screening an appropriate library under stringent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: 1 or a fragment thereof. The skilled artisan will appreciate that, in many cases, an isolated cDNA sequence will be incomplete, in that the region coding for the polypeptide is cut short at the 5' end of the cDNA. This is a consequence of reverse transcriptase, an enzyme with inherently low 'processivity' (a measure of the ability of the enzyme to remain attached to the template during the polymerisation reaction), failing to complete a DNA copy of the mRNA template during 1st strand cDNA synthesis.
There are several methods available and well known to those skilled in the art to obtain full-length cDNAs, or extend short cDNAs, for example those based on the method of Rapid Amplification of cDNA ends (RACE) (see, for example, Frohman et al., PNAS USA 85, 8998- 9002, 1988). Recent modifications of the technique, exemplified by the Marathon™' technology (Clontech Laboratories Inc.) for example, have significantly simplified the search for longer cDNAs. In the Marathon™ technology, cDNAs have been prepared from mRNA extracted from a chosen tissue and an 'adaptor' sequence ligated onto each end. Nucleic acid amplification (PCR) is then carried out to amplify the 'missing' 5' end of the cDNA using a combination of gene specific and adaptor specific oligonucleotide primers. The PCR reaction is then repeated using 'nested' primers, that is, primers designed to anneal within the amplified product (typically an adaptor specific primer that anneals further 3' in the adaptor sequence and a gene specific primer that anneals further 5' in the known gene sequence). The products of this reaction can then be analyzed by DNA sequencing and a full-length cDNA constructed either by joining the product directly to the existing cDNA to give a complete sequence, or carrying out a separate full-length PCR using the new sequence information for the design of the 5' pπmer
Recombinant polypeptides of the present mvention may be prepared by processes well known in the art from genetically engineered host cells compnsing expression systems Accordingly, in a further aspect, the present mvention relates to expression systems which compπse a polynucleotide or polynucleotides of the present mvention, to host cells which are genetically engineered with such expression systems and to the production of polypeptides of the mvention by recombinant techniques Cell-free translation systems can also be employed to produce such proteins using RNAs deπved from the DNA constructs of the present mvention For recombinant production, host cells can be genetically engineered to incorporate expression systems or portions thereof for polynucleotides of the present mvention Introduction of polynucleotides into host cells can be effected by methods descπbed m many standard laboratory manuals, such as Davis et al, Basic Methods m Molecular Biology (1986) and Sambrook et al , Molecular Cloning A Laboratory Manual, 2nd Ed , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N Y (1989) Preferred such methods mclude, for instance, calcium phosphate transfection, DEAE-dextran mediated transfection, transvection, microinjection, canonic hpid-mediated transfection, electroporation, transduction, scrape loading, ballistic introduction or infection
Representative examples of appropπate hosts mclude bacteπal cells, such as streptococci, staphylococct, E coh. Streptomyces and Bacillus subtihs cells, fungal cells, such as yeast cells and Aspergillus cells, insect cells such as Drosophtla S2 and Spodoptera Sf9 cells, animal cells such as CHO, COS, HeLa, C127, 3T3, BHK, HEK 293 and Bowes melanoma cells, and plant cells
A great variety of expression systems can be used, for instance, chromosomal, episomal and vrrus-deπve systems, e g , vectors deπved from bacteπal plasmids, from bactenophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccmia viruses, adenovrruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors deπved from combinations thereof, such as those deπved from plasmid and bactenophage genetic elements, such as cosmids and phagemids The expression systems may contain control regions that regulate as well as engender expression Generally, any system or vector which is able to maintain, propagate or express a polynucleotide to produce a polypeptide m a host may be used The appropπate nucleotide sequence may be inserted into an expression system by any of a vaπety of well-known and routine techniques, such as, for example, those set forth m Sambrook et al , MOLECULAR CLONING A LABORATORY MANUAL (supra) Appropπate secretion signals may be incorporated mto the desired polypeptide to allow secretion of the translated protem mto the lumen of the endoplasmic reticulum, the peπplasmic space or the extracellular environment These signals may be endogenous to the polypeptide or they may be heterologous signals
If a polypeptide of the present mvention is to be expressed for use m screening assays, it is generally prefeπed that the polypeptide be produced at the surface of the cell In this event, the cells may be harvested pnor to use m the screening assay If the polypeptide is secreted mto the medium, the medium can be recovered m order to recover and puπfy the polypeptide If produced lntracellularly, the cells must first be lysed before the polypeptide is recovered
Polypeptides of the present mvention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic mteraction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography Most preferably, high performance liquid chromatography is employed for purification Well known techniques for refolding proteins may be employed to regenerate active conformation when the polypeptide is denatured during isolation and or purification This mvention also relates to the use of polynucleotides of the present mvention as diagnostic reagents Detection of a mutated form of the gene characterized by the polynucleotide of SEQ ID NO 1 which is associated with a dysfunction will provide a diagnostic tool that can add to, or define, a diagnosis of a disease, or susceptibility to a disease, which results from under-expression, over- expression or altered expression of the gene Individuals carrying mutations in the gene may be detected at the DNA level by a variety of techniques
Nucleic acids for diagnosis may be obtained from a subject's cells, such as from blood, urine, saliva, tissue biopsy or autopsy mateπal The genomic DNA may be used directly for detection or may be amplified enzymatically by usmg PCR or other amplification techniques pnor to analysis RNA or cDNA may also be used in similar fashion Deletions and insertions can be detected by a change in size of the amplified product in compaπson to the normal genotype Pomt mutations can be identified by hybπdizmg amplified DNA to labeled CBFBLH05 nucleotide sequences Perfectly matched sequences can be distinguished from mismatched duplexes by RNase digestion or by differences in melting temperatures DNA sequence differences may also be detected by alterations m electrophoretic mobility of DNA fragments in gels, with or without denaturing agents, or by direct DNA sequencing (e g , Myers et al , Science (1985) 230 1242) Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and S 1 protection or the chemical cleavage method (see Cotton et al , Proc Natl Acad Sci USA (1985) 85 4397-4401) In another embodiment, an aπav of oligonucleotides probes compπsmg CBFBLH05 nucleotide sequence or fragments thereof can be constructed to conduct efficient screening of e g , genetic mutations Arrav technology methods are well known and have general apphcability and can be used to address a variety of questions in molecular genetics including gene expression, genetic linkage, and genetic variability (see for example: M.Chee et al., Science, Vol 274, pp 610-613 (1996)).
