US20030087409A1 - Human lysyl hydroxylase-like protein - Google Patents

Human lysyl hydroxylase-like protein Download PDF

Info

Publication number
US20030087409A1
US20030087409A1 US10/235,521 US23552102A US2003087409A1 US 20030087409 A1 US20030087409 A1 US 20030087409A1 US 23552102 A US23552102 A US 23552102A US 2003087409 A1 US2003087409 A1 US 2003087409A1
Authority
US
United States
Prior art keywords
polypeptide
polynucleotide
hlhlp
antibody
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/235,521
Inventor
Olga Bandman
Purvi Shah
Preeti Lal
Neil Corley
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Incyte Corp
Original Assignee
Incyte Genomics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Incyte Genomics Inc filed Critical Incyte Genomics Inc
Priority to US10/235,521 priority Critical patent/US20030087409A1/en
Publication of US20030087409A1 publication Critical patent/US20030087409A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0071Oxidoreductases (1.) acting on paired donors with incorporation of molecular oxygen (1.14)
    • 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
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes

Abstract

The invention provides a human lysyl hydroxylase-like protein (HLHLP) and polynucleotides which identify and encode HLHLP. The invention also provides expression vectors, host cells, antibodies, agonists, and antagonists. The invention also provides methods for treating or preventing disorders associated with expression of HLHLP.