The diagnostic assays offer a process for diagnosing or determining a susceptibility to the Diseases through detection of mutation in the CBFBLH05 gene by the methods described. In addition, such diseases may be diagnosed by methods comprising determining from a sample derived from a subject an abnormally decreased or increased level of polypeptide or rnRNA. Decreased or increased expression can be measured at the RNA level using any of the methods well known in the art for the quantitation of polynucleotides, such as, for example, nucleic acid amplification, for instance PCR, RT-PCR, RNase protection, Northern blotting and other hybridization methods. Assay techniques that can be used to determine levels of a protein, such as a polypeptide of the present invention, in a sample derived from a host are well-known to those of skill in the art. Such assay methods include radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays. Thus in another aspect, the present invention relates to a diagonostic kit which comprises:
(a) a polynucleotide of the present invention, preferably the nucleotide sequence of SEQ ID NO: 1, or a fragment thereof ;
(b) a nucleotide sequence complementary to that of (a);
(c) a polypeptide of the present invention, preferably the polypeptide of SEQ ID NO:2 or a fragment thereof; or
(d) an antibody to a polypeptide of the present invention, preferably to the polypeptide of SEQ ID NO:2.
It will be appreciated that in any such kit, (a), (b), (c) or (d) may comprise a substantial component. Such a kit will be of use in diagnosing a disease or susceptibility to a disease, particularly hver diseases, cancer, autoimmune diseases, and kidney disorders, amongst others. The nucleotide sequences of the present invention are also valuable for chromosome identification. The sequence is specifically targeted to, and can hybridize with, a particular location on an individual human chromosome. The mapping of relevant sequences to chromosomes according to the present invention is an important first step in correlating those sequences with gene associated disease. Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be coπelated with genetic map data. Such data are found in, for example, V. McKusick, Mendelian Inheritance in Man (available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance of physically adjacent genes).
The differences in the cDNA or genomic sequence between affected and unaffected individuals can also be determined. If a mutation is observed in some or all of the affected individuals but not in any normal individuals, then the mutation is likely to be the causative agent of the disease.
The polypeptides of the invention or their fragments or analogs thereof, or cells expressing them, can also be used as immunogens to produce antibodies irnmunospecific for polypeptides of the present invention. The term "irnmunospecific" means that the antibodies have substantially greater affinity for the polypeptides of the invention than their affinity for other related polypeptides in the prior art. Antibodies generated against polypeptides of the present invention may be obtained by adnrinistering the polypeptides or epitope-bearing fragments, analogs or cells to an ariimal, preferably a non-human animal, using routine protocols. For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler, G. and Milstein, C, Nature (1975) 256:495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al. , Immunology Today (1983) 4:72) and the EBV- hybridoma technique (Cole etal, MONOCLONAL ANTIBODIES AND CANCER THERAPY, pp. 77-96, Alan R. Liss, Inc., 1985).
Techniques for the production of single chain antibodies, such as those described in U.S. Patent No. 4,946,778, can also be adapted to produce single chain antibodies to polypeptides of this invention. Also, transgenic mice, or other organisms, including other mammals, may be used to express humanized antibodies.
The above-described antibodies may be employed to isolate or to identify clones expressing the polypeptide or to purify the polypeptides by affinity chromatography. Antibodies against polypeptides of the present invention may also be employed to treat the
Diseases, amongst others.
In a further aspect, the present invention relates to genetically engineered soluble fusion proteins comprising a polypeptide of the present invention, or a fragment thereof, and various portions of the constant regions of heavy or light chains of immunoglobulins of various subclasses (IgG, IgM, IgA, IgE). Preferred as an immunoglobulin is the constant part of the heavy chain of human IgG, particularly IgGl, where fusion takes place at the hinge region. In a particular embodiment, the Fc part can be removed simply by incorporation of a cleavage sequence which can be cleaved with blood clotting factor Xa. Furthermore, this invention relates to processes for the preparation of these fusion proteins by genetic engineering, and to the use thereof for drug screening, diagnosis and therapy. A further aspect of the invention also relates to polynucleotides encoding such fusion proteins. Examples of fusion protein technology can be found in International Patent Application Nos. W094/29458 and W094/22914.
Another aspect of the invention relates to a method for inducing an immunological response in a mammal which comprises inoculating the mammal with a polypeptide of the present invention, adequate to produce antibody and/or T cell immune response to protect said animal from the Diseases hereinbefore mentioned, amongst others. Yet another aspect of the invention relates to a method of inducing immunological response in a mammal which comprises, delivering a polypeptide of the present invention via a vector directing expression of the polynucleotide and coding for the polypeptide in vivo in order to induce such an immunological response to produce antibody to protect said animal from diseases.
A further aspect of the invention relates to an immunological/vaccine formulation (composition) which, when introduced into a mammalian host, induces an immunological response in that mammal to a polypeptide of the present invention wherein the composition comprises a polypeptide or polynucleotide of the present invention. The vaccine formulation may further comprise a suitable carrier. Since a polypeptide may be broken down in the stomach, it is preferably administered parenterally (for instance, subcutaneous, intramuscular, intravenous, or intradermal injection). Formulations suitable for parenteral aclministration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the recipient; and aqueous and non- aqueous sterile suspensions which may include suspending agents or thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampoules and vials and may be stored in a freeze-dried condition requiring only the addition of the sterile liquid carrier immediately prior to use. The vaccine formulation may also include adjuvant systems for enhancing the immunogenicity of the formulation, such as oil-in water systems and other systems known in the art. The dosage will depend on the specific activity of the vaccine and can be readily determined by routine experimentation.
Polypeptides of the present invention are responsible for many biological functions, including many disease states, in particular the Diseases hereinbefore mentioned. It is therefore desirous to devise screening methods to identify compounds which stimulate or which inhibit the function of the polypeptide. Accordingly, in a further aspect, the present invention provides for a method of screening compounds to identify those which stimulate or which inhibit the function of the polypeptide. In general, agonists or antagonists may be employed for therapeutic and prophylactic purposes for such Diseases as hereinbefore mentioned. Compounds may be identified from a variety of sources, for example, cells, cell-free preparations, chemical libraries, and natural product mixtures. Such agonists, antagonists or inhibitors so-identified may be natural or modified substrates, ligands, receptors, enzymes, etc., as the case may be, of the polypeptide; or may be stractural or functional mimetics thereof (see Coligan et al. , Current Protocols in Immunology l(2):Chapter 5 (1991)). The screening method may simply measure the binding of a candidate compound to the polypeptide, or to cells or membranes bearing the polypeptide, or a fusion protein thereof by means of a label directly or indirectly associated with the candidate compound. Alternatively, the screening method may involve competition with a labeled competitor. Further, these screening methods may test whether the candidate compound results in a signal generated by activation or inhibition of the polypeptide, using detection systems appropriate to the cells bearing the polypeptide. Inhibitors of activation are generally assayed in the presence of a known agonist and the effect on activation by the agonist by the presence of the candidate compound is observed. Constitutively active polypeptides may be employed in screening methods for inverse agonists or inhibitors, in the absence of an agonist or inhibitor, by testing whether the candidate compound results in inhibition of activation of the polypeptide. Further, the screening methods may simply comprise the steps of mixing a candidate compound with a solution containing a polypeptide of the present invention, to form a mixture, measuring CBFBLH05 activity in the mixture, and comparing the CBFBLH05 activity of the mixture to a standard. Fusion proteins, such as those made from Fc portion and CBFBLH05 polypeptide, as hereinbefore described, can also be used for high-throughput screening assays to identify antagonists for the polypeptide of the present invention (see D. Bennett et al., J Mol Recognition, 8:52-58 (1995); and K. Johanson et al., J Biol Chem, 270(16):9459-9471 (1995)).
The polynucleotides, polypeptides and antibodies to the polypeptide of the present invention may also be used to configure screening methods for detecting the effect of added compounds on the production of mRNA and polypeptide in cells. For example, an ELISA assay may be constructed for measuring secreted or cell associated levels of polypeptide using monoclonal and polyclonal antibodies by standard methods known in the art. This can be used to discover agents which may inhibit or enhance the production of polypeptide (also called antagonist or agonist, respectively) from suitably manipulated cells or tissues.
The polypeptide may be used to identify membrane bound or soluble receptors, if any, through standard receptor binding techniques known in the art. These include, but are not limited to, ligand binding and crosslinking assays in which the polypeptide is labeled with a radioactive isotope (for instance, ^^1), chemically modified (for instance, biotinylated), or fused to a peptide sequence suitable for detection or puπfication, and mcubated with a source of the putative receptor (cells, cell membranes, cell supernatants, tissue extracts, bodily fluids) Other methods mclude biophysical techniques such as surface plasmon resonance and spectroscopy These screening methods may also be used to identify agomsts and antagonists of the polypeptide which compete with the bmdmg of the polypeptide to its receptors, if any Standard methods for conductmg such assays are well understood m the art
Examples of potential polypeptide antagonists mclude antibodies or, in some cases, ohgonucleotides or proteins which are closely related to the hgands, substrates, receptors, enzymes, etc , as the case may be, of the polypeptide, e , a fragment of the hgands. substrates, receptors, enzymes, etc , or small molecules which bind to the polypeptide of the present mvention but do not elicit a response, so that the activity of the polypeptide is prevented
Thus, m another aspect, the present mvention relates to a screening kit for identifying agomsts. antagonists, hgands, receptors, substrates, enzymes, etc for polypeptides of the present mvention. or compounds which decrease or enhance the production of such polypeptides, which compπses
(a) a polypeptide of the present mvention,
(b) a recombinant cell expressmg a polypeptide of the present mvention,
(c) a cell membrane expressmg a polypeptide of the present mvention, or
(d) antibody to a polypeptide of the present mvention, which polypeptide is preferably that of SEQ ID NO 2
It will be appreciated that in any such kit, (a), (b), (c) or (d) may compπse a substantial component
It will be readily appreciated by the skilled artisan that a polypeptide of the present mvention may also be used m a method for the structure-based design of an agomst, antagonist or inhibitor of the polypeptide, by
(a) deterr mng m the first instance the three-dimensional structure of the polypeptide,
(b) deducmg the three-dimensional structure for the likely reactive or bmdmg sιte(s) of an agomst, antagomst or inhibitor,
(c) synthesizing candidate compounds that are predicted to bmd to or react with the deduced bmdmg or reactive site, and
(d) testmg whether the candidate compounds are mdeed agomsts, antagonists or inhibitors It will be further appreciated that this will normally be an interactive process In a further aspect, the present mvention provides methods of treating abnormal conditions such as, for instance, hver diseases, cancer, autoimmune diseases, and kidney disorders, related to either an excess of, or an under-expression of, CBFBLH05 polypeptide activity