Description

  • This patent application is a divisional application of U.S. application Ser. No. 09/593,826, filed Jun. 13, 2000, which is a divisional application of U.S. application Ser. No. 08/989,385 filed Dec. 12, 1997, now U.S. Pat. No. 6,130,039, issued Oct. 10, 2000, all of which applications and patents are hereby incorporated by reference.[0001]
  • FIELD OF THE INVENTION
  • This invention relates to nucleic acid and amino acid sequences of a human lysyl hydroxylase-like protein and to the use of these sequences in the diagnosis, treatment, and prevention of cancer and connective tissue disorders. [0002]
  • BACKGROUND OF THE INVENTION
  • Collagens are a family of nineteen different fibrous structural proteins that are found in several types of tissues. Collagens are the most abundant proteins in mammals, and are essential for the formation of connective tissue such as skin, bone, tendon, cartilage, blood vessels and teeth. Members of the collagen family can be distinguished from one another by the degree of cross-linking between collagen fibers and by the amount of carbohydrate units (e.g., galactose or glucosylgalactose) attached to the collagen fibers. Hydroxylated lysine residues (hydroxylysine) are essential for stability of cross-linking and as attachment points for carbohydrate units. [0003]
  • The enzyme lysyl hydroxylase catalyzes the hydroxylation of lysine residues to form hydroxylysine. Lysyl hydroxylase targets the lysine residue of the sequence X-lys-gly. Two isoforms of lysyl hydroxylase have been characterized, termed PLOD (procollagen-lysine, 2-oxoglutarate 5-dioxygenase) and PLOD2. The two enzymes share 75% sequence homology, with even higher similarity in the C-terminal region (Valtavaara, M. et al. (1997) J. Biol. Chem. 272: 6831-4). [0004]
  • Diminished lysyl hydroxylase activity is involved in certain connective tissue disorders. In particular mutations, including a truncation and duplications within the coding region of the gene for PLOD, have been described in patients with type VI Ehlers-Danos syndrome (Hyland, J. et al. (1992) Nature Genet. 2: 228-31; Hautala, T. et al. (1993) Genomics 15: 399-404). [0005]
  • The discovery of a new human lysyl hydroxylase-like protein and the polynucleotides encoding it satisfies a need in the art by providing new compositions which are useful in the diagnosis, treatment, and prevention of cancer and connective tissue disorders. [0006]
  • SUMMARY OF THE INVENTION
  • The invention features a substantially purified polypeptide, human lysyl hydroxylase-like protein (HLHLP), comprising the amino acid sequence of SEQ ID NO:1 or a fragment of SEQ ID NO:1. [0007]
  • The invention further provides a substantially purified variant of HLHLP having at least 90% amino acid identity to the amino acid sequence of SEQ ID NO:1 or a fragment of SEQ ID NO:1. The invention also provides an isolated and purified polynucleotide sequence encoding the polypeptide comprising the amino acid sequence of SEQ ID NO:1 or a fragment of SEQ ID NO:1. The invention also includes an isolated and purified polynucleotide variant having at least 90% polynucleotide identity to the polynucleotide sequence encoding the polypeptide comprising the amino acid sequence of SEQ ID NO:1 or a fragment of SEQ ID NO:1. [0008]
  • Additionally, the invention provides a composition comprising a polynucleotide sequence encoding the polypeptide comprising the amino acid sequence of SEQ ID NO:1 or a fragment of SEQ ID NO:1. The invention further provides an isolated and purified polynucleotide sequence which hybridizes under stringent conditions to the polynucleotide sequence encoding the polypeptide comprising the amino acid sequence of SEQ ID NO:1 or a fragment of SEQ ID NO:1, as well as an isolated and purified polynucleotide sequence which is complementary to the polynucleotide sequence encoding the polypeptide comprising the amino acid sequence of SEQ ID NO:1 or a fragment of SEQ ID NO:1. [0009]
  • The invention also provides an isolated and purified polynucleotide sequence comprising SEQ ID NO:2 or a fragment of SEQ ID NO:2, and an isolated and purified polynucleotide variant having at least 90% polynucleotide identity to the polynucleotide sequence comprising SEQ ID NO:2 or a fragment of SEQ ID NO:2. The invention also provides an isolated and purified polynucleotide sequence which is complementary to the polynucleotide sequence comprising SEQ ID NO:2 or a fragment of SEQ ID NO:2. [0010]
  • The invention further provides an expression vector containing at least a fragment of the polynucleotide sequence encoding the polypeptide comprising the amino acid sequence of SEQ ID NO:1 or a fragment of SEQ ID NO:1. In another aspect, the expression vector is contained within a host cell. [0011]
  • The invention also provides a method for producing a polypeptide comprising the amino acid sequence of SEQ ID NO:1 or a fragment of SEQ ID NO:1, the method comprising the steps of: (a) culturing the host cell containing an expression vector containing at least a fragment of a polynucleotide sequence encoding HLHLP under conditions suitable for the expression of the polypeptide; and (b) recovering the polypeptide from the host cell culture. [0012]
  • The invention also provides a pharmaceutical composition comprising a substantially purified HLHLP having the amino acid sequence of SEQ ID NO:1 or a fragment of SEQ ID NO:1 in conjunction with a suitable pharmaceutical carrier. [0013]
  • The invention further includes a purified antibody which binds to a polypeptide comprising the amino acid sequence of SEQ ID NO:1 or a fragment of SEQ ID NO:1, as well as a purified agonist and a purified antagonist of the polypeptide. [0014]
  • The invention also provides a method for treating or preventing a cancer, the method comprising administering to a subject in need of such a treatment an effective amount of an antagonist of HLHLP. [0015]
  • The invention also provides a method for treating or preventing a connective tissue disorder, the method comprising administering to a subject in need of such treatment an effective amount of a pharmaceutical composition comprising substantially purified HLHLP. [0016]
  • The invention also provides a method for treating or preventing a connective tissue disorder, the method comprising administering to a subject in need of such treatment an effective amount of an agonist of HLHLP. [0017]
  • The invention also provides a method for detecting a polynucleotide encoding HLHLP in a biological sample containing nucleic acids, the method comprising the steps of: (a) hybridizing the complement of the polynucleotide sequence encoding the polypeptide comprising SEQ ID NO:1 or a fragment of SEQ ID NO:1 to at least one of the nucleic acids of the biological sample, thereby forming a hybridization complex; and (b) detecting the hybridization complex, wherein the presence of the hybridization complex correlates with the presence of a polynucleotide encoding HLHLP in the biological sample. In one aspect, the nucleic acids of the biological sample are amplified by the polymerase chain reaction prior to the hybridizing step.[0018]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIGS. 1A, 1B, [0019] 1C, 1D, 1E, 1F, and 1G show the amino acid sequence (SEQ ID NO:1) and nucleic acid sequence (SEQ ID NO:2) of HLHLP. The alignment was produced using MacDNASIS PRO software (Hitachi Software Engineering Co. Ltd., S. San Francisco, Calif.).
  • FIGS. 2A, 2B, [0020] 2C, and 2D show the amino acid sequence alignments among HLHLP (1622313; SEQ ID NO:1), lysyl hydroxylase isoform 2 (PLOD2) (GI 2138314; SEQ ID NO:3), and lysyl hydroxylase (PLOD) (GI 190074; SEQ ID NO:4), produced using the multisequence alignment program of LASERGENE software (DNASTAR Inc, Madison Wis.).
  • DESCRIPTION OF THE INVENTION
  • Before the present proteins, nucleotide sequences, and methods are described, it is understood that this invention is not limited to the particular methodology, protocols, cell lines, vectors, and reagents described, as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. [0021]
  • It must be noted that as used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly dictates otherwise. Thus, for example, a reference to “a host cell” includes a plurality of such host cells, and a reference to “an antibody” is a reference to one or more antibodies and equivalents thereof known to those skilled in the art, and so forth. [0022]
  • Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods, devices, and materials are now described. All publications mentioned herein are cited for the purpose of describing and disclosing the cell lines, vectors, and methodologies which are reported in the publications and which might be used in connection with the invention. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention. [0023]
  • Definitions [0024]
  • “HLHLP,” as used herein, refers to the amino acid sequences of substantially purified HLHLP obtained from any species, particularly a mammalian species, including bovine, ovine, porcine, murine, equine, and preferably the human species, from any source, whether natural, synthetic, semi-synthetic, or recombinant. [0025]
  • The term “agonist,” as used herein, refers to a molecule which, when bound to HLHLP, increases or prolongs the duration of the effect of HLHLP. Agonists may include proteins, nucleic acids, carbohydrates, or any other molecules which bind to and modulate the effect of HLHLP. [0026]
  • An “allele” or an “allelic sequence,” as these terms are used herein, is an alternative form of the gene encoding HLHLP. Alleles may result from at least one mutation in the nucleic acid sequence and may result in altered mRNAs or in polypeptides whose structure or function may or may not be altered. Any given natural or recombinant gene may have none, one, or many allelic forms. Common mutational changes which give rise to alleles are generally ascribed to natural deletions, additions, or substitutions of nucleotides. Each of these types of changes may occur alone, or in combination with the others, one or more times in a given sequence. [0027]
  • “Altered” nucleic acid sequences encoding HLHLP, as described herein, include those sequences with deletions, insertions, or substitutions of different nucleotides, resulting in a polynucleotide the same HLHLP or a polypeptide with at least one functional characteristic of HLHLP. Included within this definition are polymorphisms which may or may not be readily detectable using a particular oligonucleotide probe of the polynucleotide encoding HLHLP, and improper or unexpected hybridization to alleles, with a locus other than the normal chromosomal locus for the polynucleotide sequence encoding HLHLP. The encoded protein may also be “altered,” and may contain deletions, insertions, or substitutions of amino acid residues which produce a silent change and result in a functionally equivalent HLHLP. Deliberate amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues, as long as the biological or immunological activity of HLHLP is retained. For example, negatively charged amino acids may include aspartic acid and glutamic acid, positively charged amino acids may include lysine and arginine, and amino acids with uncharged polar head groups having similar hydrophilicity values may include leucine, isoleucine, and valine; glycine and alanine; asparagine and glutamine; serine and threonine; and phenylalanine and tyrosine. [0028]
  • The terms “amino acid” or “amino acid sequence,” as used herein, refer to an oligopeptide, peptide, polypeptide, or protein sequence, or a fragment of any of these, and to naturally occurring or synthetic molecules. In this context, “fragments” refers to fragments of HLHLP which are preferably about 5 to about 15 amino acids in length and which retain some biological activity or immunological activity of HLHLP. Where “amino acid sequence” is recited herein to refer to an amino acid sequence of a naturally occurring protein molecule, “amino acid sequence” and like terms are not meant to limit the amino acid sequence to the complete native amino acid sequence associated with the recited protein molecule. [0029]
  • “Amplification,” as used herein, relates to the production of additional copies of a nucleic acid sequence. Amplification is generally carried out using polymerase chain reaction (PCR) technologies well known in the art. (See, e.g., Dieffenbach, C. W. and G. S. Dveksler (1995) [0030] PCR Primer, a Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y.)
  • The term “antagonist,” as it is used herein, refers to a molecule which, when bound to HLHLP, decreases the amount or the duration of the effect of the biological or immunological activity of HLHLP. Antagonists may include proteins, nucleic acids, carbohydrates, antibodies, or any other molecules which decrease the effect of HLHLP. [0031]
  • As used herein, the term “antibody” refers to intact molecules as well as to fragments thereof, such as Fa, F(ab′)[0032] 2, and Fv fragments, which are capable of binding the epitopic determinant. Antibodies that bind HLHLP polypeptides can be prepared using intact polypeptides or using fragments containing small peptides of interest as the immunizing antigen. The polypeptide or oligopeptide used to immunize an animal (e.g., a mouse, a rat, or a rabbit) can be derived from the translation of RNA, or synthesized chemically, and can be conjugated to a carrier protein if desired. Commonly used carriers that are chemically coupled to peptides include bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin (KLH). The coupled peptide is then used to immunize the animal.
  • The term “antigenic determinant,” as used herein, refers to that fragment of a molecule (i.e., an epitope) that makes contact with a particular antibody. When a protein or a fragment of a protein is used to immunize a host animal, numerous regions of the protein may induce the production of antibodies which bind specifically to antigenic determinants (given regions or three-dimensional structures on the protein). An antigenic determinant may compete with the intact antigen (i.e., the immunogen used to elicit the immune response) for binding to an antibody. [0033]
  • The term “antisense,” as used herein, refers to any composition containing a nucleic acid sequence which is complementary to a specific polynucleotide sequence. The term “antisense strand” is used in reference to a nucleic acid strand that is complementary to the “sense” strand. Antisense molecules may be produced by any method including synthesis or transcription. Once introduced into a cell, the complementary nucleotides combine with natural sequences produced by the cell to form duplexes and to block either transcription or translation. The designation “negative” can refer to the antisense strand, and the designation “positive” can refer to the sense strand. [0034]
  • As used herein, the term “biologically active,” refers to a protein having structural, regulatory, or biochemical functions of a naturally occurring molecule. Likewise, “immunologically active” refers to the capability of the natural, recombinant, or synthetic HLHLP, or of any oligopeptide thereof, to induce a specific immune response in appropriate animals or cells and to bind with specific antibodies. [0035]
  • The terms “complementary” or “complementarity,” as used herein, refer to the natural binding of polynucleotides under permissive salt and temperature conditions by base pairing. For example, the sequence “A-G-T” binds to the complementary sequence “T-C-A.” Complementarity between two single-stranded molecules may be “partial,” such that only some of the nucleic acids bind, or it may be “complete,” such that total complementarity exists between the single stranded molecules. The degree of complementarity between nucleic acid strands has significant effects on the efficiency and strength of the hybridization between the nucleic acid strands. This is of particular importance in amplification reactions, which depend upon binding between nucleic acids strands, and in the design and use of peptide nucleic acid (PNA) molecules. [0036]
  • A “composition comprising a given polynucleotide sequence” or a “composition comprising a given amino acid sequence,” as these terms are used herein, refer broadly to any composition containing the given polynucleotide or amino acid sequence. The composition may comprise a dry formulation or an aqueous solution. Compositions comprising polynucleotide sequences encoding HLHLP or fragments of HLHLP may be employed as hybridization probes. The probes may be stored in freeze-dried form and may be associated with a stabilizing agent such as a carbohydrate. In hybridizations, the probe may be deployed in an aqueous solution containing salts (e.g., NaCl), detergents (e.g., SDS), and other components (e.g., Denhardt's solution, dry milk, salmon sperm DNA, etc.). [0037]
  • The phrase “consensus sequence,” as used herein, refers to a nucleic acid sequence which has been resequenced to resolve uncalled bases, extended using XL-PCR (Perkin Elmer, Norwalk, Conn.) in the 5′ and/or the 3′ direction, and resequenced, or which has been assembled from the overlapping sequences of more than one Incyte Clone using a computer program for fragment assembly, such as the GELVIEW Fragment Assembly system (GCG, Madison, Wis.). Some sequences have been both extended and assembled to produce the consensus sequence. [0038]
  • As used herein, the term “correlates with expression of a polynucleotide” indicates that the detection of the presence of nucleic acids, the same or related to a nucleic acid sequence encoding HLHLP, by northern analysis is indicative of the presence of nucleic acids encoding HLHLP in a sample, and thereby correlates with expression of the transcript from the polynucleotide encoding HLHLP. [0039]
  • A “deletion,” as the term is used herein, refers to a change in the amino acid or nucleotide sequence that results in the absence of one or more amino acid residues or nucleotides. [0040]
  • The term “derivative,” as used herein, refers to the chemical modification of HLHLP, of a polynucleotide sequence encoding HLHLP, or of a polynucleotide sequence complementary to a polynucleotide sequence encoding HLHLP. Chemical modifications of a polynucleotide sequence can include, for example, replacement of hydrogen by an alkyl, acyl, or amino group. A derivative polynucleotide encodes a polypeptide which retains at least one biological or immunological function of the natural molecule. A derivative polypeptide is one modified by glycosylation, pegylation, or any similar process that retains a at least one biological or immunological function of the polypeptide from which it was derived. [0041]
  • The term “homology,” as used herein, refers to a degree of complementarity. There may be partial homology or complete homology. The word “identity” may substitute for the word “homology.” A partially complementary sequence that at least partially inhibits an identical sequence from hybridizing to a target nucleic acid is referred to as “substantially homologous.” The inhibition of hybridization of the completely complementary sequence to the target sequence may be examined using a hybridization assay (Southern or northern blot, solution hybridization, and the like) under conditions of reduced stringency. A substantially homologous sequence or hybridization probe will compete for and inhibit the binding of a completely homologous sequence to the target sequence under conditions of reduced stringency. This is not to say that conditions of reduced stringency are such that non-specific binding is permitted, as reduced stringency conditions require that the binding of two sequences to one another be a specific (i.e., a selective) interaction. The absence of non-specific binding may be tested by the use of a second target sequence which lacks even a partial degree of complementarity (e.g., less than about 30% homology or identity). In the absence of non-specific binding, the substantially homologous sequence or probe will not hybridize to the second non-complementary target sequence. [0042]
  • “Human artificial chromosomes” (HACs), as described herein, are linear microchromosomes which may contain DNA sequences of about 10 K to 10 M in size, and which contain all of the elements required for stable mitotic chromosome segregation and maintenance. (Harrington, J. J. et al. (1997) Nat Genet. 15:345-355.) [0043]
  • The term “humanized antibody,” as used herein, refers to antibody molecules in which the amino acid sequence in the non-antigen binding regions has been altered so that the antibody more closely resembles a human antibody, and still retains its original binding ability. [0044]
  • “Hybridization,” as the term is used herein, refers to any process by which a strand of nucleic acid binds with a complementary strand through base pairing. [0045]
  • As used herein, the term “hybridization complex” as used herein, refers to a complex formed between two nucleic acid sequences by virtue of the formation of hydrogen bonds between complementary bases. A hybridization complex may be formed in solution (e.g., C[0046] 0t or R0t analysis) or formed between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized on a solid support (e.g., paper, membranes, filters, chips, pins or glass slides, or any other appropriate substrate to which cells or their nucleic acids have been fixed).
  • The words “insertion” or “addition,” as used herein, refer to changes in an amino acid or nucleotide sequence resulting in the addition of one or more amino acid residues or nucleotides, respectively, to the sequence found in the naturally occurring molecule. [0047]
  • The term “microarray,” as used herein, refers to an array of distinct polynucleotides or oligonucleotides arrayed on a substrate, such as paper, nylon or any other type of membrane, filter, chip, glass slide, or any other suitable solid support. [0048]
  • The term “modulate,” as it appears herein, refers to a change in the activity of HLHLP. For example, modulation may cause an increase or a decrease in protein activity, binding characteristics, or any other biological, functional, or immunological properties of HLHLP. [0049]
  • The phrases “nucleic acid” or “nucleic acid sequence,” as used herein, refer to an oligonucleotide, nucleotide, polynucleotide, or any fragment thereof, to DNA or RNA of genomic or synthetic origin which may be single-stranded or double-stranded and may represent the sense or the antisense strand, to peptide nucleic acid (PNA), or to any DNA-like or RNA-like material. In this context, “fragments” refers to those nucleic acid sequences which are greater than about 60 nucleotides in length, and most preferably are at least about 100 nucleotides, at least about 1000 nucleotides, or at least about 10,000 nucleotides in length. [0050]
  • The term “oligonucleotide,” as used herein, refers to a nucleic acid sequence of at least about 6 nucleotides to 60 nucleotides, preferably about 15 to 30 nucleotides, and most preferably about 20 to 25 nucleotides, which can be used in PCR amplification or in a hybridization assay or microarray. As used herein, the term “oligonucleotide” is substantially equivalent to the terms “amplimers,” “primers,” “oligomers,” and “probes,” as these terms are commonly defined in the art. [0051]
  • “Peptide nucleic acid” (PNA), as used herein, refers to an antisense molecule or anti-gene agent which comprises an oligonucleotide of at least about 5 nucleotides in length linked to a peptide backbone of amino acid residues ending in lysine. The terminal lysine confers solubility to the composition. PNAs preferentially bind complementary single stranded DNA and RNA and stop transcript elongation, and may be pegylated to extend their lifespan in the cell. (Nielsen, P. E. et al. (1993) Anticancer Drug Des. 8:53-63. [0052]
  • The term “sample,” as used herein, is used in its broadest sense. A biological sample suspected of containing nucleic acids encoding HLHLP, or fragments thereof, or HLHLP itself may comprise a bodily fluid; an extract from a cell, chromosome, organelle, or membrane isolated from a cell; a cell; genomic DNA, RNA, or cDNA (in solution or bound to a solid support); a tissue; a tissue print; and the like. [0053]
  • As used herein, the terms “specific binding” or “specifically binding” refer to that interaction between a protein or peptide and an agonist, an antibody, or an antagonist. The interaction is dependent upon the presence of a particular structure of the protein recognized by the binding molecule (i.e., the antigenic determinant or epitope). For example, if an antibody is specific for epitope “A,” the presence of a polypeptide containing the epitope A, or the presence of free unlabeled A, in a reaction containing free labeled A and the antibody will reduce the amount of labeled A that binds to the antibody. [0054]
  • As used herein, the term “stringent conditions” refers to conditions which permit hybridization between polynucleotide sequences and the claimed polynucleotide sequences. Suitably stringent conditions can be defined by, for example, the concentrations of salt or formamide in the prehybridization and hybridization solutions, or by the hybridization temperature, and are well known in the art. In particular, stringency can be increased by reducing the concentration of salt, increasing the concentration of formamide, or raising the hybridization temperature. [0055]