If the activity of the polypeptide is in excess, several approaches are available One approach compπses admmistenng to a subject in need thereof an inhibitor compound (antagonist) as heremabove descπbed. optionally in combination with a pharmaceutically acceptable earner, in an amount effective to inhibit the function of the polypeptide, such as, for example, by blocking the bmdmg of hgands, substrates, receptors, enzymes, etc , or by inhibiting a second signal, and thereby alleviating the abnormal condition In another approach, soluble forms of the polypeptides still capable of bmdmg the ligand, substrate, enzymes, receptors, etc m competition with endogenous polypeptide may be administered Typical examples of such competitors mclude fragments of the CBFBLH05 polypeptide
In still another approach, expression of the gene encoding endogenous CBFBLH05 polypeptide can be inhibited usmg expression blocking techniques Known such techniques mvolve the use of antisense sequences, either internally generated or separately administered (see, for example, O'Connor, J Neurochem (1991) 56 560 m Ohgodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, FL (1988)) Alternatively, ohgonucleotides which form tπple helices with the gene can be supplied (see, for example, Lee et al , Nucleic Acids Res (1979) 6 3073, Cooney et al , Science (1988) 241 456, Dervan et α/ , Science (1991) 251 1360) These ohgomers can be adιmmstered »er se or the relevant oligomers can be expressed in vivo
For treating abnormal conditions related to an under-expression of CBFBLH05 and its activity, several approaches are also available One approach compπses administering to a subject a therapeutically effective amount of a compound which activates a polypeptide of the present mvention. i e , an agomst as descπbed above, in combination with a pharmaceutically acceptable earner, to thereby alleviate the abnormal condition Alternatively, gene therapy may be employed to effect the endogenous production of CBFBLH05 by the relevant cells m the subject For example, a polynucleotide of the mvention may be engineered for expression in a replication defective retroviral vector, as discussed above The retroviral expression construct may then be isolated and introduced mto a packaging cell transduced with a retroviral plasmid vector containmg RNA encoding a polypeptide of the present mvention such that the packaging cell now produces infectious viral particles containmg the gene of interest These producer cells may be administered to a subject for engineering cells in vivo and expression of the polypeptide in vivo For an overview of gene therapy, see Chapter 20, Gene Therapy and other Molecular Genetic-based Therapeutic Approaches, (and references cited therein) in Human Molecular Genetics, T Strachan and A P Read, BIOS Scientific Publishers Ltd (1996) Another approach is to administer a therapeutic amount of a polypeptide of the present invention in combination with a suitable pharmaceutical carrier.
In a further aspect, the present invention provides for pharmaceutical compositions comprising a therapeutically effective amount of a polypeptide, such as the soluble form of a polypeptide of the present invention, agonist/antagonist peptide or small molecule compound, in combination with a pharmaceutically acceptable carrier or excipient. Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The invention further relates to pharmaceutical packs and kits comprising one or more containers filled with one or more of the ingredients of the aforementioned compositions of the invention. Polypeptides and other compounds of the present invention may be employed alone or in conjunction with other compounds, such as therapeutic compounds.
The composition will be adapted to the route of administration, for instance by a systemic or an oral route. Prefeπed forms of systemic administration include injection, typically by intravenous injection. Other injection routes, such as subcutaneous, intramuscular, or intraperitoneal, can be used. Alternative means for systemic administration include transmucosal and transdermal administration using penetrants such as bile salts or fusidic acids or other detergents. In addition, if a polypeptide or other compounds of the present invention can be formulated in an enteric or an encapsulated formulation, oral administration may also be possible. Administration of these compounds may also be topical and/or localized, in the form of salves, pastes, gels, and the like. The dosage range required depends on the choice of peptide or other compounds of the present invention, the route of administration, the nature of the formulation, the nature of the subject's condition, and the judgment of the attending practitioner. Suitable dosages, however, are in the range of 0.1-100 μg/kg of subject. Wide variations in the needed dosage, however, are to be expected in view of the variety of compounds available and the differing efficiencies of various routes of administration. For example, oral administration would be expected to require higher dosages than administration by intravenous injection. Variations in these dosage levels can be adjusted using standard empirical routines for optimization, as is well understood in the art.
Polypeptides used in treatment can also be generated endogenously in the subject, in treatment modalities often refeπed to as "gene therapy" as described above. Thus, for example, cells from a subject may be engineered with a polynucleotide, such as a DNA or RNA, to encode a polypeptide ex vivo, and for example, by the use of a retroviral plasmid vector. The cells are then introduced into the subject.
Polynucleotide and polypeptide sequences form a valuable information resource with which to identify further sequences of similar homology. This is most easily facilitated by storing the sequence in a computer readable medium and then using the stored data to search a sequence database using well known searching tools, such as GCC. Accordingly, in a further aspect, the present invention provides for a computer readable medium having stored thereon a polynucleotide comprising the sequence of SEQ ID NO: 1 and or a polypeptide sequence encoded thereby.
The following definitions are provided to facilitate understanding of certain terms used frequently hereinbefore.
"Antibodies" as used herein includes polyclonal and monoclonal antibodies, chimeric, single chain, and humanized antibodies, as well as Fab fragments, including the products of an Fab or other immunoglobulin expression library.
"Isolated" means altered "by the hand of man" from the natural state. If an "isolated" composition or substance occurs in nature, it has been changed or removed from its original environment, or both. For example, a polynucleotide or a polypeptide naturally present in a living animal is not "isolated," but the same polynucleotide or polypeptide separated from the coexisting materials of its natural state is "isolated", as the term is employed herein.
"Polynucleotide" generally refers to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. "Polynucleotides" include, without limitation, single- and double-stranded DNA, DNA that is a mixture of single- and double- stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions. In addition, "polynucleotide" refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA. The term "polynucleotide" also includes DNAs or RNAs containing one or more modified bases and DNAs or RNAs with backbones modified for stability or for other reasons. "Modified" bases include, for example, tritylated bases and unusual bases such as inosine. A variety of modifications may be made to DNA and RNA; thus, "polynucleotide" embraces chemically, enzymatically or metabolically modified forms of polynucleotides as typically found in nature, as well as the chemical forms of DNA and RNA characteristic of viruses and cells. "Polynucleotide" also embraces relatively short polynucleotides, often referred to as ohgonucleotides .
"Polypeptide" refers to any peptide or protein comprising two or more a ino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres. "Polypeptide" refers to both short chains, commonly referred to as peptides, oligopeptides or oligomers, and to longer chains, generally referred to as proteins. Polypeptides may contain amino acids other than the 20 gene-encoded amino acids. "Polypeptides" include amino acid sequences modified either by natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature. Modifications may occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. It will be appreciated that the same type of modification may be present to the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Polypeptides may be branched as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched and branched cyclic polypeptides may result from post-translation natural processes or may be made by synthetic methods. Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cystine, formation of pyroglutamate, formylation, gamma- carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination (see, for instance, PROTEINS - STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York, 1993; Wold, F., Post-translational Protein Modifications: Perspectives and Prospects, pgs. 1-12 in POSTTRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York, 1983; Seifter et al, "Analysis for protein modifications and nonprotein cofactors", Meth Enzymol (1990) 182:626-646 and Rattan et al. , "Protein Synthesis: Post-translational Modifications and Aging", Ann NYAcad Sci (1992) 663 :48-62). "Variant" refers to a polynucleotide or polypeptide that differs from a reference polynucleotide or polypeptide, but retains essential properties. A typical variant of a polynucleotide differs in nucleotide sequence from another, reference polynucleotide. Changes in the nucleotide sequence of the variant may or may not alter the amino acid sequence of a polypeptide encoded by the reference polynucleotide. Nucleotide changes may result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence, as discussed below. A typical variant of a polypeptide differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference polypeptide and the variant are closely similar overall and, in many regions, identical A vaπant and reference polypeptide may differ m ammo acid sequence by one or more substitutions, additions, deletions m any combmation A substituted or inserted ammo acid residue may or may not be one encoded by the genetic code A vaπant of a polynucleotide or polypeptide may be a naturally occurring such as an allehc vaπant, or it may be a vaπant that is not known to occur naturally Non-naturally occurring vaπants of polynucleotides and polypeptides may be made by mutagenesis techniques or by direct synthesis
"Identity," as known in the art, is a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, as the case may be, as determmed by comparing the sequences In the art, "identity" also means the degree of sequence relatedness between polypeptide or polynucleotide sequences, as the case may be, as determined by the match between stπngs of such sequences "Identity" can be readily calculated by known methods, including but not limited to those descπbed m (Computational Molecular Biology, Lesk, A M , ed , Oxford University Press, New York, 1988, Biocomputing Informatics and Genome Projects, Smith, D W , ed , Academic Press, New York, 1993, Computer Analysis of Sequence Data, Part I, Gπffin, A M , and Gπffin, H G , eds , Humana Press, New Jersey, 1994, Sequence Analysis in Molecular Biology, von Hemje, G , Academic Press, 1987, and Sequence Analysis Primer, Gπbskov, M and Devereux, J , eds , M Stockton Press. New York, 1991, and Caπllo, H , and Lipman, D , SLAM J Applied Math , 48 1073 (1988) Methods to determine identity are designed to give the largest match between the sequences tested Moreover, methods to determine identity are codified in publicly available computer programs Computer program methods to determine identity between two sequences mclude, but are not limited to, the GCG program package (Devereux, J , et al , Nucleic Acids Research 12(1) 387 (1984)), BLASTP, BLASTN, and FASTA (Atschul, S F et al , J Molec Biol 215 403-410 (1990) The BLAST X program is publicly available from NCBI and other sources (BLAST Manual, Altschul, S , et al , NCBI NLM NIH Bethesda, MD 20894, Altschul, S , et al , J Mol Biol 215 403-410 (1990) The well known Smith Waterman algoπthm may also be used to determine identity
Parameters for polypeptide sequence compaπson mclude the following 1) Algoπthm Needleman and Wunsch, J Mol Biol 48 443-453 (1970) Compaπson matπx BLOSSUM62 from Hentikoff and Hentikoff, Proc Natl Acad Sci USA 89 10915-10919 (1992) Gap Penalty 12 Gap Length Penalty 4 A program useful with these parameters is publicly available as the "gap" program from Genetics Computer Group, Madison WI. The aforementioned parameters are the default parameters for peptide comparisons (along with no penalty for end gaps).
Parameters for polynucleotide comparison include the following: 1) Algorithm: Needleman and Wunsch, J. Mol Biol. 48: 443-453 (1970) Comparison matrix: matches = +10, mismatch = 0 Gap Penalty: 50 Gap Length Penalty: 3 Available as: The "gap" program from Genetics Computer Group, Madison WI. These are the default parameters for nucleic acid comparisons.
A preferred meaning for "identity" for polynucleotides and polypeptides, as the case may be, are provided in (1) and (2) below.
(1) Polynucleotide embodiments further include an isolated polynucleotide comprising a polynucleotide sequence having at least a 50, 60, 70, 80, 85, 90, 95, 97 or 100% identity to the reference sequence of SEQ ID NO: 1, wherein said polynucleotide sequence may be identical to the reference sequence of SEQ ID NO: 1 or may include up to a certain integer number of nucleotide alterations as compared to the reference sequence, wherein said alterations are selected from the group consisting of at least one nucleotide deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terrninal positions, interspersed either individually among the nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence, and wherein said number of nucleotide alterations is determined by multiplying the total number of nucleotides in SEQ ID NO: 1 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of nucleotides in SEQ ID NOT, or:
nn < xn - (xn . y),