  • For example, hybridization under high stringency conditions could occur in about 50% formamide at about 37° C. to 42° C. Hybridization could occur under reduced stringency conditions in about 35% to 25% formamide at about 30° C. to 35° C. In particular, hybridization could occur under high stringency conditions at 42° C. in 50% formamide, 5× SSPE, 0.3% SDS, and 200μg/ml sheared and denatured salmon sperm DNA. Hybridization could occur under reduced stringency conditions as described above, but in 35% formamide at a reduced temperature of 35° C. The temperature range corresponding to a particular level of stringency can be further narrowed by calculating the purine to pyrimidine ratio of the nucleic acid of interest and adjusting the temperature accordingly. Variations on the above ranges and conditions are well known in the art. [0056]
  • The term “substantially purified,” as used herein, refers to nucleic acid or amino acid sequences that are removed from their natural environment and are isolated or separated, and are at least about 60% free, preferably about 75% free, and most preferably about 90% free from other components with which they are naturally associated. [0057]
  • A “substitution,” as used herein, refers to the replacement of one or more amino acids or nucleotides by different amino acids or nucleotides, respectively. [0058]
  • “Transformation,” as defined herein, describes a process by which exogenous DNA enters and changes a recipient cell. Transformation may occur under natural or artificial conditions according to various methods well known in the art, and may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method for transformation is selected based on the type of host cell being transformed and may include, but is not limited to, viral infection, electroporation, heat shock, lipofection, and particle bombardment. The term “transformed” cells includes stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome, and refers to cells which transiently express the inserted DNA or RNA for limited periods of time. [0059]
  • A “variant” of HLHLP, as used herein, refers to an amino acid sequence that is altered by one or more amino acids. The variant may have “conservative” changes, wherein a substituted amino acid has similar structural or chemical properties (e.g., replacement of leucine with isoleucine). More rarely, a variant may have “nonconservative” changes (e.g., replacement of glycine with tryptophan). Analogous minor variations may also include amino acid deletions or insertions, or both. Guidance in determining which amino acid residues may be substituted, inserted, or deleted without abolishing biological or immunological activity may be found using computer programs well known in the art, for example, DNASTAR software. [0060]
  • The Invention [0061]
  • The invention is based on the discovery of a new human lysyl hydroxylase-like protein (HLHLP), the polynucleotides encoding HLHLP, and the use of these compositions for the diagnosis, treatment, or prevention of cancer and connective tissue disorders. [0062]
  • Nucleic acids encoding the HLHLP of the present invention were first identified in [0063] Incyte Clone 1622313 from the brain tumor cDNA library (BRAITUT13) using a computer search for amino acid sequence alignments. A consensus sequence, SEQ ID NO:2, was derived from the following overlapping and/or extended nucleic acid sequences: Incyte Clones 1622313 (BRAITUT13), 3216326 (TESTNOT07), 1484314 and 1484611 (CORPNOT02), 1436129 (PANCNOT08), 669328 (CRBLNOT01), 759207 (BRAITUT02), 1820044 (GBLATUT01), 685231 (UTRSNOT02), 1995161 (BRSTTUT03), 2058591 (OVARNOT03).
  • In one embodiment, the invention encompasses a polypeptide comprising the amino acid sequence of SEQ ID NO:1. As shown in FIGS. 1A, 1B, [0064] 1C, 1D, 1E, 1F, and 1G, HLHLP is 738 amino acids in length and has potential glycosylation sites at residues N63 and N548. HLHLP also has potential casein kinase II phosphorylation sites at residues S23, S121, S367, S433, S483, S485, S487, S496 S519 and S734. HLHLP also has potential protein kinase C phosphorylation sites at S25, T65, T159, T402, T471, S496, T518, S663, and S702, and potential tyrosine kinase phosphorylation sites at Y57 and Y380. As shown in FIGS. 2A, 2B, 2C, and 2D, HLHLP has chemical and structural homology with lysyl hydroxylase isoform 2 (PLOD2) (GI 2138314; SEQ ID NO:3), and with lysyl hydroxylase (PLOD) (GI 190074; SEQ ID NO:4). In particular, HLHLP and PLOD2 share 58% identity, while HLHLP and PLOD share 57% identity. Several potential phosphorylation sites for protein kinase C and for casein kinase II are shared among HLHLP, PLOD2, and PLOD. In addition, HLHLF, PLOD2, and PLOD share the potential tyrosine kinase phosphorylation sites as well as the glycosylation sites. Northern analysis shows the expression of this sequence in various cells and tissues, at least 57% of which are immortalized or cancerous. Of particular note is the expression of HLHLP in several tumors including brain, prostate, breast, and paraganglion.
  • The invention also encompasses HLHLP variants. A preferred HLHLP variant is one having at least about 80%, more preferably at least about 90%, and most preferably at least about 95% amino acid sequence identity to the HLHLP amino acid sequence. [0065]
  • The invention also encompasses polynucleotides which encode HLHLP. In a particular embodiment, the invention encompasses a polynucleotide sequence comprising the sequence of SEQ ID NO:2, which encodes an HLHLP. [0066]
  • The invention also encompasses a variant of a polynucleotide sequence encoding HLHLP. In particular, such a variant polynucleotide sequence will have at least about 80%, more preferably at least about 90%, and most preferably at least about 95% polynucleotide sequence identity to the polynucleotide sequence encoding HLHLP. A particular aspect of the invention encompasses a variant of SEQ ID NO:2 which has at least about 80%, more preferably at least about 90%, and most preferably at least about 95% polynucleotide sequence identity to SEQ ID NO:2. [0067]
  • It will be appreciated by those skilled in the art that as a result of the degeneracy of the genetic code, a multitude of polynucleotide sequences encoding HLHLP, some bearing minimal homology to the polynucleotide sequences of any known and naturally occurring gene, may be produced. Thus, the invention contemplates each and every possible variation of polynucleotide sequence that could be made by selecting combinations based on possible codon choices. These combinations are made in accordance with the standard triplet genetic code as applied to the polynucleotide sequence of naturally occurring HLHLP, and all such variations are to be considered as being specifically disclosed. [0068]
  • Although nucleotide sequences which encode HLHLP and its variants are preferably capable of hybridizing to the nucleotide sequence of the naturally occurring HLHLP under appropriately selected conditions of stringency, it may be advantageous to produce nucleotide sequences encoding HLHLP or its derivatives possessing a substantially different codon usage. Codons may be selected to increase the rate at which expression of the peptide occurs in a particular prokaryotic or eukaryotic host in accordance with the frequency with which particular codons are utilized by the host. Other reasons for substantially altering the nucleotide sequence encoding HLHLP and its derivatives without altering the encoded amino acid sequences include the production of RNA transcripts having more desirable properties, such as a greater half-life, than transcripts produced from the naturally occurring sequence. [0069]
  • The invention also encompasses production of DNA sequences which encode HLHLP and HLHLP derivatives, or fragments thereof, entirely by synthetic chemistry. After production, the synthetic sequence may be inserted into any of the many available expression vectors and cell systems using reagents that are well known in the art. Moreover, synthetic chemistry may be used to introduce mutations into a sequence encoding HLHLP or any fragment thereof. [0070]
  • Also encompassed by the invention are polynucleotide sequences that are capable of hybridizing to the claimed polynucleotide sequences, and, in particular, to those shown in SEQ ID NO:2, or a fragment of SEQ ID NO:2, under various conditions of stringency as taught in Wahl, G. M. and S. L. Berger (1987; Methods Enzymol. 152:399-407) and Kimmel, A. R. (1987; Methods Enzymol. 152:507-511). [0071]
  • Methods for DNA sequencing are well known and generally available in the art and may be used to practice any of the embodiments of the invention. The methods may employ such enzymes as the Klenow fragment of DNA polymerase I, SEQUENASE (US Biochemical Corp., Cleveland, Ohio), Taq DNA polymerase (Perkin Elmer), thermostable T7 polymerase (Amersham, Chicago, Ill.), or combinations of polymerases and proofreading exonucleases such as those found in the ELONGASE Amplification System marketed by GIBCO/BRL (Gaithersburg, Md.). Preferably, the process is automated with machines such as the MICROLAB 2200 (Hamilton, Reno, Nev.), DNA ENGINE thermal cycler (PTC200; MJ Research, Watertown, Mass.) and the ABI Catalyst and ABI PRISM sequencing systems (PE Biosystems. [0072]
  • The nucleic acid sequences encoding HLHLP may be extended utilizing a partial nucleotide sequence and employing various methods known in the art to detect upstream sequences, such as promoters and regulatory elements. For example, one method which may be employed, restriction-site PCR, uses universal primers to retrieve unknown sequence adjacent to a known locus. (Sarkar, G. (1993) PCR Methods Applic. 2:318-322.) In particular, genomic DNA is first amplified in the presence of a primer to a linker sequence and a primer specific to the known region. The amplified sequences are then subjected to a second round of PCR with the same linker primer and another specific primer internal to the first one. Products of each round of PCR are transcribed with an appropriate RNA polymerase and sequenced using reverse transcriptase. [0073]
  • Inverse PCR may also be used to amplify or extend sequences using divergent primers based on a known region. (Triglia, T. et al. (1988) Nucleic Acids Res. 16:8186.) The primers may be designed using commercially available software such as OLIGO 4.06 Primer Analysis software (National Biosciences Inc., Plymouth, Minn.) or another appropriate program to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the target sequence at temperatures of about 68° C. to 72° C. The method uses several restriction enzymes to generate a suitable fragment in the known region of a gene. The fragment is then circularized by intramolecular ligation and used as a PCR template. [0074]
  • Another method which may be used is capture PCR, which involves PCR amplification of DNA fragments adjacent to a known sequence in human and yeast artificial chromosome DNA. (Lagerstrom, M. et al. (1991) PCR Methods Applic. 1:111-119.) In this method, multiple restriction enzyme digestions and ligations may be used to place an engineered double-stranded sequence into an unknown fragment of the DNA molecule before performing PCR. Another method which may be used to retrieve unknown sequences is that of Parker, J. D. et al. (1991; Nucleic Acids Res. 19:3055-3060). Additionally, one may use PCR, nested primers, and PROMOTERFINDER libraries (Clontech, Palo Alto, Calif.) to walk genomic DNA. This process avoids the need to screen libraries and is useful in finding intron/exon junctions. [0075]
  • When screening for full-length cDNAs, it is preferable to use libraries that have been size-selected to include larger cDNAs. Also, random-primed libraries are preferable in that they will include more sequences which contain the 5′ regions of genes. Use of a randomly primed library may be especially preferable for situations in which an oligo d(T) library does not yield a full-length cDNA. Genomic libraries may be useful for extension of sequence into 5′ non-transcribed regulatory regions. [0076]
  • Capillary electrophoresis systems which are commercially available may be used to analyze the size or confirm the nucleotide sequence of sequencing or PCR products. In particular, capillary sequencing may employ flowable polymers for electrophoretic separation, four different fluorescent dyes (one for each nucleotide) which are laser activated, and a charge coupled device camera for detection of the emitted wavelengths. Output/light intensity may be converted to electrical signal using appropriate software (e.g., GENOTYPER and SEQUENCE NAVIGATOR, PE Biosystems), and the entire process from loading of samples to computer analysis and electronic data display may be computer controlled. Capillary electrophoresis is especially preferable for the sequencing of small pieces of DNA which might be present in limited amounts in a particular sample. [0077]
  • In another embodiment of the invention, polynucleotide sequences or fragments thereof which encode HLHLP may be used in recombinant DNA molecules to direct expression of HLHLP, or fragments or functional equivalents thereof, in appropriate host cells. Due to the inherent degeneracy of the genetic code, other DNA sequences which encode substantially the same or a functionally equivalent amino acid sequence may be produced, and these sequences may be used to clone and express HLHLP. [0078]
  • As will be understood by those of skill in the art, it may be advantageous to produce HLHLP-encoding nucleotide sequences possessing non-naturally occurring codons. For example, codons preferred by a particular prokaryotic or eukaryotic host can be selected to increase the rate of protein expression or to produce an RNA transcript having desirable properties, such as a half-life which is longer than that of a transcript generated from the naturally occurring sequence. [0079]
  • The nucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter HLHLP encoding sequences for a variety of reasons including, but not limited to, alterations which modify the cloning, processing, and/or expression of the gene product. DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides may be used to engineer the nucleotide sequences. For example, site-directed mutagenesis may be used to insert new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, introduce mutations, and so forth. [0080]
  • In another embodiment of the invention, natural, modified, or recombinant nucleic acid sequences encoding HLHLP may be ligated to a heterologous sequence to encode a fusion protein. For example, to screen peptide libraries for inhibitors of HLHLP activity, it may be useful to encode a chimeric HLHLP protein that can be recognized by a commercially available antibody. A fusion protein may also be engineered to contain a cleavage site located between the HLHLP encoding sequence and the heterologous protein sequence, so that HLHLP may be cleaved and purified away from the heterologous moiety. [0081]
  • In another embodiment, sequences encoding HLHLP may be synthesized, in whole or in part, using chemical methods well known in the art. (See Caruthers, M. H. et al. (1980) Nucl. Acids Symp. Ser. (7):215-223, and Horn, T. et al. (1980) Nucl. Acids Symp. Ser. (7):225-232.) Alternatively, the protein itself may be produced using chemical methods to synthesize the amino acid sequence of HLHLP, or a fragment thereof. For example, peptide synthesis can be performed using various solid-phase techniques (Roberge, J. Y. et al. (1995) Science 269:202-204) and automated synthesis may be achieved using the ABI 431A Peptide synthesizer (PE Biossystems). [0082]
  • The newly synthesized peptide may be substantially purified by preparative high performance liquid chromatography. (See, for example, Creighton, T. (1983) [0083] Proteins, Structures and Molecular Principles, W H Freeman and Co., New York, N.Y.) The composition of the synthetic peptides may be confirmed by amino acid analysis or by sequencing. (See, for example, the Edman degradation procedure described in Creighton, supra.) Additionally, the amino acid sequence of HLHLP, or any part thereof, may be altered during direct synthesis and/or combined with sequences from other proteins, or any part thereof, to produce a variant polypeptide.
  • In order to express a biologically active HLHLP, the nucleotide sequences encoding HLHLP or derivatives thereof may be inserted into appropriate expression vector, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted coding sequence. [0084]
  • Methods which are well known to those skilled in the art may be used to construct expression vectors containing sequences encoding HLHLP and appropriate transcriptional and translational control elements. These methods include, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. Such techniques are described in Sambrook, J. et al. (1989[0085] ; Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y.) and Ausubel, F. M. et al. (1989; Current Protocols in Molecular Biology, John Wiley & Sons, New York, N.Y.).
  • A variety of expression vector/host systems may be utilized to contain and express sequences encoding HLHLP. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus (CaMV) or tobacco mosaic virus (TMV)) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids); or animal cell systems. [0086]
  • The invention is not limited by the host cell employed. [0087]
  • The “control elements” or “regulatory sequences” are those non-translated regions of the vector (i.e., enhancers, promoters, and 5′ and 3′ untranslated regions) which interact with host cellular proteins to carry out transcription and translation. Such elements may vary in their strength and specificity. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used. For example, when cloning in bacterial systems, inducible promoters such as the hybrid lacZ promoter of the BLUESCRIPT phagemid (Stratagene, La Jolla, Calif.) or PSPORT1 plasmid (GIBCO/BRL), and the like, may be used. The baculovirus polyhedrin promoter may be used in insect cells. Promoters or enhancers derived from the genomes of plant cells (e.g., heat shock, RUBISCO, and storage protein genes) or from plant viruses (e.g., viral promoters or leader sequences) may be cloned into the vector. In mammalian cell systems, promoters from mammalian genes or from mammalian viruses are preferable. If it is necessary to generate a cell line that contains multiple copies of the sequence encoding HLHLP, vectors based on SV40 or EBV may be used with an appropriate selectable marker. [0088]
  • In bacterial systems, a number of expression vectors may be selected depending upon the use intended for HLHLP. For example, when large quantities of HLHLP are needed for the induction of antibodies, vectors which direct high level expression of fusion proteins that are readily purified may be used. Such vectors include, but are not limited to, multifunctional [0089] E. coli cloning and expression vectors such as BLUESCRIPT (Stratagene), in which the sequence encoding HLHLP may be ligated into the vector in frame with sequences for the amino-terminal Met and the subsequent 7 residues of β-galactosidase so that a hybrid protein is produced, pIN vectors (Van Heeke, G. and S. M. Schuster (1989) J. Biol. Chem. 264:5503-5509), and the like. pGEX vectors (Promega, Madison, Wis.) may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione. Proteins made in such systems may be designed to include heparin, thrombin, or factor XA protease cleavage sites so that the cloned polypeptide of interest can be released from the GST moiety at will.
  • In the yeast [0090] Saccharomyces cerevisiae, a number of vectors containing constitutive or inducible promoters, such as alpha factor, alcohol oxidase, and PGH, may be used. For reviews, see Ausubel (supra) and Grant et al. (1987; Methods Enzymol. 153:516-544).
  • In cases where plant expression vectors are used, the expression of sequences encoding HLHLP may be driven by any of a number of promoters. For example, viral promoters such as the 35S and 19S promoters of CaMV may be used alone or in combination with the omega leader sequence from TMV. (Takamatsu, N. (1987) EMBO J. 6:307-311.) Alternatively, plant promoters such as the small subunit of RUBISCO or heat shock promoters may be used. (Coruzzi, G. et al. (1984) EMBO J. 3:1671-1680; Broglie, R. et al. (1984) Science 224:838-843; and Winter, J. et al. (1991) Results Probl. Cell Differ. 17:85-105.) These constructs can be introduced into plant cells by direct DNA transformation or pathogen-mediated transfection. Such techniques are described in a number of generally available reviews. (See, for example, Hobbs, S. or Murry, L. E. in McGraw Hill Yearbook of Science and Technology (1992) McGraw Hill, New York, N.Y.; pp. 191-196.) [0091]
  • An insect system may also be used to express HLHLP. For example, in one such system, [0092] Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae. The sequences encoding HLHLP may be cloned into a non-essential region of the virus, such as the polyhedrin gene, and placed under control of the polyhedrin promoter. Successful insertion of HLHLP will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein. The recombinant viruses may then be used to infect, for example, S. frugiperda cells or Trichoplusia larvae in which HLHLP may be expressed. (Engelhard, E. K. et al. (1994) Proc. Nat. Acad. Sci. 91:3224-3227.)
  • In mammalian host cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, sequences encoding HLHLP may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential E1 or E3 region of the viral genome may be used to obtain a viable virus which is capable of expressing HLHLP in infected host cells. (Logan, J. and T. Shenk (1984) Proc. Natl. Acad. Sci. 81:3655-3659.) In addition, transcription enhancers, such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells. [0093]
  • Human artificial chromosomes (HACs) may also be employed to deliver larger fragments of DNA than can be contained and expressed in a plasmid. HACs of about 6 M to 10 M are constructed and delivered via conventional delivery methods (liposomes, polycationic amino polymers, or vesicles) for therapeutic purposes. [0094]
  • Specific initiation signals may also be used to achieve more efficient translation of sequences encoding HLHLP. Such signals include the ATG initiation codon and adjacent sequences. In cases where sequences encoding HLHLP and its initiation codon and upstream sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a fragment thereof, is inserted, exogenous translational control signals including the ATG initiation codon should be provided. Furthermore, the initiation codon should be in the correct reading frame to ensure translation of the entire insert. Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers appropriate for the particular cell system used, such as those described in the literature. (Scharf, D. et al. (1994) Results Probl. Cell Differ. 20:125-162.) [0095]
  • In addition, a host cell strain may be chosen for its ability to modulate expression of the inserted sequences or to process the expressed protein in the desired fashion. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation. Post-translational processing which cleaves a “prepro” form of the protein may also be used to facilitate correct insertion, folding, and/or function. Different host cells which have specific cellular machinery and characteristic mechanisms for post-translational activities (e.g., CHO, HeLa, MDCK, HEK293, and WI38), are available from the American Type Culture Collection (ATCC, Bethesda, Md.) and may be chosen to ensure the correct modification and processing of the foreign protein. [0096]
  • For long term, high yield production of recombinant proteins, stable expression is preferred. For example, cell lines capable of stably expressing HLHLP can be transformed using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells may be allowed to grow for about 1 to 2 days in enriched media before being switched to selective media. The purpose of the selectable marker is to confer resistance to selection, and its presence allows growth and recovery of cells which successfully express the introduced sequences. Resistant clones of stably transformed cells may be proliferated using tissue culture techniques appropriate to the cell type. [0097]
  • Any number of selection systems may be used to recover transformed cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase genes (Wigler, M. et al. (1977) Cell 11:223-32) and adenine phosphoribosyltransferase genes (Lowy, I. et al. (1980) Cell 22:817-23), which can be employed in tk[0098] or aprcells, respectively. Also, antimetabolite, antibiotic, or herbicide resistance can be used as the basis for selection. For example, dhfr confers resistance to methotrexate (Wigler, M. et al. (1980) Proc. Natl. Acad. Sci. 77:3567-70); npt confers resistance to the aminoglycosides neomycin and G-418 (Colbere-Garapin, F. et al (1981) J. Mol. Biol. 150:1-14); and als or pat confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively (Murry, supra). Additional selectable genes have been described, for example, trpB, which allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine. (Hartman, S. C. and R. C. Mulligan (1988) Proc. Natl. Acad. Sci. 85:8047-51.) Recently, the use of visible markers has gained popularity with such markers as anthocyanins, β glucuronidase and its substrate GUS, and luciferase and its substrate luciferin. These markers can be used not only to identify transformants, but also to quantify the amount of transient or stable protein expression attributable to a specific vector system. (Rhodes, C. A. et al. (1995) Methods Mol. Biol. 55:121-131.)