wherein nn is the number of nucleotide alterations, xn is the total number of nucleotides in SEQ ID NOT, y is 0.50 for 50%, 0.60 for 60%, 0.70 for 70%, 0.80 for 80%, 0.85 for 85%, 0.90 for 90%, 0.95 for 95%, 0.97 for 97% or 1.00 for 100%, and • is the symbol for the multiplication operator, and wherein any non-integer product of xn and y is rounded down to the nearest integer prior to subtracting it from xn. Alterations of a polynucleotide sequence encoding the polypeptide of SEQ ID NO:2 may create nonsense, missense or frameshift mutations in this coding sequence and thereby alter the polypeptide encoded by the polynucleotide following such alterations.
By way of example, a polynucleotide sequence of the present invention may be identical to the reference sequence of SEQ ID NO:2, that is it may be 100% identical, or it may include up to a certain integer number of amino acid alterations as compared to the reference sequence such that the percent identity is less than 100% identity. Such alterations are selected from the group consisting of at least one nucleic acid deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5' or 3' terminal positions of the reference polynucleotide sequence or anywhere between those terminal positions, interspersed either individually among the nucleic acids in the reference sequence or in one or more contiguous groups within the reference sequence. The number of nucleic acid alterations for a given percent identity is determined by multiplying the total number of amino acids in SEQ ID NO:2 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of amino acids in SEQ ID NO:2, or:
nn < xn - (xn - y),
wherein nn is the number of amino acid alterations, xn is the total number of amino acids in SEQ ID NO:2, y is, for instance 0.70 for 70%, 0.80 for 80%, 0.85 for 85% etc., • is the symbol for the multiplication operator, and wherein any non-integer product of xn and y is rounded down to the nearest integer prior to subtracting it from xn.
(2) Polypeptide embodiments further include an isolated polypeptide comprising a polypeptide having at least a 50,60, 70, 80, 85, 90, 95, 97 or 100% identity to a polypeptide reference sequence of SEQ ID NO:2, wherein said polypeptide sequence may be identical to the reference sequence of SEQ ID NO: 2 or may include up to a certain integer number of amino acid alterations as compared to the reference sequence, wherein said alterations are selected from the group consisting of at least one amino acid deletion, substitution, including conservative and non- conservative substitution, or insertion, and wherein said alterations may occur at the amino- or carboxy-terminal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the amino acids in the reference sequence or in one or more contiguous groups within the reference sequence, and wherein said number of amino acid alterations is determined by multiplying the total number of amino acids in SEQ ID NO:2 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of amino acids in SEQ ID NO:2, or: na < xa - (xa • y),
wherein na is the number of amino acid alterations, xa is the total number of amino acids in SEQ ID NO:2, y is 0.50 for 50%, 0.60 for 60%, 0.70 for 70%, 0.80 for 80%, 0.85 for 85%, 0.90 for 90%, 0.95 for 95%, 0.97 for 97% or 1.00 for 100%, and • is the symbol for the multiplication operator, and wherein any non-integer product of xa and y is rounded down to the nearest integer prior to subtracting it from xa.
By way of example, a polypeptide sequence of the present invention may be identical to the reference sequence of SEQ ID NO:2, that is it may be 100% identical, or it may include up to a certain integer number of amino acid alterations as compared to the reference sequence such that the percent identity is less than 100% identity. Such alterations are selected from the group consisting of at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion, and wherein said alterations may occur at the amino- or carboxy-terminal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the amino acids in the reference sequence or in one or more contiguous groups within the reference sequence. The number of amino acid alterations for a given % identity is determined by multiplying the total number of amino acids in SEQ ID NO:2 by the integer defining the percent identity divided.by 100 and then subtracting that product from said total number of amino acids in SEQ ID NO :2, or:
na < xa - (xa • y),
wherein na is the number of amino acid alterations, xa is the total number of amino acids in SEQ ID NO:2, y is, for instance 0.70 for 70%, 0.80 for 80%, 0.85 for 85% etc., and • is the symbol for the multiplication operator, and wherein any non-integer product of xa and y is rounded down to the nearest integer prior to subtracting it from xa.
"Fusion protein" refers to a protein encoded by two, often unrelated, fused genes or fragments thereof. In one example, EP-A-0 464 discloses fusion proteins comprising various portions of constant region of immunoglobulin molecules together with another human protein or part thereof. In many cases, employing an immunoglobulin Fc region as a part of a fusion protein is advantageous for use in therapy and diagnosis resulting in, for example, improved pharmacokinetic properties [see, e.g., EP-A 0232 262]. On the other hand, for some uses it would be desirable to be able to delete the Fc part after the fusion protein has been expressed, detected and purified.
All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth.
SEQUENCE INFORMATION SEQ ID NO:l
1 GGTGTATGTG CGGCAATAAC ATGTCAACCC CGCTGCCCGC CATCGTGCCC
51 GCCGCCCGGA AGGCCACCGC TGCGGTGATT TTCCTGCATG GATTGGGAGA
101 TACTGGGCAC GGATGGGCAG AAGCCTTTGC AGGTATCAGA AGTTCACATA
151 TCAAATATAT CTGCCCGCAT GCGTTTGATA TTATTGGGCT TTCACCAGAT
201 TCACAGGAGG ATGAATCTGG GATTAAACAG GCAGCAGAAA ATATAAAAGC
251 TTTGATTGAT CAAGAAGTGA AGAATGGCAT TCCTTCTAAC AGAATTATTT
301 TGGGAGGGTT TTCTCAGGGA GGAGCTTTAT CTTTATATAC TGCCCTTACC
351 ACACAGCAGA AACTGGCAGG TGTCACTGCA CTCAGTTGCT GGCTTCCACT
401 TCGGGCTTCC TTTCCACAGG GTCCTATCGG TGGTGCTAAT AGAGATATTT
451 CTATTCTCCA GTGCCACGGG GATTGTGACC CTTTGGTTCC CCTGATGTTT
501 GGTTCTCTTA CGGTGGAAAA ACTAAAAACA TTGGTGAATC CAGCCAATGT
551 GACCTTTAAA ACCTATGAAG GTATGATGCA CAGTTCGTGT CAACAGGAAA
601 TGATGGATGT CAAGCAATTC ATTGATAAAC TCCTACCTCC AATTGATTGA
651 CGTCACTAAG AGGCCTTGTG TAGAAGTACA CCAGCATCAT TGTAGTAGAG
701 TGTAAACCTT TTCCCATGCC CAGTCTTCAA ATTTCTAATG TTTGCAGTGT
751 TTAAATGTTT TGCAAATACA TGCCATTAAC ACAGATCAAT AATATCTCCT
801 CTGAGAATTT ATGATCTTAA GTCTATACAT GTATTCTTAT AAGACGACCC
851 AGGATCTACT ATATTAGAAT AGATGAAGCA GGTAGCTTCT TTTTTCTCAA 901 ATGTAATTCA GCAAAATAAT ACAGTACTGC CACCAGATTT TTTATTACAT
951 CATTTGAAAA TTAGCAGTAT GCTTAATGAA AATTTGTTCA GGTATAAATG
1001 AGCAGTTAAG ATATAAACAA TTTATGCATG CTGTGACTTA GTCTATGGAT
1051 TTATTCCAAA ATTGCTTAGT CACCATGCAG TGTCTGTATT TTTATATATG
1101 TGTTCATATA TACATAATGA TTATAATACA TAATAAGAAT GAGGTGGTAT
1151 TACATTATTC CTAATAATAG GGATAATGCT GTTTATTGTC AAGAAAAAGT
1201 AAAATCGTTC TCTTCAATTA ATGGCCCTTT TATTTTGGGA CCAGGCTTTT
1251 ATTTTCCCTG ATATTATTTC TATTTAATAC TCTTTTCTCT CAAGAAAAAA
SEQ ID NO: 2
1 MCGNNMSTPL PAIVPAARKA TAAVIFLHGL GDTGHGWAEA FAGIRSSHIK
51 YICPHAFDII GLSPDSQEDE SGIKQAAENI KALIDQEVKN GIPSNRIILG
101 GFSQGGALSL YTA TTQQKL AGVTALSC L P RASFPQGP IGGANRDISI
151 LQCHGDCDPL VP MFGSLTV EK KT VNPA NVTFKTYEGM MHSSCQQEMM
201 DVKQFIDKLL PPID

Claims

What is claimed is:
1. An isolated polypeptide selected from the group consisting of:
(i) an isolated polypeptide comprising an amino acid sequence selected from the group having at least:
(a) 70% identity;
(b) 80% identity;
(c) 90% identity; or
(d) 95% identity to the amino acid sequence of SEQ ID NO:2 over the entire length of SEQ ID
NO:2; (ii) an isolated polypeptide comprising the amino acid sequence of SEQ ID NO:2 or (iii) an isolated polypeptide which is the amino acid sequence of SEQ ID NO:2.
2. An isolated polynucleotide selected from the group consisting of:
(i) an isolated polynucleotide comprising a nucleotide sequence encoding a polypeptide that has at least
(a) 70% identity;
(b) 80% identity; (c) 90% identity; or
(d) 95% identity; to the amino acid sequence of SEQ ID NO:2, over the entire length of SEQ ID NO:2; (ii) an isolated polynucleotide comprising a nucleotide sequence that has at least:
(a) 70% identity (b) 80% identity;
(c) 90% identity; or
(d) 95% identity; over its entire length to a nucleotide sequence encoding the polypeptide of SEQ ID NO:2; (iii) an isolated polynucleotide comprising a nucleotide sequence which has at least:
(a) 10% identity;
(b) 80% identity;
(c) 90% identity; or
(d) 95% identity; to that of SEQ ID NO: 1 over the entire length of SEQ ID NOT; (iv) an isolated polynucleotide comprising a nucleotide sequence encoding the polypeptide of SEQ ID NO:2;
(vi) an isolated polynucleotide which is the polynucleotide of SEQ ID NO: 1; or (vi) an isolated polynucleotide obtainable by screening an appropriate library under stringent hybridization conditions with a labelled probe having the sequence of SEQ ID NO: 1 or a fragment thereof. ; or a nucleotide sequence complementary to said isolated polynucleotide.
3. An antibody irnmunospecific for the polypeptide of claim 1.
4. A method for the treatment of a subject:
(i) in need of enhanced activity or expression of the polypeptide of claim 1 comprising:
(a) administering to the subject a therapeutically effective amount of an agonist to said polypeptide; and/or
(b) providing to the subject an isolated polynucleotide comprising a nucleotide sequence encoding said polypeptide in a form so as to effect production of said polypeptide activity in vivo.; or
(ii) having need to inhibit activity or expression of the polypeptide of claim 1 comprising:
(a) administering to the subject a therapeutically effective amount of an antagonist to said polypeptide; and/or
(b) adrninistering to the subject a nucleic acid molecule that inhibits the expression of a nucleotide sequence encoding said polypeptide; and/or (c) adrninistering to the subject a therapeutically effective amount of a polypeptide that competes with said polypeptide for its ligand, substrate , or receptor.
5. A process for diagnosing a disease or a susceptibility to a disease in a subject related to expression or activity of the polypeptide of claim 1 in a subject comprising:
(a) determining the presence or absence of a mutation in the nucleotide sequence encoding said polypeptide in the genome of said subject; and/or
(b) analyzing for the presence or amount of said polypeptide expression in a sample derived from said subject.
6. A method for screening to identify compounds which stimulate or which inhibit the function of the polypeptide of claim 1 which comprises a method selected from the group consisting of:
(a) measuring the binding of a candidate compound to the polypeptide (or to the cells or membranes bearing the polypeptide) or a fusion protein thereof by means of a label directly or indirectly associated with the candidate compound;
(b) measuring the binding of a candidate compound to the polypeptide (or to the cells or membranes bearing the polypeptide) or a fusion protein thereof in the presence of a labeled competitor; (c) testing whether the candidate compound results in a signal generated by activation or inhibition of the polypeptide, using detection systems appropriate to the cells or cell membranes bearing the polypeptide;
(d) mixing a candidate compound with a solution containing a polypeptide of claim 1, to form a mixture, measuring activity of the polypeptide in the mixture, and comparing the activity of the mixture to a standard; or
(e) detecting the effect of a candidate compound on the production of mRNA encoding said polypeptide and said polypeptide in cells, using for instance, an ELISA assay.
7. An agonist or an antagonist of the polypeptide of claim 1.
8. An expression system comprising a polynucleotide capable of producing a polypeptide of claim 1 when said expression system is present in a compatible host cell.
9. A process for producing a recombinant host cell comprising transforming or transfecting a cell with the expression system of claim 8 such that the host cell, under appropriate culture conditions, produces a polypeptide comprising an amino acid sequence having at least 70% identity to the amino acid sequence of SEQ ID NO:2 over the entire length of SEQ ID NO:2.
10. A recombinant host cell produced by the process of claim 9.
11. A membrane of a recombinant host cell of claim 10 expressing a polypeptide comprising an amino acid sequence having at least 70% identity to the amino acid sequence of SEQ ID NO:2 over the entire length of SEQ ID NO:2.
12. A process for producing a polypeptide comprising culturing a host cell of claim 10 under conditions sufficient for the production of said polypeptide and recovering the polypeptide from the culture.
PCT/CN1998/000164 1998-08-11 1998-08-11 Human lysophospholipase gene (cbfblh05) WO2000009556A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
PCT/CN1998/000164 WO2000009556A1 (en) 1998-08-11 1998-08-11 Human lysophospholipase gene (cbfblh05)
CN98810060.6A CN1275131A (en) 1998-08-11 1998-08-11 Human lysophospholipase gene (CBFBLH05)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN1998/000164 WO2000009556A1 (en) 1998-08-11 1998-08-11 Human lysophospholipase gene (cbfblh05)

Publications (1)

Publication Number Publication Date
WO2000009556A1 true WO2000009556A1 (en) 2000-02-24

Family

ID=4575080

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN1998/000164 WO2000009556A1 (en) 1998-08-11 1998-08-11 Human lysophospholipase gene (cbfblh05)

Country Status (2)

Country Link
CN (1) CN1275131A (en)
WO (1) WO2000009556A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2048501B1 (en) * 2006-08-03 2016-03-09 The University of Tokyo Antibody specific to intact human autotaxin, method of screening the same and method and reagent for examining malignant lymphoma by assaying autotaxin

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DDBJ D63885, 19 September 1996, see the sequence or in: J. Biol. Chem. 1996, March 29; 271(13):7705-7711, Sugimoto H., et al.: "Purification cDNA cloning and regulation of lysophospholipase from rat liver", see the abstract. *
GenBank AF035293, 04 December 1997, see the sequence or in: Anal. Biochem. 1996, Apr. 5; 236(1):107-113, Andersson B., et al.: "A 'double adaptor' method for improved shotgun library construction", see the abstract or in: Genome Res. 1977, Apr.; 7(4):353-358, Yu W., et al.: "Large-scale concatenation cDNA sequencing", see the abstract. *
GenBank AF081281, 05 August 1998, see the sequence. *
GenBank U89352, 27 February 1997, see the sequence. *
J. Am. Soc. Nephrol 1998, July; 9(7):1178-1186, Portilla D., et al.: "cDNA cloning and expression of a novel family of enzymes with calcium-independent phospholipase A2 and lysophospholipase activities", see the abstract. *

Also Published As

Publication number Publication date
CN1275131A (en) 2000-11-29

Similar Documents

Publication Publication Date Title
EP1085897A1 (en) Acrp30r1l, a homolog of acrp30 (30 kd adipocyte complement-related protein)
WO1999002561A1 (en) Human f11 antigen: a cell surface receptor involved in platelet aggregation
US20010012836A1 (en) Human histone deacetylase gene HD4
EP1179001A1 (en) A splicing variant of human membrane-type matrix metalloproteinase-5 (mt-mmp5-l)
US6245550B1 (en) Cytokine family member EF-7
EP0866127A2 (en) HE8AN36, a steroid hormone receptor homolog
WO1999049023A1 (en) The cytokine family member 2-21
WO1999011783A2 (en) T-box polypeptides
US20010049121A1 (en) Cytokine family member, 2-19
WO2000011143A1 (en) Human phosphatidylinositol 3-kinase regulatory p101 subunit (ec 2.7.1.137)
WO2000017349A1 (en) A HUMAN Hsg III GENE
WO2000009556A1 (en) Human lysophospholipase gene (cbfblh05)
EP0899328A2 (en) Human p101/P13 kinase p101 subunit polypeptides and polynucleotides
EP0877030A2 (en) EPO primary response gene 1,(EPRG1)
US20020055476A1 (en) RAMP2a: receptor activity modifying protein-2a
WO2000034470A1 (en) Npcbac06: human transitional endoplasmic reticulum atpase gene
EP1083923A1 (en) Acrp30r1: a homolog of acrp30 (30 kd adipocyte complement-related protein)
WO2000061598A2 (en) Rat kidney specific gene profilin-3
WO2000021992A1 (en) A human fk506 binding protein (fkbp)
WO2000034328A1 (en) Tpaaoh04: human g protein sara gene
WO2000058353A1 (en) Npcahh01: human transmembrane protein e3-16 gene
WO1999062947A1 (en) A human ubiquinone oxireductase subunit ci-pdsw homolog gene (cblaic08)
EP0897986A2 (en) Aspartic proteinase 5 (ASP5)
WO2000047616A1 (en) Tpaace06: human ptd016 gene
WO2000021994A1 (en) Human ptd011 gene (tpaahe09)

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 98810060.6

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): CN

NENP Non-entry into the national phase

Ref country code: CA