  • Although the presence/absence of marker gene expression suggests that the gene of interest is also present, the presence and expression of the gene may need to be confirmed. For example, if the sequence encoding HLHLP is inserted within a marker gene sequence, transformed cells containing sequences encoding HLHLP can be identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a sequence encoding HLHLP under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well. [0099]
  • Alternatively, host cells which contain the nucleic acid sequence encoding HLHLP and express HLHLP may be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations and protein bioassay or immunoassay techniques which include membrane, solution, or chip based technologies for the detection and/or quantification of nucleic acid or protein sequences. [0100]
  • The presence of polynucleotide sequences encoding HLHLP can be detected by DNA-DNA or DNA-RNA hybridization or amplification using probes or fragments or fragments of polynucleotides encoding HLHLP. Nucleic acid amplification based assays involve the use of oligonucleotides or oligomers based on the sequences encoding HLHLP to detect transformants containing DNA or RNA encoding HLHLP. [0101]
  • A variety of protocols for detecting and measuring the expression of HLHLP, using either polyclonal or monoclonal antibodies specific for the protein, are known in the art. Examples of such techniques include enzyme-linked immunosorbent assays (ELISAs), radioimmunoassays (RIAs), and fluorescence activated cell sorting (FACS). A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on HLHLP is preferred, but a competitive binding assay may be employed. These and other assays are well described in the art, for example, in Hampton, R. et al. (1990[0102] ; Serological Methods, a Laboratory Manual, APS Press, St Paul, Minn.) and in Maddox, D. E. et al. (1983; J. Exp. Med. 158:1211-1216).
  • A wide variety of labels and conjugation techniques are known by those skilled in the art and may be used in various nucleic acid and amino acid assays. Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides encoding HLHLP include oligolabeling, nick translation, end-labeling, or PCR amplification using a labeled nucleotide. Alternatively, the sequences encoding HLHLP, or any fragments thereof, may be cloned into a vector for the production of an mRNA probe. Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by addition of an appropriate RNA polymerase such as T7, T3, or SP6 and labeled nucleotides. These procedures may be conducted using a variety of commercially available kits, such as those provided by Pharmacia & Upjohn (Kalamazoo, Mich.), Promega (Madison, Wis.), and U.S. Biochemical Corp. (Cleveland, Ohio). Suitable reporter molecules or labels which may be used for ease of detection include radionuclides, enzymes, fluorescent, chemiluminescent, or chromogenic agents, as well as substrates, cofactors, inhibitors, magnetic particles, and the like. [0103]
  • Host cells transformed with nucleotide sequences encoding HLHLP may be cultured under conditions suitable for the expression and recovery of the protein from cell culture. The protein produced by a transformed cell may be secreted or contained intracellularly depending on the sequence and/or the vector used. As will be understood by those of skill in the art, expression vectors containing polynucleotides which encode HLHLP may be designed to contain signal sequences which direct secretion of HLHLP through a prokaryotic or eukaryotic cell membrane. Other constructions may be used to join sequences encoding HLHLP to nucleotide sequences encoding a polypeptide domain which will facilitate purification of soluble proteins. Such purification facilitating domains include, but are not limited to, metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp., Seattle, Wash.). The inclusion of cleavable linker sequences, such as those specific for Factor XA or enterokinase (Invitrogen, San Diego, Calif.), between the purification domain and the HLHLP encoding sequence may be used to facilitate purification. One such expression vector provides for expression of a fusion protein containing HLHLP and a nucleic acid encoding 6 histidine residues preceding a thioredoxin or an enterokinase cleavage site. The histidine residues facilitate purification on immobilized metal ion affinity chromatography (IMAC; described in Porath, J. et al. (1992) Prot. Exp. Purif. 3: 263-281)), while the enterokinase cleavage site provides a means for purifying HLHLP from the fusion protein. A discussion of vectors which contain fusion proteins is provided in Kroll, D. J. et al. (1993; DNA Cell Biol. 12:441-453). [0104]
  • Fragments of HLHLP may be produced not only by recombinant production, but also by direct peptide synthesis using solid-phase techniques. (Merrifield J. (1963) J. Am. Chem. Soc. 85:2149-2154.) Protein synthesis may be performed by manual techniques or by automation. Automated synthesis may be achieved, for example, using the ABI 431A Peptide synthesizer (PE Biosystems). Various fragments of HLHLP may be synthesized separately and then combined to produce the full length molecule. [0105]
  • Therapeutics [0106]
  • Chemical and structural homology exists among HLHLP, PLOD2 (GI 2138314) and PLOD (GI 190074). In addition, HLHLP is expressed in numerous cancerous tissues. Therefore, HLHLP appears to play a role in cancer and connective tissue disorders. [0107]
  • Therefore, in one embodiment, an antagonist of HLHLP may be administered to a subject to treat or prevent a cancer. Such cancers may include, but are not limited to, adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus. In one aspect, an antibody which specifically binds HLHLP may be used directly as an antagonist or indirectly as a targeting or delivery mechanism for bringing a pharmaceutical agent to cells or tissue which express HLHLP. [0108]
  • In an additional embodiment, a vector expressing the complement of the polynucleotide encoding HLHLP may be administered to a subject to treat or prevent a cancer including, but not limited to, those described above. [0109]
  • In another embodiment, HLHLP or a fragment or derivative thereof may be administered to a subject to treat or prevent a connective tissue disorder. Such disorders can include, but are not limited to, Ehlers-Danos syndrome type VI, ankylosing spondylitis, atherosclerosis, Dupuytren's contracture, eosinophilic fasciitis, Felty syndrome, Goodpasture's disease, Hunter syndrome, Hurler syndrome, keloids, Marfan syndrome, nodular fasciitis, osteogenesis imperfecta, polyarthritis nodosa, rheumatoid arthritis, scleroderma, systemic lupus erythematosus, and restenosis following angioplasty. [0110]
  • In another embodiment, a vector capable of expressing HLHLP or a fragment or derivative thereof may be administered to a subject to treat or prevent a connective tissue disorder including, but not limited to, those described above. [0111]
  • In a further embodiment, a pharmaceutical composition comprising a substantially purified HLHLP in conjunction with a suitable pharmaceutical carrier may be administered to a subject to treat or prevent a connective tissue disorder including, but not limited to, those provided above. [0112]
  • In still another embodiment, an agonist which modulates the activity of HLHLP may be administered to a subject to treat or prevent a connective tissue disorder including, but not limited to, those listed above. [0113]
  • In other embodiments, any of the proteins, antagonists, antibodies, agonists, complementary sequences, or vectors of the invention may be administered in combination with other appropriate therapeutic agents. Selection of the appropriate agents for use in combination therapy may be made by one of ordinary skill in the art, according to conventional pharmaceutical principles. The combination of therapeutic agents may act synergistically to effect the treatment or prevention of the various disorders described above. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects. [0114]
  • An antagonist of HLHLP may be produced using methods which are generally known in the art. In particular, purified HLHLP may be used to produce antibodies or to screen libraries of pharmaceutical agents to identify those which specifically bind HLHLP. Antibodies to HLHLP may also be generated using methods that are well known in the art. Such antibodies may include, but are not limited to, polyclonal, monoclonal, chimeric, and single chain antibodies, Fab fragments, and fragments produced by a Fab expression library. Neutralizing antibodies (i.e., those which inhibit dimer formation) are especially preferred for therapeutic use. [0115]
  • For the production of antibodies, various hosts including goats, rabbits, rats, mice, humans, and others may be immunized by injection with HLHLP or with any fragment or oligopeptide thereof which has immunogenic properties. Depending on the host species, various adjuvants may be used to increase immunological response. Such adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, KLH, and dinitrophenol. Among adjuvants used in humans, BCG (bacilli Calmette-Guerin) and [0116] Corynebacterium parvum are especially preferable.
  • It is preferred that the oligopeptides, peptides, or fragments used to induce antibodies to HLHLP have an amino acid sequence consisting of at least about 5 amino acids, and, more preferably, of at least about 10 amino acids. It is also preferable that these oligopeptides, peptides, or fragments are identical to a portion of the amino acid sequence of the natural protein and contain the entire amino acid sequence of a small, naturally occurring molecule. Short stretches of HLHLP amino acids may be fused with those of another protein, such as KLH, and antibodies to the chimeric molecule may be produced. [0117]
  • Monoclonal antibodies to HLHLP may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique. (Kohler, G. et al. (1975) Nature 256:495-497; Kozbor, D. et al. (1985) J. Immunol. Methods 81:31-42; Cote, R. J. et al. (1983) Proc. Natl. Acad. Sci. 80:2026-2030; and Cole, S. P. et al. (1984) Mol. Cell Biol. 62:109-120.) [0118]
  • In addition, techniques developed for the production of “chimeric antibodies,” such as the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used. (Morrison, S. L. et al. (1984) Proc. Natl. Acad. Sci. 81:6851-6855; Neuberger, M. S. et al. (1984) Nature 312:604-608; and Takeda, S. et al. (1985) Nature 314:452-454.) Alternatively, techniques described for the production of single chain antibodies may be adapted, using methods known in the art, to produce HLHLP-specific single chain antibodies. Antibodies with related specificity, but of distinct idiotypic composition, may be generated by chain shuffling from random combinatorial immunoglobulin libraries. (Burton D. R. (1991) Proc. Natl. Acad. Sci. 88:11120-11123.) [0119]
  • Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature. (Orlandi, R. et al. (1989) Proc. Natl. Acad. Sci. 86: 3833-3837, and Winter, G. et al. (1991) Nature 349:293-299.) [0120]
  • Antibody fragments which contain specific binding sites for HLHLP may also be generated. For example, such fragments include, but are not limited to, F(ab′)2 fragments produced by pepsin digestion of the antibody molecule and Fab fragments generated by reducing the disulfide bridges of the F(ab′)2 fragments. Alternatively, Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity. (Huse, W. D. et al. (1989) Science 254:1275-1281.) [0121]
  • Various immunoassays may be used for screening to identify antibodies having the desired specificity. Numerous protocols for competitive binding or immunoradiometric assays using either polyclonal or monoclonal antibodies with established specificities are well known in the art. Such immunoassays typically involve the measurement of complex formation between HLHLP and its specific antibody. A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering HLHLP epitopes is preferred, but a competitive binding assay may also be employed. (Maddox, supra.) [0122]
  • In another embodiment of the invention, the polynucleotides encoding HLHLP, or any fragment or complement thereof, may be used for therapeutic purposes. In one aspect, the complement of the polynucleotide encoding HLHLP may be used in situations in which it would be desirable to block the transcription of the mRNA. In particular, cells may be transformed with sequences complementary to polynucleotides encoding HLHLP. Thus, complementary molecules or fragments may be used to modulate HLHLP activity, or to achieve regulation of gene function. Such technology is now well known in the art, and sense or antisense oligonucleotides or larger fragments can be designed from various locations along the coding or control regions of sequences encoding HLHLP. [0123]
  • Expression vectors derived from retroviruses, adenoviruses, or herpes or vaccinia viruses, or from various bacterial plasmids, may be used for delivery of nucleotide sequences to the targeted organ, tissue, or cell population. Methods which are well known to those skilled in the art can be used to construct vectors which will express nucleic acid sequence complementary to the polynucleotides of the gene encoding HLHLP. These techniques are described, for example, in Sambrook (supra) and in Ausubel (supra). [0124]
  • Genes encoding HLHLP can be turned off by transforming a cell or tissue with expression vectors which express high levels of a polynucleotide or fragment thereof encoding HLHLP. Such constructs may be used to introduce untranslatable sense or antisense sequences into a cell. Even in the absence of integration into the DNA, such vectors may continue to transcribe RNA molecules until they are disabled by endogenous nucleases. Transient expression may last for a month or more with a non-replicating vector, and may last even longer if appropriate replication elements are part of the vector system. [0125]
  • As mentioned above, modifications of gene expression can be obtained by designing complementary sequences or antisense molecules (DNA, RNA, or PNA) to the control, 5′, or regulatory regions of the gene encoding HLHLP. Oligonucleotides derived from the transcription initiation site, e.g., between about positions −10 and +10 from the start site, are preferred. Similarly, inhibition can be achieved using triple helix base-pairing methodology. Triple helix pairing is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or regulatory molecules. Recent therapeutic advances using triplex DNA have been described in the literature. (Gee, J. E. et al. (1994) in Huber, B. E. and B. I. Carr, [0126] Molecular and Immunologic Approaches, Futura Publishing Co., Mt. Kisco, N.Y.) A complementary sequence or antisense molecule may also be designed to block translation of mRNA by preventing the transcript from binding to ribosomes.
  • Ribozymes, enzymatic RNA molecules, may also be used to catalyze the specific cleavage of RNA. The mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage. For example, engineered hammerhead motif ribozyme molecules specifically and efficiently catalyze endonucleolytic cleavage of sequences encoding HLHLP. [0127]
  • Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, including the following sequences: GUA, GUU, and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site may be evaluated for secondary structural features which may render the oligonucleotide inoperable. The suitability of candidate targets may also be evaluated by testing accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays. [0128]
  • Complementary ribonucleic acid molecules and ribozymes of the invention may be prepared by any method known in the art for the synthesis of nucleic acid molecules. These include techniques for chemically synthesizing oligonucleotides such as solid phase phosphoramidite chemical synthesis. Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding HLHLP. Such DNA sequences may be incorporated into a wide variety of vectors with suitable RNA polymerase promoters such as T7 or SP6. Alternatively, these cDNA constructs that synthesize complementary RNA constitutively or inducibly can be introduced into cell lines, cells, or tissues. [0129]
  • RNA molecules may be modified to increase intracellular stability and half-life. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5′ and/or 3′ ends of the molecule or the use of phosphorothioate or 2′ O-methyl rather than phosphodiesterase linkages within the backbone of the molecule. This concept is inherent in the production of PNAs and can be extended in all of these molecules by the inclusion of nontraditional bases such as inosine, queosine, and wybutosine, as well as acetyl-, methyl-, thio-, and similarly modified forms of adenine, cytidine, guanine, thymine, and uridine which are not as easily recognized by endogenous endonucleases. [0130]
  • Many methods for introducing vectors into cells or tissues are available and equally suitable for use in vivo, in vitro, and ex vivo. For ex vivo therapy, vectors may be introduced into stem cells taken from the patient and clonally propagated for autologous transplant back into that same patient. Delivery by transfection, by liposome injections, or by polycationic amino polymers may be achieved using methods which are well known in the art, such as those described in Goldman, C. K. et al. (1997; Nature Biotechnology 15:462-466). [0131]
  • Any of the therapeutic methods described above may be applied to any subject in need of such therapy, including, for example, mammals such as dogs, cats, cows, horses, rabbits, monkeys, and most preferably, humans. [0132]
  • An additional embodiment of the invention relates to the administration of a pharmaceutical composition, in conjunction with a pharmaceutically acceptable carrier, for any of the therapeutic effects discussed above. Such pharmaceutical compositions may consist of HLHLP, antibodies to HLHLP, and mimetics, agonists, antagonists, or inhibitors of HLHLP. The compositions may be administered alone or in combination with at least one other agent, such as a stabilizing compound, which may be administered in any sterile, biocompatible pharmaceutical carrier including, but not limited to, saline, buffered saline, dextrose, and water. The compositions may be administered to a patient alone, or in combination with other agents, drugs or hormones. [0133]
  • The pharmaceutical compositions utilized in this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, or rectal means. [0134]
  • In addition to the active ingredients, these pharmaceutical compositions may contain suitable pharmaceutically-acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Further details on techniques for formulation and administration may be found in the latest edition of Remington's Pharmaceutical Sciences (Maack Publishing Co., Easton, Pa.). [0135]
  • Pharmaceutical compositions for oral administration can be formulated using pharmaceutically acceptable carriers well known in the art in dosages suitable for oral administration. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for ingestion by the patient. [0136]
  • Pharmaceutical preparations for oral use can be obtained through combining active compounds with solid excipient and processing the resultant mixture of granules (optionally, after grinding) to obtain tablets or dragee cores. Suitable auxiliaries can be added, if desired. Suitable excipients include carbohydrate or protein fillers, such as sugars, including lactose, sucrose, mannitol, and sorbitol; starch from corn, wheat, rice, potato, or other plants; cellulose, such as methyl cellulose, hydroxypropylmethyl-cellulose, or sodium carboxymethylcellulose; gums, including arabic and tragacanth; and proteins, such as gelatin and collagen. If desired, disintegrating or solubilizing agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, and alginic acid or a salt thereof, such as sodium alginate. [0137]
  • Dragee cores may be used in conjunction with suitable coatings, such as concentrated sugar solutions, which may also contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for product identification or to characterize the quantity of active compound, i.e., dosage. [0138]
  • Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a coating, such as glycerol or sorbitol. Push-fit capsules can contain active ingredients mixed with fillers or binders, such as lactose or starches, lubricants, such as talc or magnesium stearate, and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid, or liquid polyethylene glycol with or without stabilizers. [0139]
  • Pharmaceutical formulations suitable for parenteral administration may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiologically buffered saline. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils, such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate, triglycerides, or liposomes. Non-lipid polycationic amino polymers may also be used for delivery. Optionally, the suspension may also contain suitable stabilizers or agents to increase the solubility of the compounds and allow for the preparation of highly concentrated solutions. [0140]
  • For topical or nasal administration, penetrants appropriate to the particular barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. [0141]
  • The pharmaceutical compositions of the present invention may be manufactured in a manner that is known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes. [0142]
  • The pharmaceutical composition may be provided as a salt and can be formed with many acids, including but not limited to, hydrochloric, sulfuric, acetic, lactic, tartaric, malic, and succinic acid. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free base forms. In other cases, the preferred preparation may be a lyophilized powder which may contain any or all of the following: 1 mM to 50 mM histidine, 0.1% to 2% sucrose, and 2% to 7% mannitol, at a pH range of 4.5 to 5.5, that is combined with buffer prior to use. [0143]
  • After pharmaceutical compositions have been prepared, they can be placed in an appropriate container and labeled for treatment of an indicated condition. For administration of HLHLP, such labeling would include amount, frequency, and method of administration. [0144]
  • Pharmaceutical compositions suitable for use in the invention include compositions wherein the active ingredients are contained in an effective amount to achieve the intended purpose. The determination of an effective dose is well within the capability of those skilled in the art. [0145]
  • For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays of neoplastic cells, for example, or in animal models, usually mice, rabbits, dogs, or pigs. An animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. [0146]
  • A therapeutically effective dose refers to that amount of active ingredient, for example HLHLP or fragments thereof, antibodies of HLHLP, and agonists, antagonists or inhibitors of HLHLP, which ameliorates the symptoms or condition. Therapeutic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or with experimental animals, such as by calculating the ED50 (the dose therapeutically effective in 50% of the population) or LD50 (the dose lethal to 50% of the population) statistics. The dose ratio between therapeutic and toxic effects is the therapeutic index, and it can be expressed as the LD50/ED50 ratio. Pharmaceutical compositions which exhibit large therapeutic indices are preferred. The data obtained from cell culture assays and animal studies is used in formulating a range of dosage for human use. The dosage contained in such compositions is preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, the sensitivity of the patient, and the route of administration. [0147]
  • The exact dosage will be determined by the practitioner, in light of factors related to the subject requiring treatment. Dosage and administration are adjusted to provide sufficient levels of the active moiety or to maintain the desired effect. Factors which may be taken into account include the severity of the disease state, the general health of the subject, the age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. Long-acting pharmaceutical compositions may be administered every 3 to 4 days, every week, or once every two weeks depending on the half-life and clearance rate of the particular formulation. [0148]
  • Normal dosage amounts may vary from 0.1 μg to 100,000 μg, up to a total dose of about 1 gram, depending upon the route of administration. Guidance as to particular dosages and methods of delivery is provided in the literature and generally available to practitioners in the art. Those skilled in the art will employ different formulations for nucleotides than for proteins or their inhibitors. Similarly, delivery of polynucleotides or polypeptides will be specific to particular cells, conditions, locations, etc. [0149]
  • Diagnostics [0150]
  • In another embodiment, antibodies which specifically bind HLHLP may be used for the diagnosis of disorders characterized by expression of HLHLP, or in assays to monitor patients being treated with HLHLP or agonists, antagonists, and inhibitors of HLHLP. Antibodies useful for diagnostic purposes may be prepared in the same manner as those described above for therapeutics. Diagnostic assays for HLHLP include methods which utilize the antibody and a label to detect HLHLP in human body fluids or in extracts of cells or tissues. The antibodies may be used with or without modification, and may be labeled by covalent or non-covalent joining with a reporter molecule. A wide variety of reporter molecules, several of which are described above, are known in the art and may be used. [0151]
  • A variety of protocols for measuring HLHLP, including ELISAs, RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of HLHLP expression. Normal or standard values for HLHLP expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, preferably human, with antibody to HLHLP under conditions suitable for complex formation. The amount of standard complex formation may be quantified by various methods, preferably by photometric means. Quantities of HLHLP expressed in subject, control, and disease samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease. [0152]
  • In another embodiment of the invention, the polynucleotides encoding HLHLP may be used for diagnostic purposes. The polynucleotides which may be used include oligonucleotide sequences, complementary RNA and DNA molecules, and PNAs. The polynucleotides may be used to detect and quantitate gene expression in biopsied tissues in which expression of HLHLP may be correlated with disease. The diagnostic assay may be used to distinguish between absence, presence, and excess expression of HLHLP, and to monitor regulation of HLHLP levels during therapeutic intervention. [0153]
  • In one aspect, hybridization with PCR probes which are capable of detecting polynucleotide sequences, including genomic sequences, encoding HLHLP or closely related molecules may be used to identify nucleic acid sequences which encode HLHLP. The specificity of the probe, whether it is made from a highly specific region (e.g., the 5′ regulatory region) or from a less specific region (e.g., the 3′ coding region), and the stringency of the hybridization or amplification (maximal, high, intermediate, or low), will determine whether the probe identifies only naturally occurring sequences encoding HLHLP, alleles, or related sequences. [0154]
  • Probes may also be used for the detection of related sequences, and should preferably contain at least 50% of the nucleotides from any of the HLHLP encoding sequences. The hybridization probes of the subject invention may be DNA or RNA and may be derived from the sequence of SEQ ID NO:2 or from genomic sequences including promoter and enhancer elements and introns of the naturally occurring HLHLP. [0155]
  • Means for producing specific hybridization probes for DNAs encoding HLHLP include the cloning of polynucleotide sequences encoding HLHLP or HLHLP derivatives into vectors for the production of mRNA probes. Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by means of the addition of the appropriate RNA polymerases and the appropriate labeled nucleotides. Hybridization probes may be labeled by a variety of reporter groups, for example, by radionuclides such as [0156] 32p or 35S, or by enzymatic labels, such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems, and the like.
  • Polynucleotide sequences encoding HLHLP may be used for the diagnosis of a disorder associated with expression of HLHLP. Examples of such a disorder include, but are not limited to, cancers, such as adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; and connective tissue disorders such as Ehlers-Danos syndrome type VI, ankylosing spondylitis, atherosclerosis, Dupuytren's contracture, eosinophilic fasciitis, Felty syndrome, Goodpasture's disease, Hunter syndrome, Hurler syndrome, keloids, Marfan syndrome, nodular fasciitis, osteogenesis imperfecta, polyarthritis nodosa, rheumatoid arthritis, scleroderma, systemic lupus erythematosus, and restenosis following angioplasty. The polynucleotide sequences encoding HLHLP may be used in Southern or northern analysis, dot blot, or other membrane-based technologies; in PCR technologies; in dipstick, pin, and ELISA assays; and in microarrays utilizing fluids or tissues from patient biopsies to detect altered HLHLP expression. Such qualitative or quantitative methods are well known in the art. [0157]
  • In a particular aspect, the nucleotide sequences encoding HLHLP may be useful in assays that detect the presence of associated disorders, particularly those mentioned above. The nucleotide sequences encoding HLHLP may be labeled by standard methods and added to a fluid or tissue sample from a patient under conditions suitable for the formation of hybridization complexes. After a suitable incubation period, the sample is washed and the signal is quantitated and compared with a standard value. If the amount of signal in the patient sample is significantly altered from that of a comparable control sample, the nucleotide sequences have hybridized with nucleotide sequences in the sample, and the presence of altered levels of nucleotide sequences encoding HLHLP in the sample indicates the presence of the associated disorder. Such assays may also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies, in clinical trials, or in monitoring the treatment of an individual patient. [0158]
  • In order to provide a basis for the diagnosis of a disorder associated with expression of HLHLP, a normal or standard profile for expression is established. This may be accomplished by combining body fluids or cell extracts taken from normal subjects, either animal or human, with a sequence, or a fragment thereof, encoding HLHLP, under conditions suitable for hybridization or amplification. Standard hybridization may be quantified by comparing the values obtained from normal subjects with values from an experiment in which a known amount of a substantially purified polynucleotide is used. Standard values obtained from normal samples may be compared with values obtained from samples from patients who are symptomatic for a disorder. Deviation from standard values is used to establish the presence of a disorder. [0159]
  • Once the presence of a disorder is established and a treatment protocol is initiated, hybridization assays may be repeated on a regular basis to evaluate whether the level of expression in the patient begins to approximate that which is observed in the normal subject. The results obtained from successive assays may be used to show the efficacy of treatment over a period ranging from several days to months. [0160]
  • With respect to cancer, the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer. [0161]
  • Additional diagnostic uses for oligonucleotides designed from the sequences encoding HLHLP may involve the use of PCR. These oligomers may be chemically synthesized, generated enzymatically, or produced in vitro. Oligomers will preferably contain a fragment of a polynucleotide encoding HLHLP, or a fragment of a polynucleotide complementary to the polynucleotide encoding HLHLP, and will be employed under optimized conditions for identification of a specific gene or condition. Oligomers may also be employed under less stringent conditions for detection or quantitation of closely related DNA or RNA sequences. [0162]
  • Methods which may also be used to quantitate the expression of HLHLP include radiolabeling or biotinylating nucleotides, coamplification of a control nucleic acid, and interpolating results from standard curves. (Melby, P. C. et al. (1993) J. Immunol. Methods 159:235-244, and Duplaa, C. et al. (1993) Anal. Biochem. 229-236.) The speed of quantitation of multiple samples may be accelerated by running the assay in an ELISA format where the oligomer of interest is presented in various dilutions and a spectrophotometric or colorimetric response gives rapid quantitation. [0163]
  • In further embodiments, oligonucleotides or longer fragments derived from any of the polynucleotide sequences described herein may be used as targets in a microarray. The microarray can be used to monitor the expression level of large numbers of genes simultaneously (to produce a transcript image) and to identify genetic variants, mutations, and polymorphisms. This information may be used in determining gene function, in understanding the genetic basis of a disorder, in diagnosing a disorder, and in developing and monitoring the activities of therapeutic agents. [0164]
  • In one embodiment, the microarray is prepared and used according to methods known in the art, such as those described in published PCT application WO95/11995 (Chee et al.), Lockhart, D. J. et al. (1996; Nat. Biotech. 14:1675-1680), and Schena, M. et al. (1996; Proc. Natl. Acad. Sci. 93:10614-10619). [0165]
  • The microarray is preferably composed of a large number of unique single-stranded nucleic acid sequences, usually either synthetic antisense oligonucleotides or fragments of cDNAs, fixed to a solid support. The oligonucleotides are preferably about 6 to 60 nucleotides in length, more preferably about 15 to 30 nucleotides in length, and most preferably about 20 to 25 nucleotides in length. For a certain type of microarray, it may be preferable to use oligonucleotides which are about 7 to 10 nucleotides in length. The microarray may contain oligonucleotides which cover the known 5′ or 3′ sequence, or may contain sequential oligonucleotides which cover the full length sequence or unique oligonucleotides selected from particular areas along the length of the sequence. Polynucleotides used in the microarray may be oligonucleotides specific to a gene or genes of interest in which at least a fragment of the sequence is known or oligonucleotides specific to one or more unidentified cDNAs common to a particular cell or tissue type or to a normal, developmental, or disease state. In certain situations, it may be appropriate to use pairs of oligonucleotides on a microarray. The pairs will be identical, except for one nucleotide preferably located in the center of the sequence. The second oligonucleotide in the pair (mismatched by one) serves as a control. The number of oligonucleotide pairs may range from about 2 to 1,000,000. [0166]
  • In order to produce oligonucleotides to a known sequence for a microarray, the gene of interest is examined using a computer algorithm which starts at the 5′ end, or, more preferably, at the 3′ end of the nucleotide sequence. The algorithm identifies oligomers of defined length that are unique to the gene, have a GC content within a range suitable for hybridization, and lack predicted secondary structure that may interfere with hybridization. In one aspect, the oligomers are synthesized at designated areas on a substrate using a light-directed chemical process. The substrate may be paper, nylon, any other type of membrane, filter, chip, glass slide, or any other suitable solid support. [0167]
  • In one aspect, the oligonucleotides may be synthesized on the surface of the substrate by using a chemical coupling procedure and an ink jet application apparatus, such as that described in published PCT application WO95/251116 (Baldeschweiler et al.). In another aspect, a grid array analogous to a dot or slot blot (HYBRIDOT apparatus, GIBCO/BRL) may be used to arrange and link cDNA fragments or oligonucleotides to the surface of a substrate using a vacuum system or thermal, UV, mechanical or chemical bonding procedures. In yet another aspect, an array may be produced by hand or by using available devices, materials, and machines (including Brinkmann multichannel pipettors or robotic instruments), and may contain 8, 24, 96, 384, 1536, or 6144 oligonucleotides, or any other multiple from 2 to 1,000,000 which lends itself to the efficient use of commercially available instrumentation. [0168]
  • In order to conduct sample analysis using the microarrays, polynucleotides are extracted from a biological sample. The biological samples may be obtained from any bodily fluid (blood, urine, saliva, phlegm, gastric juices, etc.), cultured cells, biopsies, or other tissue preparations. To produce probes, the polynucleotides extracted from the sample are used to produce nucleic acid sequences which are complementary to the nucleic acids on the microarray. If the microarray consists of cDNAs, antisense RNAs (aRNA) are appropriate probes. Therefore, in one aspect, mRNA is used to produce cDNA which, in turn and in the presence of fluorescent nucleotides, is used to produce fragment or oligonucleotide aRNA probes. These fluorescently labeled probes are incubated with the microarray so that the probe sequences hybridize to the cDNA oligonucleotides of the microarray. In another aspect, nucleic acid sequences used as probes can include polynucleotides, fragments, and complementary or antisense sequences produced using restriction enzymes, PCR technologies, and Oligolabeling or TransProbe kits (Pharmacia & Upjohn) well known in the area of hybridization technology. [0169]
  • Incubation conditions are adjusted so that hybridization occurs with precise complementary matches or with various degrees of less complementarity. After removal of nonhybridized probes, a scanner is used to determine the levels and patterns of fluorescence. The scanned images are examined to determine the degree of complementarity and the relative abundance of each oligonucleotide sequence on the microarray. A detection system may be used to measure the absence, presence, and amount of hybridization for all of the distinct sequences simultaneously. This data may be used for large scale correlation studies or for functional analysis of the sequences, mutations, variants, or polymorphisms among samples. (Heller, R. A. et al. (1997) Proc. Natl. Acad. Sci. 94:2150-2155.) [0170]
  • In another embodiment of the invention, nucleic acid sequences encoding HLHLP may be used to generate hybridization probes useful for mapping the naturally occurring genomic sequence. The sequences may be mapped to a particular chromosome, to a specific region of a chromosome, or to artificial chromosome constructions, such as human artificial chromosomes (HACs), yeast artificial chromosomes (YACs), bacterial artificial chromosomes (BACs), bacterial P1 constructions, or single chromosome cDNA libraries, such as those reviewed in Price, C. M. (1993; Blood Rev. 7:127-134) and Trask, B. J. (1991; Trends Genet. 7:149-154). [0171]
  • Fluorescent in situ hybridization (FISH, as described in Verma et al. (1988) [0172] Human Chromosomes: A Manual of Basic Techniques, Pergamon Press, New York, N.Y.) may be correlated with other physical chromosome mapping techniques and genetic map data. Examples of genetic map data can be found in various scientific journals or at the Online Mendelian Inheritance in Man (OMIM) site. Correlation between the location of the gene encoding HLHLP on a physical chromosomal map and a specific disorder, or predisposition to a specific disorder, may help define the region of DNA associated with that disorder. The nucleotide sequences of the subject invention may be used to detect differences in gene sequences between normal, carrier, and affected individuals.
  • In situ hybridization of chromosomal preparations and physical mapping techniques, such as linkage analysis using established chromosomal markers, may be used for extending genetic maps. Often the placement of a gene on the chromosome of another mammalian species, such as mouse, may reveal associated markers even if the number or arm of a particular human chromosome is not known. New sequences can be assigned to chromosomal arms, or parts thereof, by physical mapping. This provides valuable information to investigators searching for disease genes using positional cloning or other gene discovery techniques. Once the disease or syndrome has been crudely localized by genetic linkage to a particular genomic region, for example, AT to 11q22-23 (Gatti, R. A. et al. (1988) Nature 336:577-580), any sequences mapping to that area may represent associated or regulatory genes for further investigation. The nucleotide sequence of the subject invention may also be used to detect differences in the chromosomal location due to translocation, inversion, etc., among normal, carrier, or affected individuals. [0173]
  • In another embodiment of the invention, HLHLP, its catalytic or immunogenic fragments, or oligopeptides thereof can be used for screening libraries of compounds in any of a variety of drug screening techniques. The fragment employed in such screening may be free in solution, affixed to a solid support, borne on a cell surface, or located intracellularly. The formation of binding complexes between HLHLP and the agent being tested may be measured. [0174]
  • Another technique for drug screening which may be used provides for high throughput screening of compounds having suitable binding affinity to the protein of interest as described in published PCT application WO84/03564. In this method, large numbers of different small test compounds are synthesized on a solid substrate, such as plastic pins or some other surface. The test compounds are reacted with HLHLP, or fragments thereof, and washed. Bound HLHLP is then detected by methods well known in the art. Purified HLHLP can also be coated directly onto plates for use in the aforementioned drug screening techniques. Alternatively, non-neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support. [0175]
  • In another embodiment, one may use competitive drug screening assays in which neutralizing antibodies capable of binding HLHLP specifically compete with a test compound for binding HLHLP. In this manner, antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants with HLHLP. [0176]
  • In additional embodiments, the nucleotide sequences which encode HLHLP may be used in any molecular biology techniques that have yet to be developed, provided the new techniques rely on properties of nucleotide sequences that are currently known, including, but not limited to, such properties as the triplet genetic code and specific base pair interactions. [0177]
  • The examples below are provided to illustrate the subject invention and are not included for the purpose of limiting the invention. [0178]
  • EXAMPLES
  • I. BRAITUT13 cDNA Library Construction [0179]
  • The brain tumor BRAITUT13 cDNA library was constructed from cancerous brain tissue obtained from a 68-year-old Caucasian male during cerebral meningeal excision following diagnosis of meningioma localized in the left frontal part of the brain. [0180]
  • The frozen tissue was homogenized and lysed using a POLYTRON homogenizer (PT-3000; Brinkmann Instruments, Westbury, N.Y.) in guanidinium isothiocyanate solution. The lysate was centrifuged over a 5.7 M CsCl cushion using an SW28 rotor in an L8-70M ultracentrifuge (Beckman Coulter, Fullerton, Calif.) for 18 hours at 25,000 rpm at ambient temperature. The RNA was extracted with acid phenol pH 4.7, precipitated using 0.3 M sodium acetate and 2.5 volumes of ethanol, resuspended in RNAse-free water, and DNase treated at 37° C. RNA extraction and precipitation were repeated as before. The mRNA was then isolated with the OLIGOTEX kit (QIAGEN; Chatsworth, Calif.) and used to construct the cDNA library. [0181]
  • The mRNA was handled according to the recommended protocols in the SUPERSCRIPT Plasmid System for cDNA Synthesis and Plasmid Cloning (Cat. #18248-013; Gibco/BRL). cDNAs were fractionated on a SEPHAROSE CL4B column (Amersham Pharmacia Biotech, Piscataway N.J.), and those cDNAs exceeding 400 bp were ligated into PSPORT1. The plasmid PSPORT1 was subsequently transformed into DH5α competent cells (Cat. #18258-012; Gibco/BRL). [0182]
  • II. Isolation and Sequencing of cDNA Clones [0183]
  • Plasmid DNA was released from the cells and purified using the REAL Prep 96 Plasmid Kit (Catalog #26173; QIAGEN).The recommended protocol was employed except for the following changes: 1) the bacteria were cultured in 1 ml of sterile Terrific Broth (Catalog #22711, Gibco/BRL) with carbenicillin at 25 mg/L and glycerol at 0.4%; 2) after inoculation, the cultures were incubated for 19 hours and at the end of incubation, the cells were lysed with 0.3 ml of lysis buffer; and 3) following isopropanol precipitation, the plasmid DNA pellet was resuspended in 0. 1 ml of distilled water. After the last step in the protocol, samples were transferred to a 96-well block for storage at 4° C. [0184]
  • The cDNAs were sequenced by the method of Sanger et al. (1975, J. Mol. Biol. 94: 441f), using a MICROLAB 2200 (Hamilton, Reno, Nev.) in combination with DNA ENGINE thermal cyclers (PTC200 from MJ Research) and ABI PRISM 377 DNA Sequencing systems (PE Biosystems). [0185]
  • III. Homology Searching of cDNA Clones and Their Deduced Proteins [0186]
  • The nucleotide sequences and/or amino acid sequences of the Sequence Listing were used to query sequences in the GenBank, SwissProt, BLOCKS, and Pima II databases. These databases, which contain previously identified and annotated sequences, were searched for regions of homology using BLAST (Basic Local Alignment Search Tool). (Altschul, S. F. (1993) J. Mol. Evol 36:290-300; and Altschul et al. (1990) J. Mol. Biol. 215:403-410.) [0187]
  • BLAST produced alignments of both nucleotide and amino acid sequences to determine sequence similarity. Because of the local nature of the alignments, BLAST was especially useful in determining exact matches or in identifying homologs which may be of prokaryotic (bacterial) or eukaryotic (animal, fungal, or plant) origin. Other algorithms such as the one described in Smith, T. et al. (1992; Protein Engineering 5:35-51), could have been used when dealing with primary sequence patterns and secondary structure gap penalties. The sequences disclosed in this application have lengths of at least 49 nucleotides and have no more than 12% uncalled bases (where N is recorded rather than A, C, G, or T). [0188]
  • The BLAST approach searched for matches between a query sequence and a database sequence. BLAST evaluated the statistical significance of any matches found, and reported only those matches that satisfy the user-selected threshold of significance. In this application, threshold was set at 10[0189] −25 for nucleotides and 10−10 for peptides.
  • Incyte nucleotide sequences were searched against the GenBank databases for primate (pri), rodent (rod), and other mammalian sequences (mam), and deduced amino acid sequences from the same clones were then searched against GenBank functional protein databases, mammalian (mamp), vertebrate (vrtp), and eukaryote (eukp), for homology. [0190]
  • IV. Northern Analysis [0191]
  • Northern analysis is a laboratory technique used to detect the presence of a transcript of a gene and involves the hybridization of a labeled nucleotide sequence to a membrane on which RNAs from a particular cell type or tissue have been bound. (Sambrook, supra). [0192]
  • Analogous computer techniques applying BLAST are used to search for identical or related molecules in nucleotide databases such as GenBank or the LIFESEQ database (Incyte Genomics, Palo Alto, Calif.). This analysis is much faster than multiple membrane-based hybridizations. In addition, the sensitivity of the computer search can be modified to determine whether any particular match is categorized as exact or homologous. [0193]
  • The basis of the search is the product score, which is defined as: [0194] % sequence identity × % maximum BLAST score 100
    Figure US20030087409A1-20030508-M00001
  • The product score takes into account both the degree of similarity between two sequences and the length of the sequence match. For example, with a product score of 40, the match will be exact within a 1% to 2% error, and, with a product score of 70, the match will be exact. Homologous molecules are usually identified by selecting those which show product scores between 15 and 40, although lower scores may identify related molecules. [0195]
  • The results of northern analysis are reported as a list of libraries in which the transcript encoding HLHLP occurs. Abundance and percent abundance are also reported. Abundance directly reflects the number of times a particular transcript is represented in a cDNA library, and percent abundance is abundance divided by the total number of sequences examined in the cDNA library. [0196]
  • V. Extension of HLHLP Encoding Polynucleotides [0197]
  • The nucleic acid sequence of [0198] Incyte Clone 1622313 was used to design oligonucleotide primers for extending a partial nucleotide sequence to full length. One primer was synthesized to initiate extension of an antisense polynucleotide, and the other was synthesized to extend the sequence of a sense polynucleotide. Primers were used to facilitate the extension of the known sequence “outward” generating amplicons containing new unknown nucleotide sequence for the region of interest. The initial primers were designed from the cDNA using OLIGO 4.06 software (National Biosciences), or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the target sequence at temperatures of about 68° C. to about 72° C. Any stretch of nucleotides which would result in hairpin structures and primer-primer dimerizations was avoided.
  • Selected human cDNA libraries (GIBCO/BRL) were used to extend the sequence. If more than one extension is necessary or desired, additional sets of primers are designed to further extend the known region. [0199]
  • High fidelity amplification was obtained by following the instructions for the XL-PCR kit (PE Biosystems) and thoroughly mixing the enzyme and reaction mix. PCR was performed using the DNA ENGINE thermal cyclers (PTC200; M.J. Research), beginning with 40 pmol of each primer and the recommended concentrations of all other components of the kit, with the following parameters: [0200]
    Step 1 94° C. for 1 min (initial denaturation)
    Step 2 65° C. for 1 min
    Step
    3 68° C. for 6 min
    Step 4 94° C. for 15 sec
    Step
    5 65° C. for 1 min
    Step 6 68° C. for 7 min
    Step 7 Repeat steps 4 through 6 for an additional 15 cycles
    Step 8 94° C. for 15 sec
    Step
    9 65° C. for 1 min
    Step 10 68° C. for 7:15 min
    Step 11 Repeat steps 8 through 10 for an additional 12 cycles
    Step 12 72° C. for 8 min
    Step 13  4° C. (and holding)
  • A 5 μl to 10 μl aliquot of the reaction mixture was analyzed by electrophoresis on a low concentration (about 0.6% to 0.8%) agarose mini-gel to determine which reactions were successful in extending the sequence. Bands thought to contain the largest products were excised from the gel, purified using a QIAQUICK kit (QIAGEN), and trimmed of overhangs using Klenow enzyme to facilitate religation and cloning. [0201]
  • After ethanol precipitation, the products were redissolved in 13 μl of ligation buffer, 1 μl T4-DNA ligase (15 units) and 1 μl T4 polynucleotide kinase were added, and the mixture was incubated at room temperature for 2 to 3 hours, or overnight at 16° C. Competent [0202] E. coli cells (in 40 μl of appropriate media) were transformed with 3 μl of ligation mixture and cultured in 80 μl of SOC medium. (Sambrook, supra). After incubation for one hour at 37° C., the E. coli mixture was plated on Luria Bertani (LB) agar (Sambrook, supra) containing 233 Carb. The following day, several colonies were randomly picked from each plate and cultured in 150 μl of liquid LB/2× Carb medium placed in an individual well of an appropriate commercially-available sterile 96-well microtiter plate. The following day, 5 μl of each overnight culture was transferred into a non-sterile 96-well plate and, after dilution 1:10 with water, 5 μl from each sample was transferred into a PCR array.
  • For PCR amplification, 18 μl of concentrated PCR reaction mix (3.3×) containing 4 units of rTth DNA polymerase, a vector primer, and one or both of the gene specific primers used for the extension reaction were added to each well. Amplification was performed using the following conditions: [0203]
    Step 1 94° C. for 60 sec
    Step 2 94° C. for 20 sec
    Step
    3 55° C. for 30 sec
    Step 4 72° C. for 90 sec
    Step
    5 Repeat steps 2 through 4 for an additional 29 cycles
    Step 6 72° C. for 180 sec
    Step 7  4° C. (and holding)
  • Aliquots of the PCR reactions were run on agarose gels together with molecular weight markers. The sizes of the PCR products were compared to the original partial cDNAs, and appropriate clones were selected, ligated into plasmid, and sequenced. [0204]
  • In like manner, the nucleotide sequence of SEQ ID NO:2 is used to obtain 5′ regulatory sequences using the procedure above, oligonucleotides designed for 5′ extension, and an appropriate genomic library. [0205]
  • VI. Labeling and Use of Individual Hybridization Probes [0206]
  • Hybridization probes derived from SEQ ID NO:2 are employed to screen cDNAs, genomic DNAs, or mRNAs. Although the labeling of oligonucleotides, consisting of about 20 base pairs, is specifically described, essentially the same procedure is used with larger nucleotide fragments. Oligonucleotides are designed using state-of-the-art software such as OLIGO 4.06 software (National Biosciences) and labeled by combining 50 pmol of each oligomer and 250 μCi of [γ-[0207] 32P] adenosine triphosphate (Amersham) and T4 polynucleotide kinase (NEN Life Sciences Products, Boston, Mass.). The labeled oligonucleotides are substantially purified using a SEPHADEX G-25 superfine resin column (Pharmacia & Upjohn). An aliquot containing 107 counts per minute of the labeled probe is used in a typical membrane-based hybridization analysis of human genomic DNA digested with one of the following endonucleases: Ase I, Bgl II, Eco RI, Pst I, Xba 1, or Pvu II (NEN Life Sciences Products).
  • The DNA from each digest is fractionated on a 0.7 percent agarose gel and transferred to nylon membranes (NYTRAN PLUS, Schleicher & Schuell, Durham, N.H.). Hybridization is carried out for 16 hours at 40° C. To remove nonspecific signals, blots are sequentially washed at room temperature under increasingly stringent conditions up to 0.1× saline sodium citrate and 0.5% sodium dodecyl sulfate. After XOMAT AR film (Kodak, Rochester, N.Y.) is exposed to the blots in a Phosphoimager cassette (Molecular Dynamics, Sunnyvale, Calif.) for several hours, hybridization patterns are compared. [0208]
  • VII. Microarrays [0209]
  • To produce oligonucleotides for a microarray, one of the nucleotide sequences of the present invention is examined using a computer algorithm which starts at the 3′ end of the nucleotide sequence. The algorithm identifies oligomers of defined length that are unique to the gene, have a GC content within a range suitable for hybridization, and lack predicted secondary structure that would interfere with hybridization. The algorithm identifies approximately 20 sequence-specific oligonucleotides of 20 nucleotides in length (20-mers). A matched set of oligonucleotides are created in which one nucleotide in the center of each sequence is altered. This process is repeated for each gene in the microarray, and double sets of twenty 20-mers are synthesized and arranged on the surface of the silicon chip using a light-directed chemical process, such as that described in Chee (supra). [0210]
  • In the alternative, a chemical coupling procedure and an ink jet device are used to synthesize oligomers on the surface of a substrate. (See Baldeschweiler, supra.) In another alternative, a grid array analogous to a dot or slot blot is used to arrange and link cDNA fragments or oligonucleotides to the surface of a substrate using a vacuum system or thermal, UV, mechanical, or chemical bonding procedures. A typical array may be produced by hand or using available materials and machines and contain grids of 8 dots, 24 dots, 96 dots, 384 dots, 1536 dots, or 6144 dots. After hybridization, the microarray is washed to remove nonhybridized probes, and a scanner is used to determine the levels and patterns of fluorescence. The scanned image is examined to determine the degree of complementarity and the relative abundance/expression level of each oligonucleotide sequence in the microarray. [0211]
  • VIII. Complementary Polynucleotides [0212]
  • Sequences complementary to the HLHLP-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occurring HLHLP. Although use of oligonucleotides comprising from about 15 to 30 base pairs is described, essentially the same procedure is used with smaller or with larger sequence fragments. Appropriate oligonucleotides are designed using OLIGO 4.06 software (National Biosciences) and the coding sequence of HLHLP. To inhibit transcription, a complementary oligonucleotide is designed from the most unique 5′ sequence and used to prevent promoter binding to the coding sequence. To inhibit translation, a complementary oligonucleotide is designed to prevent ribosomal binding to the HLHLP-encoding transcript. [0213]
  • IX. Expression of HLHLP [0214]
  • Expression of HLHLP is accomplished by subcloning the cDNAs into appropriate vectors and transforming the vectors into host cells. In this case, the cloning vector is also used to express HLHLP in [0215] E. coli. This vector contains a promoter for β-galactosidase upstream of the cloning site, followed by sequence containing the amino-terminal Met and the subsequent seven residues of β-galactosidase. Immediately following these eight residues is a bacteriophage promoter useful for transcription and a linker containing a number of unique restriction sites.
  • Induction of an isolated, transformed bacterial strain with isopropyl beta-D-thiogalactopyranoside (IPTG) using standard methods produces a fusion protein which consists of the first 8 residues of β-galactosidase, about 5 to 15 residues of linker, and the full length protein. The signal residues direct the secretion of HLHLP into bacterial growth media which can be used directly in the following assay for activity. [0216]
  • X. Demonstration of HLHLP Activity [0217]
  • The activity of HLHLP is determined by measuring the production of hydroxy[[0218] 14C]lysine from [14C]lysine. Radiolabeled protocollagen is incubated with HLHLP in buffer containing ascorbic acid, iron sulfate, dithiothreitol, bovine serum albumin, and catalase. Following a 30 minute incubation, the reaction is stopped by the addition of acetone, and centrifuged. The sedimented material is dried, and the hydroxy[14C]lysine is converted to [14C]formaldehyde by oxidation with periodate, and then extracted into toluene. The amount of 14C extracted into toluene is quantified by scintillation counting, and is proportional to the activity of HLHLP in the sample (Kivirikko, K., and Myllyla, R. (1982) Methods Enzymol 82: 245-304).
  • XI. Production of HLHLP Specific Antibodies [0219]
  • HLHLP substantially purified using PAGE electrophoresis (Sambrook, supra), or other purification techniques, is used to immunize rabbits and to produce antibodies using standard protocols. The HLHLP amino acid sequence is analyzed using LASERGENE software (DNASTAR Inc) to determine regions of high immunogenicity, and a corresponding oligopeptide is synthesized and used to raise antibodies by means known to those of skill in the art. Selection of appropriate epitopes, such as those near the C-terminus or in hydrophilic regions, is described by Ausubel (supra) and by others. [0220]
  • Typically, the oligopeptides are 15 residues in length, and are synthesized using an ABI 431A peptide synthesizer using Fmoc-chemistry and coupled to KLH (Sigma, St. Louis, Mo.) by reaction with N-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS), following the procedure described in Ausubel et al., supra. Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant. Resulting antisera are tested for antipeptide activity, for example, by binding the peptide to plastic, blocking with 1% BSA, reacting with rabbit antisera, washing, and reacting with radio-iodinated goat anti-rabbit IgG. [0221]
  • XII. Purification of Naturally Occurring HLHLP Using Specific Antibodies [0222]
  • Naturally occurring or recombinant HLHLP is substantially purified by immunoaffinity chromatography using antibodies specific for HLHLP. An immunoaffinity column is constructed by covalently coupling HLHLP antibody to an activated chromatographic resin, such as CNBr-activated SEPHAROSE (Pharmacia & Upjohn). After the coupling, the resin is blocked and washed according to the manufacturer's instructions. [0223]
  • Media containing HLHLP are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of HLHLP (e.g., high ionic strength buffers in the presence of detergent). The column is eluted under conditions that disrupt antibody/HLHLP binding (e.g., a buffer of pH 2 to [0224] pH 3, or a high concentration of a chaotrope, such as urea or thiocyanate ion), and HLHLP is collected.
  • XIII. Identification of Molecules Which Interact with HLHLP [0225]
  • HLHLP or biologically active fragments thereof are labeled with [0226] 125I Bolton-Hunter reagent. (Bolton et al. (1973) Biochem. J. 133:529-539). Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled HLHLP, washed, and any wells with labeled HLHLP complex are assayed. Data obtained using different concentrations of HLHLP are used to calculate values for the number, affinity, and association of HLHLP with the candidate molecules.
  • Various modifications and variations of the described methods and systems of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in molecular biology or related fields are intended to be within the scope of the following claims. [0227]
  • 1 4 1 738 PRT Homo sapiens misc_feature Incyte ID No 1622313 1 Met Thr Ser Ser Gly Pro Gly Pro Arg Phe Leu Leu Leu Leu Pro 1 5 10 15 Leu Leu Leu Pro Pro Ala Ala Ser Ala Ser Asp Arg Pro Arg Gly 20 25 30 Arg Asp Pro Val Asn Pro Glu Lys Leu Leu Val Ile Thr Val Ala 35 40 45 Thr Ala Glu Thr Glu Gly Tyr Leu Arg Phe Leu Arg Ser Ala Glu 50 55 60 Phe Phe Asn Tyr Thr Val Arg Thr Leu Gly Leu Gly Glu Glu Trp 65 70 75 Arg Gly Gly Asp Val Ala Arg Thr Val Gly Gly Gly Gln Lys Val 80 85 90 Arg Trp Leu Lys Lys Glu Met Glu Lys Tyr Ala Asp Arg Glu Asp 95 100 105 Met Ile Ile Met Phe Val Asp Ser Tyr Asp Val Ile Leu Ala Gly 110 115 120 Ser Pro Thr Glu Leu Leu Lys Lys Phe Val Gln Ser Gly Ser Arg 125 130 135 Leu Leu Phe Ser Ala Glu Ser Phe Cys Trp Pro Glu Trp Gly Leu 140 145 150 Ala Glu Gln Tyr Pro Glu Val Gly Thr Gly Lys Arg Phe Leu Asn 155 160 165 Ser Gly Gly Phe Ile Gly Phe Ala Thr Thr Ile His Gln Ile Val 170 175 180 Arg Gln Trp Lys Tyr Lys Asp Asp Asp Asp Asp Gln Leu Phe Tyr 185 190 195 Thr Arg Leu Tyr Leu Asp Pro Gly Leu Arg Glu Lys Leu Ser Leu 200 205 210 Asn Leu Asp His Lys Ser Arg Ile Phe Gln Asn Leu Asn Gly Ala 215 220 225 Leu Asp Glu Val Val Leu Lys Phe Asp Arg Asn Arg Val Arg Ile 230 235 240 Arg Asn Val Ala Tyr Asp Thr Leu Pro Ile Val Val His Gly Asn 245 250 255 Gly Pro Thr Lys Leu Gln Leu Asn Tyr Leu Gly Asn Tyr Val Pro 260 265 270 Asn Gly Trp Thr Pro Glu Gly Gly Cys Gly Phe Cys Asn Gln Asp 275 280 285 Arg Arg Thr Leu Pro Gly Gly Gln Pro Pro Pro Arg Val Phe Leu 290 295 300 Ala Val Phe Val Glu Gln Pro Thr Pro Phe Leu Pro Arg Phe Leu 305 310 315 Gln Arg Leu Leu Leu Leu Asp Tyr Pro Pro Asp Arg Val Thr Leu 320 325 330 Phe Leu His Asn Asn Glu Val Phe His Glu Pro His Ile Ala Asp 335 340 345 Ser Trp Pro Gln Leu Gln Asp His Phe Ser Ala Val Lys Leu Val 350 355 360 Gly Pro Glu Glu Ala Leu Ser Pro Gly Glu Ala Arg Asp Met Ala 365 370 375 Met Asp Leu Cys Arg Gln Asp Pro Glu Cys Glu Phe Tyr Phe Ser 380 385 390 Leu Asp Ala Asp Ala Val Leu Thr Asn Leu Gln Thr Leu Arg Ile 395 400 405 Leu Ile Glu Glu Asn Arg Lys Val Ile Ala Pro Met Leu Ser Arg 410 415 420 His Gly Lys Leu Trp Ser Asn Phe Trp Gly Ala Leu Ser Pro Asp 425 430 435 Glu Tyr Tyr Ala Arg Ser Glu Asp Tyr Val Glu Leu Val Gln Arg 440 445 450 Lys Arg Val Gly Val Trp Asn Val Pro Tyr Ile Ser Gln Ala Tyr 455 460 465 Val Ile Arg Gly Asp Thr Leu Arg Met Glu Leu Pro Gln Arg Asp 470 475 480 Val Phe Ser Gly Ser Asp Thr Asp Pro Asp Met Ala Phe Cys Lys 485 490 495 Ser Phe Arg Asp Lys Gly Ile Phe Leu His Leu Ser Asn Gln His 500 505 510 Glu Phe Gly Arg Leu Leu Ala Thr Ser Arg Tyr Asp Thr Glu His 515 520 525 Leu His Pro Asp Leu Trp Gln Ile Phe Asp Asn Pro Val Asp Trp 530 535 540 Lys Glu Gln Tyr Ile His Glu Asn Tyr Ser Arg Ala Leu Glu Gly 545 550 555 Glu Gly Ile Val Glu Gln Pro Cys Pro Asp Val Tyr Trp Phe Pro 560 565 570 Leu Leu Ser Glu Gln Met Cys Asp Glu Leu Val Ala Glu Met Glu 575 580 585 His Tyr Gly Gln Trp Ser Gly Gly Arg His Glu Asp Ser Arg Leu 590 595 600 Ala Gly Gly Tyr Glu Asn Val Pro Thr Val Asp Ile His Met Lys 605 610 615 Gln Val Gly Tyr Glu Asp Gln Trp Leu Gln Leu Leu Arg Thr Tyr 620 625 630 Val Gly Pro Met Thr Glu Ser Leu Phe Pro Gly Tyr His Thr Lys 635 640 645 Ala Arg Ala Val Met Asn Phe Val Val Arg Tyr Arg Pro Asp Glu 650 655 660 Gln Pro Ser Leu Arg Pro His His Asp Ser Ser Thr Phe Thr Leu 665 670 675 Asn Val Ala Leu Asn His Lys Gly Leu Asp Tyr Glu Gly Gly Gly 680 685 690 Cys Arg Phe Leu Arg Tyr Asp Cys Val Ile Ser Ser Pro Arg Lys 695 700 705 Gly Trp Ala Leu Leu His Pro Gly Arg Leu Thr His Tyr His Glu 710 715 720 Gly Leu Pro Thr Thr Trp Gly Thr Arg Tyr Ile Met Val Ser Phe 725 730 735 Val Asp Pro 2 2594 DNA Homo sapiens misc_feature Incyte ID No 1622313 2 ctgggatcca ggcgcgtagg cagatcccag gatcctgggt gctgtctggg cccgctcccc 60 accatgacct cctcggggcc tggaccccgg ttcctgctgc tgctgccgct gctgctgccc 120 cctgcggcct cagcctccga ccggccccgg ggccgagacc cggtcaaccc agagaagctg 180 ctggtgatca ctgtggccac agctgaaacc gaggggtacc tgcgtttcct gcgctctgcg 240 gagttcttca actacactgt gcggaccctg ggcctgggag aggagtggcg agggggtgat 300 gtggctcgaa cagttggtgg aggacagaag gtccggtggt taaagaagga aatggagaaa 360 tacgctgacc gggaggatat gatcatcatg tttgtggata gctacgacgt gattctggcc 420 ggcagcccca cagagctgct gaagaagttc gtccagagtg gcagccgcct gctcttctct 480 gcagagagct tctgctggcc cgagtggggg ctggcggagc agtaccctga ggtgggcacg 540 gggaagcgct tcctcaattc tggtggattc atcggttttg ccaccaccat ccaccaaatc 600 gtgcgccagt ggaagtacaa ggatgatgac gacgaccagc tgttctacac acggctctac 660 ctggacccag gactgaggga gaaactcagc cttaatctgg atcataagtc tcggatcttt 720 cagaacctca acggggcttt agatgaagtg gttttaaagt ttgatcggaa ccgtgtgcgt 780 atccggaacg tggcctacga cacgctcccc attgtggtcc atggaaacgg tcccactaag 840 ctgcagctca actacctggg aaactacgtc cccaatggct ggactcctga gggaggctgt 900 ggcttctgca accaggaccg gaggacactc ccgggggggc agcctccccc ccgggtgttt 960 ctggccgtgt ttgtggaaca gcctactccg tttctgcccc gattcctgca gcggctgcta 1020 ctcctggact atccccccga cagggtcacc cttttcctgc acaacaacga ggtcttccat 1080 gaaccccaca tcgctgactc ctggccgcag ctccaggacc acttctcagc tgtgaagctc 1140 gtggggccgg aggaggctct gagcccaggc gaggccaggg acatggccat ggacctgtgt 1200 cggcaggacc ccgagtgtga gttctacttc agcctggacg ccgacgctgt cctcaccaac 1260 ctgcagaccc tgcgtatcct cattgaggag aacaggaagg tgatcgcccc catgctgtcc 1320 cgccacggca agctgtggtc caacttctgg ggcgccctga gccccgatga gtactacgcc 1380 cgctccgagg actacgtgga gctggtgcag cggaagcgag tgggtgtgtg gaatgtacca 1440 tacatctccc aggcctatgt gatccggggt gataccctgc ggatggagct gccccagagg 1500 gatgtgttct cgggcagtga cacagacccg gacatggcct tctgtaagag ctttcgagac 1560 aagggcatct tcctccatct gagcaatcag catgaatttg gccggctcct ggccacttcc 1620 agatacgaca cggagcacct gcaccccgac ctctggcaga tcttcgacaa ccccgtcgac 1680 tggaaggagc agtacatcca cgagaactac agccgggccc tggaagggga aggaatcgtg 1740 gagcagccat gcccggacgt gtactggttc ccactgctgt cagaacaaat gtgtgatgag 1800 ctggtggcag agatggagca ctacggccag tggtcaggcg gccggcatga ggattcaagg 1860 ctggctggag gctacgagaa tgtgcccacc gtggacatcc acatgaagca ggtggggtac 1920 gaggaccagt ggctgcagct gctgcggacg tatgtgggcc ccatgaccga gagcctgttt 1980 cccggttacc acaccaaggc gcgggcggtg atgaactttg tggttcgcta ccggccagac 2040 gagcagccgt ctctgcggcc acaccacgac tcatccacct tcaccctcaa cgttgccctc 2100 aaccacaagg gcctggacta tgagggaggt ggctgccgct tcctgcgcta cgactgtgtg 2160 atctcctccc cgaggaaggg ctgggcactc ctgcaccccg gccgcctcac ccactaccac 2220 gaggggctgc caacgacctg gggcacacgc tacatcatgg tgtcctttgt cgacccctga 2280 cactcaacca ctctgccaaa cctgccctgc cattgtgcct ttttaggggg cctggccccc 2340 gtcctgggag ttgggggatg ggtctctctg tctccccact tcctgagttc atgttccgcg 2400 tgcctgaact gaatatgtca ccttgctccc aagacacggc cctctcagga agctcccgga 2460 gtccccgcct ctctcctccg cccacagggg ttcgtgggca cagggcttct ggggactccc 2520 cgcgtgataa attattaatg ttccgcagtc tcactctgaa taaaggacag tttgtaagtc 2580 ttgaaaaaaa aaaa 2594 3 737 PRT Homo sapiens misc_feature GenBank ID No g2138314 3 Met Gly Gly Cys Thr Val Lys Pro Gln Leu Leu Leu Leu Ala Leu 1 5 10 15 Val Leu His Pro Trp Asn Pro Cys Leu Gly Ala Asp Ser Glu Lys 20 25 30 Pro Ser Ser Ile Pro Thr Asp Lys Leu Leu Val Ile Thr Val Ala 35 40 45 Thr Lys Glu Ser Asp Gly Phe His Arg Phe Met Gln Ser Ala Lys 50 55 60 Tyr Phe Asn Tyr Thr Val Lys Val Leu Gly Gln Gly Glu Glu Trp 65 70 75 Arg Gly Gly Asp Gly Ile Asn Ser Ile Gly Gly Gly Gln Lys Val 80 85 90 Arg Leu Met Lys Glu Val Met Glu His Tyr Ala Asp Gln Asp Asp 95 100 105 Leu Val Val Met Phe Thr Glu Cys Phe Asp Val Ile Phe Ala Gly 110 115 120 Gly Pro Glu Glu Val Leu Lys Lys Phe Gln Lys Ala Asn His Lys 125 130 135 Val Val Phe Ala Ala Asp Gly Ile Leu Trp Pro Asp Lys Arg Leu 140 145 150 Ala Asp Lys Tyr Pro Val Val His Ile Gly Lys Arg Tyr Leu Asn 155 160 165 Ser Gly Gly Phe Ile Gly Tyr Ala Pro Tyr Val Asn Arg Ile Val 170 175 180 Gln Gln Trp Asn Leu Gln Asp Asn Asp Asp Asp Gln Leu Phe Tyr 185 190 195 Thr Lys Val Tyr Ile Asp Pro Leu Lys Arg Glu Ala Ile Asn Ile 200 205 210 Thr Leu Asp His Lys Cys Lys Ile Phe Gln Thr Leu Asn Gly Ala 215 220 225 Val Asp Glu Val Val Leu Lys Phe Glu Asn Gly Lys Ala Arg Ala 230 235 240 Lys Asn Thr Phe Tyr Glu Thr Leu Pro Val Ala Ile Asn Gly Asn 245 250 255 Gly Pro Thr Lys Ile Leu Leu Asn Tyr Phe Gly Asn Tyr Val Pro 260 265 270 Asn Ser Trp Thr Gln Asp Asn Gly Cys Thr Leu Cys Glu Phe Asp 275 280 285 Thr Val Asp Leu Ser Ala Val Asp Val His Pro Asn Val Ser Ile 290 295 300 Gly Val Phe Ile Glu Gln Pro Thr Pro Phe Leu Pro Arg Phe Leu 305 310 315 Asp Ile Leu Leu Thr Leu Asp Tyr Pro Lys Glu Ala Leu Lys Leu 320 325 330 Phe Ile His Asn Lys Glu Val Tyr His Glu Lys Asp Ile Lys Val 335 340 345 Phe Phe Asp Lys Ala Lys His Glu Ile Lys Thr Ile Lys Ile Val 350 355 360 Gly Pro Glu Glu Asn Leu Ser Gln Ala Glu Ala Arg Asn Met Gly 365 370 375 Met Asp Phe Cys Arg Gln Asp Glu Lys Cys Asp Tyr Tyr Phe Ser 380 385 390 Val Asp Ala Asp Val Val Leu Thr Asn Pro Arg Thr Leu Lys Ile 395 400 405 Leu Ile Glu Gln Asn Arg Lys Ile Ile Ala Pro Leu Val Thr Arg 410 415 420 His Gly Lys Leu Trp Ser Asn Phe Trp Gly Ala Leu Ser Pro Asp 425 430 435 Gly Tyr Tyr Ala Arg Ser Glu Asp Tyr Val Asp Ile Val Gln Gly 440 445 450 Asn Arg Val Gly Val Trp Asn Val Pro Tyr Met Ala Asn Val Tyr 455 460 465 Leu Ile Lys Gly Lys Thr Leu Arg Ser Glu Met Asn Glu Arg Asn 470 475 480 Tyr Phe Val Arg Asp Lys Leu Asp Pro Asp Met Ala Leu Cys Arg 485 490 495 Asn Ala Arg Glu Met Gly Val Phe Met Tyr Ile Ser Asn Arg His 500 505 510 Glu Phe Gly Arg Leu Leu Ser Thr Ala Asn Tyr Asn Thr Ser His 515 520 525 Tyr Asn Asn Asp Leu Trp Gln Ile Phe Glu Asn Pro Val Asp Trp 530 535 540 Lys Glu Lys Tyr Ile Asn Arg Asp Tyr Ser Lys Ile Phe Thr Glu 545 550 555 Asn Ile Val Glu Gln Pro Cys Pro Asp Val Phe Trp Phe Pro Ile 560 565 570 Phe Ser Glu Lys Ala Cys Asp Glu Leu Val Glu Glu Met Glu His 575 580 585 Tyr Gly Lys Trp Ser Gly Gly Lys His His Asp Ser Arg Ile Ser 590 595 600 Gly Gly Tyr Glu Asn Val Pro Thr Asp Asp Ile His Met Lys Gln 605 610 615 Val Asp Leu Glu Asn Val Trp Leu Asp Phe Ile Arg Glu Phe Ile 620 625 630 Ala Pro Val Thr Leu Lys Val Phe Ala Gly Tyr Tyr Thr Lys Gly 635 640 645 Phe Ala Leu Leu Asn Phe Val Val Lys Tyr Ser Pro Glu Arg Gln 650 655 660 Arg Ser Leu Arg Pro His His Asp Ala Ser Thr Phe Thr Ile Asn 665 670 675 Ile Ala Leu Asn Asn Val Gly Glu Asp Phe Gln Gly Gly Gly Cys 680 685 690 Lys Phe Leu Arg Tyr Asn Cys Ser Ile Glu Ser Pro Arg Lys Gly 695 700 705 Trp Ser Phe Met His Pro Gly Arg Leu Thr His Leu His Glu Gly 710 715 720 Leu Pro Val Lys Asn Gly Thr Arg Tyr Ile Ala Val Ser Phe Ile 725 730 735 Asp Pro 4 727 PRT Homo sapiens misc_feature GenBank ID No g190074 4 Met Arg Pro Leu Leu Leu Leu Ala Leu Leu Gly Trp Leu Leu Leu 1 5 10 15 Ala Glu Ala Lys Gly Asp Ala Lys Pro Glu Asp Asn Leu Leu Val 20 25 30 Leu Thr Val Ala Thr Lys Glu Thr Glu Gly Phe Arg Arg Phe Lys 35 40 45 Arg Ser Ala Gln Phe Phe Asn Tyr Lys Ile Gln Ala Leu Gly Leu 50 55 60 Gly Glu Asp Trp Asn Val Glu Lys Gly Thr Ser Ala Gly Gly Gly 65 70 75 Gln Lys Val Arg Leu Leu Lys Lys Ala Leu Glu Lys His Ala Asp 80 85 90 Lys Glu Asp Leu Val Ile Leu Phe Thr Asp Ser Tyr Asp Val Leu 95 100 105 Phe Ala Ser Gly Pro Arg Glu Leu Leu Lys Lys Phe Arg Gln Ala 110 115 120 Arg Ser Gln Val Val Phe Ser Ala Glu Glu Leu Ile Tyr Pro Asp 125 130 135 Arg Arg Leu Glu Thr Lys Tyr Pro Val Val Ser Asp Gly Lys Arg 140 145 150 Phe Leu Gly Ser Gly Gly Phe Ile Gly Tyr Ala Pro Asn Leu Ser 155 160 165 Lys Leu Val Ala Glu Trp Glu Gly Gln Asp Ser Asp Ser Asp Gln 170 175 180 Leu Phe Tyr Thr Lys Ile Phe Leu Asp Pro Glu Lys Arg Glu Gln 185 190 195 Ile Asn Ile Thr Leu Asp His Arg Cys Arg Ile Phe Gln Asn Leu 200 205 210 Asp Gly Ala Leu Asp Glu Val Val Leu Lys Phe Glu Met Gly His 215 220 225 Val Arg Ala Arg Asn Leu Ala Tyr Asp Thr Leu Pro Val Leu Ile 230 235 240 His Gly Asn Gly Pro Thr Lys Leu Gln Leu Asn Tyr Leu Gly Asn 245 250 255 Tyr Ile Pro Arg Phe Trp Thr Phe Glu Thr Gly Cys Thr Val Cys 260 265 270 Asp Glu Gly Leu Arg Ser Leu Lys Gly Ile Gly Asp Glu Ala Leu 275 280 285 Pro Thr Val Leu Val Gly Val Phe Ile Glu Gln Pro Thr Pro Phe 290 295 300 Val Ser Leu Phe Phe Gln Arg Leu Leu Arg Leu His Tyr Pro Gln 305 310 315 Lys His Met Arg Leu Phe Ile His Asn His Glu Gln His His Lys 320 325 330 Ala Gln Val Glu Glu Phe Leu Ala Gln His Gly Ser Glu Tyr Gln 335 340 345 Ser Val Lys Leu Val Gly Pro Glu Val Arg Met Ala Asn Ala Asp 350 355 360 Ala Arg Asn Met Gly Ala Asp Leu Cys Arg Gln Asp Arg Ser Cys 365 370 375 Thr Tyr Tyr Phe Ser Val Asp Ala Asp Val Ala Leu Thr Glu Pro 380 385 390 Asn Ser Leu Arg Leu Leu Ile Gln Gln Asn Lys Asn Val Ile Ala 395 400 405 Pro Leu Met Thr Arg His Gly Arg Leu Trp Ser Asn Phe Trp Gly 410 415 420 Ala Leu Ser Ala Asp Gly Tyr Tyr Ala Arg Ser Glu Asp Tyr Val 425 430 435 Asp Ile Val Gln Gly Arg Arg Val Gly Val Trp Asn Val Pro Tyr 440 445 450 Ile Ser Asn Ile Tyr Leu Ile Lys Gly Ser Ala Leu Arg Gly Glu 455 460 465 Leu Gln Ser Ser Asp Leu Phe His His Ser Lys Leu Asp Pro Asp 470 475 480 Met Ala Phe Cys Ala Asn Ile Arg Gln Gln Asp Val Phe Met Phe 485 490 495 Leu Thr Asn Arg His Thr Leu Gly His Leu Leu Ser Leu Asp Ser 500 505 510 Tyr Arg Thr Thr His Leu His Asn Asp Leu Trp Glu Val Phe Ser 515 520 525 Asn Pro Glu Asp Trp Lys Glu Lys Tyr Ile His Gln Asn Tyr Thr 530 535 540 Lys Ala Leu Ala Gly Lys Leu Val Glu Thr Pro Cys Pro Asp Val 545 550 555 Tyr Trp Phe Pro Ile Phe Thr Glu Val Ala Cys Asp Glu Leu Val 560 565 570 Glu Glu Met Glu His Phe Gly Gln Trp Ser Leu Gly Asn Asn Lys 575 580 585 Asp Asn Arg Ile Gln Gly Gly Tyr Glu Asn Val Pro Thr Ile Asp 590 595 600 Ile His Met Asn Gln Ile Gly Phe Glu Arg Glu Trp His Lys Phe 605 610 615 Leu Leu Glu Tyr Ile Ala Pro Met Thr Glu Lys Leu Tyr Pro Gly 620 625 630 Tyr Tyr Thr Arg Ala Gln Phe Asp Leu Ala Phe Val Val Arg Tyr 635 640 645 Lys Pro Asp Glu Gln Pro Ser Leu Met Pro His His Asp Ala Ser 650 655 660 Thr Phe Thr Ile Asn Ile Ala Leu Asn Arg Val Gly Val Asp Tyr 665 670 675 Glu Gly Gly Gly Cys Arg Phe Leu Arg Tyr Asn Cys Ser Ile Arg 680 685 690 Ala Pro Arg Lys Gly Trp Thr Leu Met His Pro Gly Arg Leu Thr 695 700 705 His Tyr His Glu Gly Leu Pro Thr Thr Arg Gly Thr Arg Tyr Ile 710 715 720 Ala Val Ser Phe Val Asp Pro 725

Claims (55)

What is claimed is:
1. An isolated polypeptide selected from the group consisting of:
a) a polypeptide comprising an amino acid sequence of SEQ ID NO:1,
b) a polypeptide comprising a naturally occurring an amino acid sequence at least 90% identical to an amino acid sequence of SEQ ID NO:1,
c) a biologically active fragment of a polypeptide having an amino acid sequence of SEQ ID NO:1, and
d) an immunogenic fragment of a polypeptide having an amino acid sequence of SEQ ID NO:1.
2. An isolated polypeptide of claim 1, comprising an amino acid sequence of SEQ ID NO:1.
3. An isolated polynucleotide encoding a polypeptide of claim 1.
4. An isolated polynucleotide encoding a polypeptide of claim 2.
5. An isolated polynucleotide of claim 4, having a sequence of SEQ ID NO:2.
6. A recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide of claim 3.
7. A cell transformed with a recombinant polynucleotide of claim 6.
8. A transgenic organism comprising a recombinant polynucleotide of claim 6.
9. A method of producing a polypeptide of claim 1, the method comprising:
a) culturing a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide, and said recombinant polynucleotide comprises a promoter sequence operably linked to a polynucleotide encoding the polypeptide of claim 1, and
b) recovering the polypeptide so expressed.
10. A method of claim 9, wherein the polypeptide comprises the amino acid sequence of SEQ ID NO:1.
11. An isolated antibody which specifically binds to a polypeptide of claim 1.
12. An isolated polynucleotide selected from the group consisting of:
a) a polynucleotide comprising a polynucleotide sequence of SEQ ID NO:2,
b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence of SEQ ID NO:2,
c) a polynucleotide complementary to a polynucleotide of a),
d) a polynucleotide complementary to a polynucleotide of b) and
e) an RNA equivalent of a)-d).
13. An isolated polynucleotide comprising at least 60 contiguous nucleotides of a polynucleotide of claim 12.
14. A method of detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide of claim 12, the method comprising:
a) hybridizing the sample with a probe comprising at least 20 contiguous nucleotides comprising a sequence complementary to said target polynucleotide in the sample, and which probe specifically hybridizes to said target polynucleotide, under conditions whereby a hybridization complex is formed between said probe and said target polynucleotide or fragments thereof, and
b) detecting the presence or absence of said hybridization complex, and, optionally, if present, the amount thereof.
15. A method of claim 14, wherein the probe comprises at least 60 contiguous nucleotides.
16. A method of detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide of claim 12, the method comprising:
a) amplifying said target polynucleotide or fragment thereof using polymerase chain reaction amplification, and
b) detecting the presence or absence of said amplified target polynucleotide or fragment thereof, and, optionally, if present, the amount thereof.
17. A composition comprising a polypeptide of claim 1 and a pharmaceutically acceptable excipient.
18. A composition of claim 17, wherein the polypeptide comprises an amino acid sequence of SEQ ID NO:1.
19. A method for treating a disease or condition associated with decreased expression of functional HLHLP, comprising administering to a patient in need of such treatment the composition of claim 17.
20. A method of screening a compound for effectiveness as an agonist of a polypeptide of claim 1, the method comprising:
a) exposing a sample comprising a polypeptide of claim 1 to a compound, and
b) detecting agonist activity in the sample.
21. A composition comprising an agonist compound identified by a method of claim 20 and a pharmaceutically acceptable excipient.
22. A method for treating a disease or condition associated with decreased expression of functional HLHLP, comprising administering to a patient in need of such treatment a composition of claim 21.
23. A method of screening a compound for effectiveness as an antagonist of a polypeptide of claim 1, the method comprising:
a) exposing a sample comprising a polypeptide of claim 1 to a compound, and
b) detecting antagonist activity in the sample.
24. A composition comprising an antagonist compound identified by a method of claim 23 and a pharmaceutically acceptable excipient.
25. A method for treating a disease or condition associated with overexpression of functional HLHLP, comprising administering to a patient in need of such treatment a composition of claim 24.
26. A method of screening for a compound that specifically binds to the polypeptide of claim 1, the method comprising:
a) combining the polypeptide of claim 1 with at least one test compound under suitable conditions, and
b) detecting binding of the polypeptide of claim 1 to the test compound, thereby identifying a compound that specifically binds to the polypeptide of claim 1.
27. A method of screening for a compound that modulates the activity of the polypeptide of claim 1, said method comprising:
a) combining the polypeptide of claim 1 with at least one test compound under conditions permissive for the activity of the polypeptide of claim 1,
b) assessing the activity of the polypeptide of claim 1 in the presence of the test compound, and
c) comparing the activity of the polypeptide of claim 1 in the presence of the test compound with the activity of the polypeptide of claim 1 in the absence of the test compound, wherein a change in the activity of the polypeptide of claim 1 in the presence of the test compound is indicative of a compound that modulates the activity of the polypeptide of claim 1.
28. A method of screening a compound for effectiveness in altering expression of a target polynucleotide, wherein said target polynucleotide comprises a polynucleotide sequence of claim 5, the method comprising:
a) exposing a sample comprising the target polynucleotide to a compound, under conditions suitable for the expression of the target polynucleotide,
b) detecting altered expression of the target polynucleotide, and
c) comparing the expression of the target polynucleotide in the presence of varying amounts of the compound and in the absence of the compound.
29. A method of assessing toxicity of a test compound, the method comprising:
a) treating a biological sample containing nucleic acids with the test compound,
b) hybridizing the nucleic acids of the treated biological sample with a probe comprising at least 20 contiguous nucleotides of a polynucleotide of claim 12 under conditions whereby a specific hybridization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide comprising a polynucleotide sequence of a polynucleotide of claim 12 or fragment thereof,
c) quantifying the amount of hybridization complex, and
d) comparing the amount of hybridization complex in the treated biological sample with the amount of hybridization complex in an untreated biological sample, wherein a difference in the amount of hybridization complex in the treated biological sample is indicative of toxicity of the test compound.
30. A diagnostic test for a condition or disease associated with the expression of HLHLP in a biological sample, the method comprising:
a) combining the biological sample with an antibody of claim 11, under conditions suitable for the antibody to bind the polypeptide and form an antibody:polypeptide complex, and
b) detecting the complex, wherein the presence of the complex correlates with the presence of the polypeptide in the biological sample.
31. The antibody of claim 11, wherein the antibody is:
a) a chimeric antibody,
b) a single chain antibody,
c) a Fab fragment,·
d) a F(ab′)2 fragment, or
e) a humanized antibody.
32. A composition comprising an antibody of claim 11 and an acceptable excipient.
33. A method of diagnosing a condition or disease associated with the expression of HLHLP in a subject, comprising administering to said subject an effective amount of the composition of claim 32.
34. A composition of claim 32, wherein the antibody is labeled.
35. A method of diagnosing a condition or disease associated with the expression of HLHLP in a subject, comprising administering to said subject an effective amount of the composition of claim 34.
36. A method of preparing a polyclonal antibody with the specificity of the antibody of claim 11, the method comprising:
a) immunizing an animal with a polypeptide consisting of an amino acid sequence of SEQ ID NO:1, or an immunogenic fragment thereof, under conditions to elicit an antibody response,
b) isolating antibodies from said animal, and
c) screening the isolated antibodies with the polypeptide, thereby identifying a polyclonal antibody which binds specifically to a polypeptide comprising an amino acid sequence of SEQ ID NO:1.
37. A polyclonal antibody produced by a method of claim 36.
38. A composition comprising the polyclonal antibody of claim 37 and a suitable carrier.
39. A method of making a monoclonal antibody with the specificity of the antibody of claim 11, the method comprising:
a) immunizing an animal with a polypeptide consisting of an amino acid sequence of SEQ ID NO:1, or an immunogenic fragment thereof, under conditions to elicit an antibody response,
b) isolating antibody producing cells from the animal,
c) fusing the antibody producing cells with immortalized cells to form monoclonal antibody-producing hybridoma cells,
d) culturing the hybridoma cells, and
e) isolating from the culture monoclonal antibody which binds specifically to a polypeptide comprising an amino acid sequence of SEQ ID NO:1.
40. A monoclonal antibody produced by a method of claim 39.
41. A composition comprising the monoclonal antibody of claim 40 and a suitable carrier.
42. The antibody of claim II, wherein the monoclonal antibody is produced by screening a Fab expression library.
43. The antibody of claim 11, wherein the antibody is produced by screening a recombinant immunoglobulin library.
44. A method of detecting a polypeptide comprising an amino acid sequence of SEQ ID NO:1 in a sample, the method comprising:
a) incubating the antibody of claim 11 with a sample under conditions to allow specific binding of the antibody and the polypeptide, and
b) detecting specific binding, wherein specific binding indicates the presence of a polypeptide comprising an amino acid sequence of SEQ ID NO:1 in the sample.
45. A method of purifying a polypeptide comprising an amino acid sequence of SEQ ID NO:1 from a sample, the method comprising:
a) incubating the antibody of claim 11 with a sample under conditions to allow specific binding of the antibody and the polypeptide, and
b) separating the antibody from the sample and obtaining the purified polypeptide comprising an amino acid sequence of SEQ ID NO:1.
46. A microarray wherein at least one element of the microarray is a polynucleotide of claim 13.
47. A method of generating an expression profile of a sample which contains polynucleotides, the method comprising:
a) labeling the polynucleotides of the sample,
b) contacting the elements of the microarray of claim 46 with the labeled polynucleotides of the sample under conditions suitable for the formation of a hybridization complex, and
c) quantifying the expression of the polynucleotides in the sample.
48. An array comprising different nucleotide molecules affixed in distinct physical locations on a solid substrate, wherein at least one of said nucleotide molecules comprises a first oligonucleotide or polynucleotide sequence specifically hybridizable with at least 30 contiguous nucleotides of a target polynucleotide, and wherein said target polynucleotide is a polynucleotide of claim 12.
49. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to at least 30 contiguous nucleotides of said target polynucleotide.
50. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to at least 60 contiguous nucleotides of said target polynucleotide.
51. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to said target polynucleotide.
52. An array of claim 48, which is a microarray.
53. An array of claim 48, further comprising said target polynucleotide hybridized to a nucleotide molecule comprising said first oligonucleotide or polynucleotide sequence.
54. An array of claim 48, wherein a linker joins at least one of said nucleotide molecules to said solid substrate.
55. An array of claim 48, wherein each distinct physical location on the substrate contains multiple nucleotide molecules, and the multiple nucleotide molecules at any single distinct physical location have the same sequence, and each distinct physical location on the substrate contains nucleotide molecules having a sequence which differs from the sequence of nucleotide molecules at another distinct physical location on the substrate.
US10/235,521 1997-12-12 2002-09-04 Human lysyl hydroxylase-like protein Abandoned US20030087409A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/235,521 US20030087409A1 (en) 1997-12-12 2002-09-04 Human lysyl hydroxylase-like protein

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/989,385 US6130039A (en) 1997-12-12 1997-12-12 Polynucleotide encoding human lysyl hydroxylase-like protein
US09/593,826 US6489444B1 (en) 1997-12-12 2000-06-13 Human lysyl hydroxylase-like protein
US10/235,521 US20030087409A1 (en) 1997-12-12 2002-09-04 Human lysyl hydroxylase-like protein

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/593,826 Division US6489444B1 (en) 1997-12-12 2000-06-13 Human lysyl hydroxylase-like protein

Publications (1)

Publication Number Publication Date
US20030087409A1 true US20030087409A1 (en) 2003-05-08

Family

ID=25535071

Family Applications (3)

Application Number Title Priority Date Filing Date
US08/989,385 Expired - Fee Related US6130039A (en) 1997-12-12 1997-12-12 Polynucleotide encoding human lysyl hydroxylase-like protein
US09/593,826 Expired - Fee Related US6489444B1 (en) 1997-12-12 2000-06-13 Human lysyl hydroxylase-like protein
US10/235,521 Abandoned US20030087409A1 (en) 1997-12-12 2002-09-04 Human lysyl hydroxylase-like protein

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US08/989,385 Expired - Fee Related US6130039A (en) 1997-12-12 1997-12-12 Polynucleotide encoding human lysyl hydroxylase-like protein
US09/593,826 Expired - Fee Related US6489444B1 (en) 1997-12-12 2000-06-13 Human lysyl hydroxylase-like protein

Country Status (1)

Country Link
US (3) US6130039A (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009080898A2 (en) * 2007-12-21 2009-07-02 Oulun Yliopisto Products and methods for preventing tumour growth
US20100035271A1 (en) * 1999-11-29 2010-02-11 Nederlandse Organisatie Voor Toegepast-Natuurwetenschappelijk Onderzoek Tno Modification of Collagenous Materials and Medical Treatment, Diagnosis and Monitoring of Fibrotic Conditions
US20100163651A1 (en) * 2008-12-31 2010-07-01 Feith Raymond P Low Flow Irrigation Emitter
US11932838B2 (en) 2014-11-25 2024-03-19 Corning Incorporated Cell culture media extending materials and methods

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FI20001328A (en) * 2000-06-02 2001-12-03 Oulun Yliopisto Process for production of a multifunctional enzyme
CN1615131A (en) * 2001-11-23 2005-05-11 中外制药株式会社 Method for identification of tumor targeting enzymes
WO2004073601A2 (en) * 2003-02-21 2004-09-02 Chugai Seiyaku Kabushiki Kaisha Process for the preparation of hexacyclic camptothecin derivatives
BRPI0509732A (en) * 2004-04-09 2007-09-25 Chugai Pharmaceutical Co Ltd water soluble prodrug, pharmaceutical composition, therapeutic agent, antifungal agent and use of said prodrug
TW200744603A (en) * 2005-08-22 2007-12-16 Chugai Pharmaceutical Co Ltd Novel anticancer concomitant drug
WO2007046456A1 (en) * 2005-10-19 2007-04-26 Chugai Seiyaku Kabushiki Kaisha Agent for preventing or treating pancreas cancer, ovary cancer or liver cancer containing novel water-soluble prodrug

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4889806A (en) * 1987-04-15 1989-12-26 Washington University Large DNA cloning system based on yeast artificial chromosomes

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4889806A (en) * 1987-04-15 1989-12-26 Washington University Large DNA cloning system based on yeast artificial chromosomes

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100035271A1 (en) * 1999-11-29 2010-02-11 Nederlandse Organisatie Voor Toegepast-Natuurwetenschappelijk Onderzoek Tno Modification of Collagenous Materials and Medical Treatment, Diagnosis and Monitoring of Fibrotic Conditions
WO2009080898A2 (en) * 2007-12-21 2009-07-02 Oulun Yliopisto Products and methods for preventing tumour growth
WO2009080899A2 (en) * 2007-12-21 2009-07-02 Oulun Yliopisto Products and methods affecting cell and tissue growth
WO2009080898A3 (en) * 2007-12-21 2009-10-01 Oulun Yliopisto Products and methods for preventing tumour growth
WO2009080899A3 (en) * 2007-12-21 2009-10-01 Oulun Yliopisto Products and methods affecting cell and tissue growth
US20100163651A1 (en) * 2008-12-31 2010-07-01 Feith Raymond P Low Flow Irrigation Emitter
US11932838B2 (en) 2014-11-25 2024-03-19 Corning Incorporated Cell culture media extending materials and methods

Also Published As

Publication number Publication date
US6130039A (en) 2000-10-10
US6489444B1 (en) 2002-12-03

Similar Documents

Publication Publication Date Title
US6313266B1 (en) Human nucleolin-like protein
US20020086006A1 (en) Human mitochondrial malate dehydrogenase
US6017744A (en) Human N-acetyl transferase
US6210890B1 (en) Human peroxisomal thioesterase
US6365358B1 (en) Ubiquitin-like conjugating protein
US6489444B1 (en) Human lysyl hydroxylase-like protein
US6368810B1 (en) Glutathione S-transferase homolog
US6506571B1 (en) Human glutathione-S-transferase
US20030077747A1 (en) Delta -6 desaturase homologs
US6242179B1 (en) Human phosphatases
US20020173022A1 (en) Human protein phosphatase
US5925543A (en) Isolated polynucleotide sequence encoding NADH dehydrogenase B17 subunit
US6416755B1 (en) Human beta-alanine-pyruvate aminotransferase
US6251389B1 (en) Human nucleotide pyrophosphohydrolase-2
US20030077802A1 (en) Human PRL1 phosphatase
US5952177A (en) Human cytosolic isocitrate dehydrogenase
US20020086393A1 (en) Mitochondrial adenylate kinase
US5869311A (en) Mitochondrial processing peptidase subunit
US6323328B1 (en) Human citrate synthase homolog
US6806071B1 (en) Glutathione S-transferase
US7238506B1 (en) Electron transport proteins
US6004778A (en) Embryogenesis protein
US20030119046A1 (en) Human fatty acid beta-oxidation enzymes
US5976865A (en) Vesicle transport related protein
US20010016648A1 (en) Autoantigen-like protein

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION