WO2004104575A2 - Diagnostics and therapeutics for diseases associated with x-prolyl aminopeptidase 2 (xpnpep2) - Google Patents

Diagnostics and therapeutics for diseases associated with x-prolyl aminopeptidase 2 (xpnpep2) Download PDF

Info

Publication number
WO2004104575A2
WO2004104575A2 PCT/EP2004/005021 EP2004005021W WO2004104575A2 WO 2004104575 A2 WO2004104575 A2 WO 2004104575A2 EP 2004005021 W EP2004005021 W EP 2004005021W WO 2004104575 A2 WO2004104575 A2 WO 2004104575A2
Authority
WO
WIPO (PCT)
Prior art keywords
xpnpep2
disorders
diseases
polypeptide
cancer
Prior art date
Application number
PCT/EP2004/005021
Other languages
French (fr)
Other versions
WO2004104575A3 (en
Inventor
Stefan Golz
Ulf Brüggemeier
Andreas Geerts
Ralf Thiele
Original Assignee
Bayer Healthcare Ag
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 Bayer Healthcare Ag filed Critical Bayer Healthcare Ag
Publication of WO2004104575A2 publication Critical patent/WO2004104575A2/en
Publication of WO2004104575A3 publication Critical patent/WO2004104575A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/37Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving peptidase or proteinase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • the present invention is in the field of molecular biology, more particularly, the present invention relates to nucleic acid sequences and amino acid sequences of a human XP PEP2 and its regulation for the treatment of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in mammals.
  • XP PEP2 is a member of the group of protease enzymes [WO200261131, Nenema et al. (1997), Prueitt et al. (2000)].
  • proteases were recognized very early in the history of biochemistry. In the nineteenth century, one primary focus of research was on digestive proteases, like pepsin and trypsin. Proteases belong systematically to the C- ⁇ Hydrolases. More specifically, proteases catalyze the hyprolytic cleavage of a peptide bond and are therefore called peptidases as well.
  • Proteases can be classified according to several criteria, e.g. by localisation. Digestive proteases are located in the gastro-intestinal tract. These proteases are responsible for the digestion of food proteins. Peptidases located extracellularly in the blood or other extracellular compartments of the body play often regulatory roles in processes like for example blood clotting, fibrinolysis, or activation of complement constituents.
  • Proteases are most commonly classified according to their mechanism of action, or to specific active groups that are present in the active center. The following groups can be distinguished:
  • Serin-peptidases 2. cystein-peptidases, 3. aspartyl- or acidic-peptidases, 4. metallo-peptidases, or 5. peptidases with yet unclear reaction mechanism.
  • a catalytic triad comprised of an aspartate, a histidine and the above mentioned serine is found. This triad functions in the reaction mechanism as a charge relay system.
  • t he digestive enzymes trypsin and chymotrypsin, components of the complement cascade, enzymes involved in the blood clotting cascade, as well as enzymes that function in degradation, rebuilding and maintenace of constituents of the extracellular matrix.
  • serine protease family is the broad range of substrate specificity.
  • trypase subgroup cleave after arginine or lysine, chymases after phenylalanine or leucine, aspases after aspartate, metases after methionine and serases after serine.
  • thioester intermediate is formed by a nucleophilic attack of the cysteine on the caboxy carbon of the peptide bond.
  • a catalytic triad comprised of the cysteine, a histidine and an asparagine is found which functions as a charge relay system to facilitate the formation of the thioester intermediate.
  • cysteine proteases include lysosomal cathepsines, and cytosolic calpains.
  • the catalytic site of aspartyl proteases is composed of two aspartate residues.
  • Aspartyl proteases (2-3) one of the aspartyl carboxy groups is ionized and the other is neutral, which is important for the catalytic reaction to occur.
  • aspartyl proteases are gastric pepsins A and C, chymosin, as well as mammalian renin.
  • Metallo-peptidases are proteases, whose proteolytic activity depends on the presence of divalent cations in the active center. Examples of members of this class are carboxypeptidase A, which represents a pancreatic digestive enzyme, the Angiotension Converting Enzymes (ACE), which are responsible for the conversion of angiotensin I to angiotensin ⁇ , or the Extracellular Matrix Metalloprotienases.
  • ACE Angiotension Converting Enzymes
  • the protease encoded by the HIN genome is used as a target for drugs for the treatment of HIN infections
  • the proteasom complex h as b een discovered as an anti-cancer target, o r C ys-proteases have been implemented as drug targets for inflammatory disorders.
  • Selective inhibitors have been developed as therapeutic agents for diseases such as HIN.
  • the identification of further disease implications of protease species and their splice variants may lead to the development of specific inhibitors or modulators, or suggest new utilities for known compounds affecting proteases. That in turn will provide additional pharmacological approaches to treat diseases and conditions in which protease activities are involved.
  • This diseases may include, but are not limited to, infections such as bacterial, fungal, protozoan, and viral infections, particularly those caused by HIN viruses, cancers, allergies including asthma, cardiovascular diseases including acute heart failure, hypotension, hypertension, angina pectoris, myocardial infarction, hematological diseases, genito-urinary diseases including urinary incontinence and benign prostate hyperplasia, osteoporosis, and peripheral and central nervous system disorders including pain, Alzheimer's disease and Parkinson's disease.
  • infections such as bacterial, fungal, protozoan, and viral infections, particularly those caused by HIN viruses, cancers, allergies including asthma, cardiovascular diseases including acute heart failure, hypotension, hypertension, angina pectoris, myocardial infarction, hematological diseases, genito-urinary diseases including urinary incontinence and benign prostate hyperplasia, osteoporosis, and peripheral and central nervous system disorders including pain, Alzheimer's disease and Parkinson's disease.
  • TaqMan is a recently developed technique, in which the release of a fluorescent reporter dye from a hybridisation probe in real-time during a polymerase chain reaction (PCR) is proportional to the accumulation of the PCR product.
  • PCR polymerase chain reaction
  • Quantification is based on the early, linear part of the reaction, and by determining the threshold cycle (CT), at which fluorescence above background is first detected.
  • CT threshold cycle
  • Gene expression technologies may be useful in several areas of drug discovery and development, such as target identification, lead optimization, and identification of mechanisms of action.
  • the TaqMan technology can be used to compare differences between expression profiles of normal tissue and diseased tissue.
  • Expression profiling has b een used in identifying genes, which are up- or downregulated in a variety of diseases.
  • An interesting application of expression pro filing is temporal monitoring of changes in gene expression during disease progression and drug treatment or in patients versus healthy individuals.
  • the premise in this approach is that changes in pattern of gene expression in response to physiological or environmental stimuli (e.g., drugs) may serve as indirect clues about disease-causing genes or drug targets.
  • physiological or environmental stimuli e.g., drugs
  • the effects of drugs with established efficacy on global gene expression patterns may provide a guidepost, or a genetic signature, against which a new drug candidate can be compared.
  • the nucleotide sequence of XPNPEP2 is accessible in public databases by the accession number NM_003399 and is given in SEQ ID NO:l.
  • the amino acid sequence of XP PEP2 is depicted in SEQ ID NO:2.
  • Aminopeptidase P is a hydrolase specific for N-terminal imido bonds, which are common to several collagen degradation products, neuropeptides, vasoactive peptides, and cytokines. Structurally, the enzyme is a member of the 'pita bread fold' family and occurs in mammalian tissues in both soluble and GPI-anchored membrane-bound forms. A membrane-bound and soluble form of this enzyme have been identified as products of two separate genes.
  • RT-PCR u sing d egenerate o ligonucleotides b ased o n the pig aminopeptidase P amino acid sequence, and by 5-prime and 3-prime RACE, Nenema et al. (1997) isolated human kidney and lung cD ⁇ As encoding XP ⁇ PEP2.
  • the deduced XPNPEP2 protein has 673 amino acids and an estimated molecular mass of 75,490 Da.
  • the authors stated that the human and pig XPNPEP2 amino acid sequences show significant evolutionary divergence, with 83% identity; 5 of 6 potential N- glycosylation sites, and 5 of 6 cysteine residues that are potentially involved in disulfide bond formation, are conserved.
  • Northern blot analysis detected a 3.5-kb XPNPEP2 transcript in human kidney, lung, heart, placenta, liver, small intestine, and colon, but not in brain, skeletal muscle, pancreas, spleen, thymus, prostate, testis, ovary, or leukocytes. Based on these results and on RT-PCR studies, Nenema et al. (1997) suggested that the membrane-bound form and the soluble form, which is found in brain and leukocytes, are the products of 2 different genes or of alternative splicing of a single primary transcript
  • XPNPEP2 is published in WO200261131. XPNPEP2 shows the highest homology (42%) to the human X-Pro aminopeptidase 1 as shown in example 1.
  • the invention relates to novel disease associations of XPNPEP2 polypeptides and polynucleotides.
  • the invention also relates to novel methods of screening for therapeutic agents for the treatment of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal.
  • the invention also relates to pharmaceutical compositions for the treatment of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a XPNPEP2 polypeptide, a XPNPEP2 polynucleotide, or regulators of XPNPEP2 or modulators of XPNPEP2 activity.
  • the invention further comprises methods of diagnosing cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal.
  • Fig. 1 shows the nucleotide sequence of a XPNPEP2 polynucleotide (SEQ ID NO: 1).
  • Fig. 2 shows the amino acid sequence of a XPNPEP2 polypeptide (SEQ ID NO:2).
  • Fig. 3 shows the nucleotide sequence of a primer useful for the invention (SEQ ID NO:
  • Fig. 4 shows the nucleotide sequence of a primer useful for the invention (SEQ ID NO:4).
  • Fig. 5 shows a nucleotide sequence useful as a probe to detect proteins of the invention (SEQ ID NO:5).
  • oligonucleotide is a stretch of nucleotide residues which has a sufficient number of bases to be used as an oligomer, amplimer or probe in a polymerase chain reaction (PCR). Oligonucleotides are prepared from genomic or cDNA sequence and are used to amplify, reveal, or confirm the presence of a similar DNA or RNA in a particular cell or tissue. Oligonucleotides or oligomers comprise portions of a DNA sequence having at least about 10 nucleotides and as many as about 35 nucleotides, preferably about 25 nucleotides.
  • Probes may be derived from naturally occurring or recombinant single- or double- stranded nucleic acids or may be chemically synthesized. They are useful in detecting the presence of identical or similar sequences. Such probes may be labeled with reporter molecules using nick translation, Klenow fill-in reaction, PCR or other methods well known in the art. Nucleic acid probes may be used in southern, northern or in situ hybridizations to determine whether DNA or RNA encoding a certain protein is present in a cell type, tissue, or organ.
  • a “fragment of a polynucleotide” is a nucleic acid that comprises all or any part of a given nucleotide molecule, the fragment having fewer nucleotides than about 6 kb, preferably fewer than about 1 kb.
  • Reporter molecules are radionuclides, enzymes, fluorescent, chemiluminescent, or chromogenic agents which associate with a particular nucleotide or amino acid sequence, thereby establishing the presence of a certain sequence, or allowing for the quantification of a certain sequence.
  • Chimeric molecules may be constructed by introducing all or part of the nucleotide sequence of this invention into a vector containing additional nucleic acid sequence which might be expected to change any one or several of the following XPNPEP2 characteristics: cellular location, distribution, ligand-binding affinities, interchain affinities, degradation/turnover rate, signaling, etc.
  • XPNPEP2 polypeptide refers to those forms, fragments, or domains of a XPNPEP2 polypeptide which retain the biological and/or antigenic activity of a XPNPEP2 polypeptide.
  • “Naturally occurring XPNPEP2 polypeptide” refers to a polypeptide produced by cells which have not been genetically engineered and specifically contemplates various polypeptides arising from post-translational modifications of the polypeptide including but not limited to acetylation, carboxylation, glycosylation, phos- phorylation, lipidation and acylation.
  • Derivative refers to polypeptides which have been chemically modified by techniques such as ubiquitination, labeling (see above), pegylation (derivatization with polyethylene glycol), and chemical insertion or substitution of amino acids such as ornithine which do not normally occur in human proteins.
  • Constant amino acid substitutions result from replacing one amino acid with another having similar structural and/or chemical properties, such as the replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, or a threonine with a serine.
  • “Insertions” or “deletions” are typically in the range of about 1 to 5 amino acids. The variation allowed may be experimentally determined by producing the peptide synthetically while systematically making insertions, deletions, or substitutions of nucleotides in the sequence using recombinant DNA techniques.
  • a “signal sequence” or “leader sequence” can be used, when desired, to direct the polypeptide through a membrane of a cell.
  • Such a sequence may be naturally present on the polypeptides of the present invention or provided from heterologous sources by recombinant DNA techniques.
  • oligopeptide i s a s hort s tretch of amino acid residues and may be expressed from an oligonucleotide.
  • Ohgopeptides comprise a stretch of amino acid residues of at least 3, 5, 10 amino acids and at most 10, 15, 25 amino acids, typically of at least 9 to 13 amino acids, and of sufficient length to display biological and/or antigenic activity.
  • “Inhibitor” i s any substance which retards or prevents a chemical or physiological reaction or response. Common inhibitors include but are not limited to antisense molecules, antibodies, and antagonists.
  • Standard expression is a quantitative or qualitative measurement for comparison. It is based on a statistically appropriate number of normal samples and is created to use as a basis of comparison when performing diagnostic assays, running clinical trials, or following patient treatment profiles.
  • Animal as used herein may be defined to include human, domestic (e.g., cats, dogs, etc.), agricultural (e.g., cows, horses, sheep, etc.) or test species (e.g., mouse, rat, rabbit, etc.).
  • domestic e.g., cats, dogs, etc.
  • agricultural e.g., cows, horses, sheep, etc.
  • test species e.g., mouse, rat, rabbit, etc.
  • nucleic acid molecules encoding a polypeptide comprising the amino acid sequence of SEQ ID NO: 2,
  • nucleic acid molecules comprising the sequence of SEQ ID NO: 1,
  • nucleic acid molecules the complementary strand of which hybridizes under stringent conditions to a nucleic acid molecule of (i), (ii), or (iii);
  • nucleic acid molecules the sequence of which differs from the sequence of a nucleic acid molecule of (iii) due to the degeneracy of the genetic code; wherein the polypeptide encoded by said nucleic acid molecule has XPNPEP2 activity.
  • XPNPEP2 polypeptide within the meaning of the invention, shall be understood as being a polypeptide selected from a group consisting of
  • polypeptides comprising the sequence of SEQ LD NO: 2
  • polypeptides which show at least 99%, 98%, 95%, 90%, or 80% homology with a polypeptide of (i), (ii), or (iii);
  • polypeptide has XPNPEP2 activity.
  • nucleotide sequences encoding a XPNPEP2 have numerous applications in techniques known to those skilled in the art of molecular biology. These techniques include use as hybridization probes, use in the construction of oligomers for PCR, use for chromosome and gene mapping, use in the recombinant production of XPNPEP2, and use in generation of antisense DNA or RNA, their chemical analogs and the like. Uses of nucleotides encoding a XPNPEP2 disclosed h erein are exemplary o f known techniques and are not intended to limit their use in any technique known to a person of ordinary skill in the art.
  • nucleotide sequences disclosed herein may be used in molecular biology techniques that have not yet been developed, provided the new techniques rely on properties of nucleotide sequences that are currently known, e.g., the triplet genetic code, specific base pair interactions, etc. It will be appreciated by those skilled in the art that as a result of the degeneracy of the genetic code, a multitude of XPNPEP2 - encoding nucleotide sequences may be produced. Some of these will only bear minimal homology to the nucleotide sequence of the known and naturally occurring XPNPEP2.
  • the invention has specifically contemplated each and every possible variation of nucleotide 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 nucleotide sequence of naturally occurring XPNPEP2, and all such variations are to be considered as being specifically disclosed.
  • nucleotide sequences which encode a XPNPEP2 are preferably capable of hybridizing to the nucleotide sequence of the naturally occurring XPNPEP2 polynucleotide under stringent conditions, it may be advantageous to produce nucleotide sequences encoding XPNPEP2 polypeptides or its derivatives possessing a substantially different codon usage. Codons can be selected to increase the rate at which expression of the peptide occurs in a particular prokaryotic or eukaryotic expression host in accordance with the frequency with which particular codons are utilized by the host.
  • RNA transcripts having more desirable properties such as a greater half-life, than transcripts produced from the naturally occurring sequence.
  • Nucleotide sequences encoding a XPNPEP2 polypeptide may be joined to a variety of other nucleotide sequences by means of well established recombinant DNA techniques.
  • U seful nucleotide sequences for j oining to XPNPEP2 polynucleotides include an assortment of cloning vectors such as plasmids, cosmids, lambda phage derivatives, phagemids, and the like.
  • Vectors of interest include expression vectors, replication vectors, probe generation vectors, sequencing vectors, etc. In general, vectors of interest may contain an origin of replication functional in at least one organism, convenient restriction endonuclease sensitive sites, and selectable markers for one or more host cell systems.
  • Another aspect of the subject invention is to provide for XPNPEP2-specific hybridi- zation probes capable of hybridizing with naturally occurring nucleotide sequences encoding XPNPEP2. Such probes may also be used for the detection of similar protease encoding sequences and should preferably show at least 40% nucleotide identity to XPNPEP2 polynucleotides.
  • the hybridization probes of the subject invention may be derived from the nucleotide sequence presented as SEQ ID NO: 1 or from genomic sequences including promoter, enhancers or introns of the native gene. Hybridization probes may be labelled by a variety of reporter molecules using techniques well known in the art.
  • the invention relates to nucleic acid sequences that hybridize with such XPNPEP2 encoding nucleic acid sequences under stringent conditions.
  • Stringent conditions refers to conditions that allow for the hybridization of substantially related nucleic acid sequences. For instance, such conditions will generally allow hybridization of sequence with at least about 85% sequence identity, preferably with at least about 90% sequence identity, more preferably with at least about 95% sequence identity. Hybridization conditions and probes can be adjusted in well-characterized ways to achieve selective hybridization of human-derived probes. Stringent conditions, within the meaning of the invention are 65°C in a buffer containing 1 mM EDTA, 0.5 MNaHPO 4 (pH 7.2), 7 % (w/v) SDS.
  • Nucleic acid molecules that will hybridize to XPNPEP2 polynucleotides under stringent conditions can be identified functionally.
  • examples of the uses for hybridization probes include: histochemical uses such as identifying tissues that express XPNPEP2; measuring mRNA levels, for instance to identify a sample's tissue type or to identify cells that express abnormal levels of XPNPEP2; and detecting polymorphisms of XPNPEP2.
  • PCR provides additional uses for oligonucleotides based upon the nucleotide sequence which encodes XPNPEP2.
  • probes used in PCR may be of recombinant origin, chemically synthesized, or a mixture of both.
  • Oligomers may comprise discrete nucleotide sequences employed under optimized conditions for identification of XPNPEP2 in specific tissues or diagnostic use. The same two oligomers, a nested set of oligomers, or even a degenerate pool of oligomers may be employed under less stringent conditions for identification of closely related DNAs or RNAs.
  • PCR primers i.e., preparations of primers that are heterogeneous at given sequence locations, can be designed to amplify nucleic acid sequences that are highly homologous to, but not identical with XPNPEP2.
  • nucleic acid primers can be ligated to the nucleic acid sought to be amplified to provide the hybridization partner for one of the primers. In this way, only one of the primers need be based on the sequence of the nucleic acid sought to be amplified.
  • PCR methods for amplifying nucleic acid will utilize at least two primers.
  • One of these primers will be capable of hybridizing to a first strand of the nucleic acid to be amplified and of priming enzyme-driven nucleic acid synthesis in a first direction.
  • the other will be capable of hybridizing the reciprocal sequence of the first strand (if the sequence to be amplified is single stranded, this sequence will initially be hypothetical, but will be synthesized in the first amplification cycle) and of priming nucleic acid synthesis from that strand in the direction opposite the first direction and towards the site of hybridization for the first primer.
  • Conditions for conducting such amplifications particularly under preferred stringent hybridization conditions, are well known.
  • RNA polymerase as T7 or SP6 RNA polymerase and the appropriate reporter molecules.
  • nucleic acid sequence can be inserted into any of the many available DNA vectors and their respective host cells using techniques which are well known in the art.
  • synthetic chemistry may be used to introduce mutations into the nucleotide sequence. Alternately, a portion of sequence in which a mutation is desired can be synthesized and recombined with longer portion of an existing genomic or recombinant sequence.
  • XPNPEP2 polynucleotides may be used to produce a purified oligo-or polypeptide using well known methods of recombinant DNA technology.
  • the ohgopeptide may be expressed in a variety of host cells, either prokaryotic or eukaryotic. Host cells may be from the same species from which the nucleotide sequence was derived or from a different species. Advantages of producing an oligonucleotide by recombinant DNA technology include obtaining adequate amounts of the protein for purification and the availability of simplified purification procedures.
  • Chromosome-based techniques such as comparative genomic hybridization (CGH) and fluorescent in situ hybridization (FISH) facilitate efforts to cytogenetically localize genomic regions that are altered in tumor cells. Regions of genomic alteration can be narrowed further using loss of heterozygosity analysis (LOH), in which disease DNA is analyzed and compared with normal DNA for the loss of a heterozygous polymorphic marker.
  • LH loss of heterozygosity analysis
  • RFLPs restriction fragment length polymorphisms
  • Johnson, (1989) or hyper- variable minisatellite DNA [Barnes, 2000].
  • LOH has been performed primarily using PCR amplification of microsatellite markers and electrophoresis of the radio labelled [Jeffreys, (1985)] or fluorescently labelled PCR products [Weber, (1990)] and compared between paired normal and disease DNAs.
  • a gene sequence contained in all samples at relatively constant quantity is typically utilized for sample amplification efficiency normalization.
  • This approach suffers from several drawbacks.
  • the method requires that each s ample has equal input amounts of the nucleic acid and that the amplification efficiency between samples is identical until the time of analysis.
  • Fluorogenic nuclease assays are a real time quantitation method that uses a probe to monitor formation of amplification product.
  • the basis for this method of monitoring the formation of amplification product is to measure continuously PCR product accumulation u sing a dual-labelled fluorogenic oligonucleotide probe, an approach frequently referred to in the literature simply as the "TaqMan method” [Piatak,(1993), Science; Heid, (1996); Gibson, (1996); Holland. (1991)].
  • the probe used in such assays is typically a short (about 20-25 bases) oligonucleotide that is labeled with two different fluorescent dyes.
  • the 5' terminus of the probe is attached to a reporter dye and the 3' terminus is attached to a quenching dye, although the dyes could be attached at other locations on the probe as well.
  • the probe is designed to have at least substantial sequence complementarity with the probe binding site. Upstream and downstream PCR primers which bind to flanking regions of the locus are added to the reaction mixture. When the probe is intact, energy transfer between the two fluorophors occurs and the quencher quenches emission from the reporter.
  • the probe is cleaved by the 5' nuclease activity of a nucleic acid polymerase such as Taq polymerase, thereby releasing the reporter from the oligonucleotide-quencher and resulting in an increase of reporter emission intensity which can be measured by an appropriate detector.
  • a nucleic acid polymerase such as Taq polymerase
  • One detector which is specifically adapted for measuring fluorescence emissions such as those created during a fluorogenic assay is the ABI 7700 or 4700 HT manufactured by Applied Biosystems, Inc. in Foster City, Calif.
  • the ABI 7700 uses fiber o ptics c onnected w ith e ach w ell i n a 96-or 3 84 w ell P CR tube arrangement.
  • the instrument includes a laser for exciting the labels and is capable of measuring the fluorescence spectra intensity from each tube with continuous monitoring during PCR amplification. Each tube is re-examined every 8.5 seconds.
  • Computer software provided with the instrument is capable of recording the fluorescence intensity of reporter and quencher over the course of the amplification. The recorded values will then be used to calculate the increase in normalized reporter emission intensity on a continuous basis. The increase in emission intensity is plotted versus time, i.e., the number of amplification cycles, to produce a continuous measure of amplification.
  • the amplification plot is examined at a point during the log phase of product accumulation. This is accomplished by assigning a fluorescence threshold intensity above background and determining the point at which each amplification plot crosses the threshold (defined as the threshold cycle number or Ct). Differences in threshold cycle number are used to quantify the relative amount of PCR target contained within each tube. Assuming that each reaction functions at 100% PCR efficiency, a difference of one Ct represents a two-fold difference in the amount of starting template.
  • the fluorescence value can be used in conjunction with a standard curve to determine the amount of amplification product present.
  • amplification product (which is double stranded) binds dye molecules in solution to form a complex.
  • dyes it is possible to distinguish between dye molecules free in solution and dye molecules bound to amplification product. For example, certain dyes fluoresce only when bound to amplification product. Examples of dyes which can be used in methods of this general type include, but are not limited to, Syber Green.TM.
  • These detection methods involve some alteration to the structure or conformation of a probe hybridized to the locus between the amplification primer pair.
  • the alteration is caused by the template-dependent extension catalyzed by a nucleic acid polymerase during the amplification process.
  • the alteration generates a detectable signal which is an indirect measure of the amount of amplification product formed.
  • some methods involve the degradation or digestion of the probe during the extension reaction. These methods are a consequence of the 5'-3' nuclease activity associated with some nucleic acid polymerases. Polymerases having this activity cleave mononucleotides or small o ligonucleotides from an o ligonucleotide probe annealed to its complementary sequence located within the locus.
  • the 3' end of the upstream primer provides the initial binding site for the nucleic acid polymerase.
  • the nucleic acid polymerase displaces a portion of the 5' end of the probe and through its nuclease activity cleaves mononucleotides or oligonucleotides from the probe.
  • the upstream primer and the probe can be designed such that they anneal to the complementary strand in close proximity to one another. In fact, the 3' end of the upstream primer and the 5' end of the probe may abut one another. In this situation, extension of the upstream primer is not necessary in order for the nucleic acid polymerase to begin cleaving the probe. In the case in which intervening nucleotides separate the upstream primer and the probe, extension of the primer is necessary before the nucleic acid polymerase encounters the 5' end of the probe.
  • the 5'-3' exonuclease activity of the nucleic acid polymerase begins cleaving mononucleotides or oligonucleotides from the 5' end of the probe. Digestion of the probe continues until the remaining portion of the probe dissociates from the complementary strand.
  • the two end sections can hybridize with each other to form a hairpin loop.
  • the reporter and quencher dye are in sufficiently close proximity that fluorescence from the reporter dye is effectively quenched by the quencher dye.
  • Hybridized probe in contrast, results in a linearized conformation in which the extent of quenching is decreased.
  • the labeled probe is selected so that its sequence is substantially complementary to a segment of the test locus or a reference locus. As indicated above, the nucleic acid site to which the probe binds should be located between the primer binding sites for the upstream and downstream amplification primers.
  • the primers used in the amplification are selected so as to be capable of hybridizing to sequences at flanking regions of the locus being amplified.
  • the primers are chosen to have at least substantial complementarity with the different strands of the nucleic acid b eing amplified.
  • the primers are selected in such that they flank the probe, i.e. are located upstream and downstream of the probe.
  • the primer must have sufficient length so that it is capable of priming the synthesis of extension products in the presence of an agent for polymerization.
  • the length and composition of the primer depends on many parameters, including, for example, the temperature at which the annealing reaction is conducted, proximity of the probe binding site to that of the primer, relative concentrations of the primer and probe and the particular nucleic acid composition of the probe.
  • the primer includes
  • the length of the primer may be more or less depending on the complexity of the primer binding site and the factors listed above.
  • the labels used for labeling the probes or primers of the current invention and which can provide the signal corresponding to the quantity of amplification product can take a variety o f forms.
  • a s indicated above with regard to the 5' fluorogenic nuclease method a fluorescent signal is one signal which can be measured. However, measurements may also be made, for example, by monitoring radioactivity, colorimetry, absorption, magnetic parameters, or enzymatic activity.
  • labels which can be employed include, but are not limited to, fluorophors, chromophores, radioactive isotopes, electron dense reagents, enzymes, and ligands having specific binding partners (e.g., biotin-avidin).
  • a number of labels useful for attachment to probes or primers are commercially available including fluorescein and various fluorescein derivatives such as FAM, HEX, TET and JOE (all which are available from Applied Biosystems, Foster City, Calif); lucifer yellow, and coumarin derivatives.
  • Labels may be attached to the probe or primer using a variety of techniques and can be attached at the 5' end, and or the 3' end and/or at an internal nucleotide.
  • the label can also be attached to spacer arms of various sizes which are attached to the probe or primer. T hese spacer arms are useful for obtaining a desired distance between multiple labels attached to the probe or primer.
  • a single label may be utilized; whereas, in other instances, such as with the 5' fluorogenic nuclease assays for example, two or more labels are attached to the probe.
  • the probe includes multiple labels, it is generally advisable to maintain spacing between the labels which is sufficient to permit separation of the labels during digestion of the probe through the 5'-3' nuclease activity of the nucleic acid polymerase.
  • a number of diseases are associated with changes in the copy number of a certain gene.
  • the real-time PCR method can be used to determine if the patient has copy number alterations which are known to be linked with diseases that are associated with the symptoms the patient has.
  • Fusion proteins are useful for generating antibodies against XPNPEP2 polypeptides and for use in various assay systems. For example, fusion proteins can be used to identify proteins which interact with portions of XPNPEP2 polypeptides. Protein affinity chromatography or library-based assays for protein-protein interactions, such as the yeast two-hybrid or phage display systems, can be used for this purpose. Such methods are well known in the art and also can be used as drug screens.
  • a XPNPEP2 fusion protein comprises two polypeptide segments fused together by means of a peptide bond.
  • the first polypeptide segment can comprise at least 54, 75, 100, 125, 139, 150, 175, 200, 225, 250, 275, 300, 325 or 350 contiguous amino acids of SEQ ID NO: 2 or of a biologically active variant, such as those described above.
  • the first polypeptide segment also can comprise full-length XPNPEP2.
  • the second polypeptide segment can be a full-length protein or a protein fragment.
  • Proteins commonly used in fusion protein construction include, but are not limited to ⁇ galactosidase, ⁇ -glucuronidase, green fluorescent protein (GFP), autofluorescent proteins, including blue fluorescent protein (BFP), glutathione-S-transferase (GST), luciferase, horseradish peroxidase (HRP), and chloramphenicol acetyltransferase (CAT).
  • epitope tags are used in fusion protein constructions, including histidine (His) tags, FLAG tags, influenza hemagglutinin (HA) tags, Myc tags, NSN- G tags, and thioredoxin (Trx) tags.
  • Other fusion constructions can include maltose binding protein (MBP), S -tag, Lex a D ⁇ A binding domain (DBD) fusions, GAL4 D ⁇ A binding domain fusions, and herpes simplex virus (HSN) BP16 protein fusions.
  • a fusion protein also can be engineered to contain a cleavage site located adjacent to the XP ⁇ PEP2.
  • a naturally occurring XPNPEP2 polynucleotide can be isolated free of other cellular components such as membrane components, proteins, and lipids.
  • Polynucleotides can be made by a cell and isolated using standard nucleic acid purification techniques, or synthesized using an amplification technique, such as the polymerase chain reaction (PCR), or by using an automatic synthesizer. Methods for isolating polynucleotides are routine and are known in the art. Any such technique for obtaining a polynucleotide can be used to obtain isolated XPNPEP2 polynucleotides. For example, restriction enzymes and probes can be used to isolate polynucleotide fragments which comprise XPNPEP2 nucleotide sequences. Isolated polynucleotides are in preparations which are free or at least 70, 80, or 90% free of other molecules.
  • XPNPEP2 cDNA molecules can be made with standard molecular biology techniques, using XPNPEP2 mRNA as a template. XPNPEP2 cDNA molecules can thereafter be replicated using molecular biology techniques known in the art. A n amplification technique, such as PCR, can be used to obtain additional copies of polynucleotides of the invention, using either human genomic DNA or cDNA as a template.
  • XPNPEP2 polynucleotides can be synthesized using synthetic chemistry techniques to synthesize XPNPEP2 polynucleotides.
  • the degeneracy of the genetic code allows alternate nucleotide sequences to be synthesized which will encode XPNPEP2 having, for example, an amino acid sequence shown in SEQ ID NO: 2 or a biologically active variant thereof.
  • PCR-based methods can be used to extend nucleic acid sequences encoding human XPNPEP2, for example to detect upstream sequences of XPNPEP2 gene such as promoters and regulatory elements.
  • restriction-site PCR uses universal primers to retrieve unknown sequence adjacent to a known locus. 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.
  • Inverse PCR also can be used to amplify or extend sequences using divergent primers based on a known region.
  • 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.
  • capture PCR which involves PCR amplification of DNA fragments adjacent to a known sequence in human and yeast artificial chromosome DNA.
  • multiple restriction enzyme digestions and ligations also can be used to place an engineered double-stranded sequence into an unknown fragment of the DNA molecule before performing PCR.
  • Randomly-primed libraries are preferable, in that they will contain 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 can be useful for extension of sequence into 5' non-transcribed regulatory regions.
  • capillary electrophoresis systems can be used to analyze the size or confirm the nucleotide sequence of PCR or sequencing products.
  • capillary sequencing can employ flowable polymers for electrophoretic separation, four different fluorescent dyes (one for each nucleotide) which are laser activated, and detection of the emitted wavelengths by a charge coupled device camera.
  • Output/light intensity can be converted to electrical signal using appropriate equipment and software (e.g., GENOTYPER and Sequence NAVIGATOR, Perkin Elmer), and the entire process from loading of samples to computer analysis and electronic data display can 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.
  • XPNPEP2 can be obtained, for example, by purification from human cells, by expression of XPNPEP2 polynucleotides, or by direct chemical synthesis.
  • XPNPEP2 can be purified from any human cell which expresses the enzyme, including those which have been transfected with expression constructs which express XPNPEP2.
  • a purified XPNPEP2 is separated from other compounds which normally associate with XPNPEP2 in the cell, such as certain proteins, carbohydrates, or lipids, using methods well-known in the art. Such methods include, but are not limited to, size exclusion chromatography, ammonium sulfate fractionation, ion exchange chromatography, affinity chromatography, and preparative gel electrophoresis.
  • XPNPEP2 polynucleotides can be inserted into an expression vector which contains the necessary elements for the transcription and translation of the inserted coding sequence.
  • Methods which are well known to those skilled in the art can be used to construct expression vectors containing sequences encoding XPNPEP2 and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
  • a variety of expression vector/host systems can be utilized to contain and express sequences encoding XPNPEP2. 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., baculo virus), plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMN) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids), or animal cell systems.
  • 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., baculo virus), plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco
  • control elements or regulatory sequences are those non-translated regions of the vector - enhancers, promoters, 5' and 3' untranslated regions — which interact with host cellular proteins to carry out transcription and translation. Such elements can 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 i nducible p romoters, c an b e used. F or example, when cloning in bacterial systems, inducible promoters such as the hybrid lacZ promoter of the
  • BLUESCREPT phagemid (Stratagene, LaJolla, Calif.) or pSPORTl plasmid (Life Technologies) and the like can be used.
  • the baculovirus polyhedrin promoter can 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) can be cloned into the vector. In mammalian cell systems, promoters from mammalian genes or from mammalian viruses are preferable.
  • vectors based on SN40 or EBN can be used with an appropriate selectable marker.
  • a number of expression vectors can be selected.
  • vectors which direct high level expression of fusion proteins that are readily purified can be used.
  • Such vectors include, but are not limited to, multifunctional E. coli cloning and expression vectors such as BLUESCRIPT (Stratagene).
  • BLUESCRIPT a sequence encoding XPNPEP2 can be ligated into the vector in frame with sequences for the amino-terminal Met and the subsequent 7 residues of ⁇ -galactosidase so that a hybrid protein is produced.
  • pIN vectors or pGEX vectors also can be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adso ⁇ tion to glutathione-agarose beads followed by elution in the presence of free glutathione.
  • Proteins made in such systems can 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.
  • sequences encoding XPNPEP2 can be driven by any of a number of promoters.
  • viral promoters such as the 35S and 19S promoters of CaMV can be used alone or in combination with the omega leader sequence from TMN.
  • plant promoters such as the small subunit of RUBISCO or heat shock promoters can be used. These constructs can be introduced into plant cells by direct D ⁇ A transformation or by pathogen-mediated transfection.
  • An insect system also can be used to express XP ⁇ PEP2.
  • Autographa californica nuclear polyhedrosis virus (AcNPN) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae.
  • Sequences encoding XP ⁇ PEP2 can 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 XPNPEP2 will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein.
  • the recombinant viruses can then be used to infect S. frugiperda cells or Trichoplusia larvae in which XPNPEP2 can be expressed.
  • a number of viral-based expression systems can b e used to express XPNPEP2 in mammalian host cells.
  • sequences encoding XPNPEP2 can be ligated into an adenovirus transcription/- translation complex comprising the late promoter and tripartite leader sequence. Insertion in a non-essential El or E3 region of the viral genome can be used to obtain a viable virus which is capable of expressing XPNPEP2 in infected host cells [Engelhard, 1 994)].
  • transcription enhancers such as the Rous sarcoma virus (RSV) enhancer, can be used to increase expression in mammalian host cells.
  • RSV Rous sarcoma virus
  • HACs Human artificial chromosomes
  • HACs also can be used to deliver larger fragments of DNA than can be contained and expressed in a plasmid.
  • HACs of 6M to 10M are constructed and delivered to cells via conventional delivery methods (e.g., liposomes, polycationic amino polymers, or vesicles).
  • Specific initiation signals also can be used to achieve more efficient translation of sequences encoding XPNPEP2. Such signals include the ATG initiation codon and adjacent sequences. I n cases where sequences encoding XPNPEP2, its initiation codon, and upstream sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed.
  • exogenous translational control signals including the ATG initiation codon
  • the initiation codon should be in the correct reading frame to ensure translation of the entire insert.
  • Exogenous translational elements and initiation codons can be of various origins, both natural and synthetic.
  • a host cell strain can be chosen for its ability to modulate the expression of the inserted sequences or to process the expressed XPNPEP2 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 polypeptide also can 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; 10801 University Boulevard, Manassas, VA 20110-2209) and can be chosen to ensure the correct modification and processing of the foreign protein.
  • cell lines which stably express XPNPEP2 can be transformed using expression vectors which can contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate v ector. F ollowing t he i ntroduction o f t he v ector, c ells can be allowed to grow for 1-2 days in an enriched medium before they are switched to a selective medium.
  • 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 XPNPEP2 sequences.
  • Resistant clones of stably transformed cells can be proliferated using tissue culture techniques appropriate to the cell type. Any number of selection systems can be used to recover transformed cell lines. These include, but are not limited to, the he ⁇ es simplex virus thymidine kinase [Logan, (1984)] and adenine phosphoribosyltransferase [Wigler, (1977)] genes which can be employed in tic or aprf cells, respectively. Also, antimetabolite, antibiotic, or herbicide resistance can be used as the basis for selection.
  • dhfr confers resistance to methotrexate [Lowy, (1980)]
  • npt confers resistance to the aminoglycosides, neomycin and G-418 [Wigler, (1980)]
  • als and pat confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively [Colbere-Garapin, 1981]. Additional selectable genes have been described.
  • tYpB allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine.
  • Visible markers such as anthocyanins, ⁇ -glucuronidase and its substrate GUS, and luciferase and its substrate luciferin, can be used to identify transformants and to quantify the amount of transient or stable protein expression attributable to a specific vector system
  • a XPNPEP2 polynucleotide is also present, its presence and expression may need to be confirmed.
  • a sequence encoding XPNPEP2 is inserted within a marker gene sequence, transformed cells containing sequences which encode XPNPEP2 can b e identified by the absence of marker gene function.
  • a marker gene can be placed in tandem with a sequence encoding XPNPEP2 under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of XPNPEP2 polynucleotide.
  • host cells which contain a XPNPEP2 polynucleotide and which express XPNPEP2 can 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.
  • nucleic acid amplification-based assays involve the use of oligonucleotides selected from sequences encoding XPNPEP2 to detect transformants which contain a XPNPEP2 polynucleotide.
  • a variety of protocols for detecting and measuring the expression of XPNPEP2, using either polyclonal or monoclonal antibodies specific for the polypeptide, are known in the art.
  • Examples include enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and fluorescence activated cell sorting (FACS).
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • FACS fluorescence activated cell sorting
  • Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides encoding XPNPEP2 include oligolabeling, nick translation, end- labeling, or PCR amplification using a labeled nucleotide.
  • sequences encoding XPNPEP2 can be cloned into a vector for the production of an mRNA probe.
  • Such vectors are known in the art, are commercially available, and can be used t o s ynthesize RNA p robes i n v itro b y a ddition o f 1 abeled nucleotides and an appropriate RNA polymerase such as T7, T3, or SP6. These procedures can be conducted using a variety of commercially available kits (Amersham Pharmacia Biotech, Promega, and US Biochemical). Suitable reporter molecules or labels which can be used for ease of detection include radionuclides, enzymes, and fluorescent, chemiluminescent, or chromogenic agents, as well as substrates, cofactors, inhibitors, magnetic particles, and the like.
  • Host cells transformed with XPNPEP2 polynucleotides can be cultured under conditions suitable for the expression and recovery of the protein from cell culture.
  • the polypeptide produced by a transformed cell can be secreted or contained intracellularly depending on the sequence and/or the vector used.
  • expression vectors containing XPNPEP2 polynucleotides can be designed to contain signal sequences which direct secretion of soluble XPNPEP2 through a prokaryotic or eukaryotic cell membrane or which direct the membrane insertion of membrane-bound XPNPEP2.
  • XPNPEP2 to a nucleotide sequence encoding a polypeptide domain which will facilitate purification of soluble proteins.
  • 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
  • cleavable linker sequences such as those specific for Factor XA or enterokinase (Invitrogen, San Diego, CA) between the purification domain and XPNPEP2 also can be used to facilitate purification.
  • cleavable linker sequences such as those specific for Factor XA or enterokinase (Invitrogen, San Diego, CA) between the purification domain and XPNPEP2 also can be used to facilitate purification.
  • cleavable linker sequences such as those specific for Factor XA or enterokinase (Invitrogen, San Diego, CA) between the purification domain and XPNPEP2 also can be used to facilitate purification.
  • One such expression vector provides for expression
  • Sequences encoding XPNPEP2 can be synthesized, in whole or in part, using chemical methods well known in the art. Alternatively, XPNPEP2 itself can be
  • XPNPEP2 can be separately synthesized and 0 combined using chemical methods to produce a full-length molecule.
  • the newly synthesized peptide can be substantially purified by preparative high performance liquid chromatography.
  • the composition of a synthetic XPNPEP2 can be confirmed by amino acid analysis or sequencing. Additionally, any portion of the amino acid sequence of XPNPEP2 can be altered during direct synthesis and/or combined using chemical methods with sequences from other proteins to produce a variant polypeptide or a fusion protein.
  • XPNPEP2 polynucleotides possessing non-narurally 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.
  • nucleotide sequences referred to herein can be engineered using methods generally known in the art to alter XPNPEP2 polynucleotides for a variety of reasons, including but not limited to, alterations which modify the cloning, processing, and/or expression of the polypeptide or mRNA product.
  • DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides can be used to engineer the nucleotide sequences.
  • site-directed mutagenesis can be used to insert new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, introduce mutations, and so forth.
  • XPNPEP2 analogs are variants having an amino acid sequence that is a mutation of the amino acid sequence disclosed herein.
  • Another general class of XPNPEP2 analogs is provided by anti-idiotype antibodies, and fragments thereof, as described below.
  • recombinant antibodies comprising anti-idiotype variable domains can be used as analogs (see, for example, [Monfardini et al., (1996)]). Since the variable domains of anti-idiotype XPNPEP2 antibodies mimic XPNPEP2, these domains can provide XPNPEP2 enzymatic activity.
  • Methods of producing anti-idiotypic catalytic antibodies are known to those of skill in the art [Joron et al., (1992), Friboulet et al. (1994), Avalle et al., (1998)].
  • XPNPEP2 One illustrative in vitro use of XPNPEP2 and its analogs is the production of labeled peptides from a labeled protein substrate.
  • Proteases can also be used in detergents and cleaning solutions.
  • serine proteases are used in solutions to clean and to disinfect contact lenses (see, for example, [U.S. 5,985,629]).
  • Another use for a serine protease is in the formulation of vaccines (see, for example, [U.S. 5,885,814]).
  • Those of skill in the art can devise other uses for molecules having XPNPEP2 activity.
  • Any type of antibody known in the art can be generated to bind specifically to an epitope of XPNPEP2.
  • Antibody as used herein includes intact immunoglobulin molecules, as well as fragments thereof, such as Fab, F(ab') 2 , and Fv, which are capable of binding an epitope of XPNPEP2.
  • Fab fragment antigen binding protein
  • F(ab') 2 fragment antigen binding protein 2
  • Fv fragment antigen binding protein 2
  • Fab fragment antigen binding protein 2
  • Fv fragment antigen binding protein 2
  • epitope Typically, at least 6, 8, 10, or 12 contiguous amino acids are required to form an epitope. However, epitopes which involve non-contiguous amino acids may require more, e.g., at least 15, 25, or 50 amino acid.
  • An antibody which specifically binds to an epitope of XPNPEP2 can be used therapeutically, as well as in immunochemical assays, such as Western blots, ELISAs, radioimmunoassays, immunohistochemical assays, immunoprecipitations, or other immunochemical assays known i n t he a rt.
  • immunochemical assays such as Western blots, ELISAs, radioimmunoassays, immunohistochemical assays, immunoprecipitations, or other immunochemical assays known i n t he a rt.
  • N arious i mmunoassays c an b e u sed t identify antibodies having the desired specificity.
  • Numerous protocols for competitive binding or immunoradiometric assays are well known in the art.
  • Such immunoassays typically involve the measurement of complex formation between an immunogen and an antibody which specifically binds to the XPNPEP2
  • an antibody which specifically binds to XPNPEP2 provides a d etection signal at least 5-, 10-, or 20-fold higher than a detection signal provided with other proteins when used in an immunochemical assay.
  • antibodies which specifically bind to XPNPEP2 do not detect other proteins in immunochemical assays and can immunoprecipitate XPNPEP2 from solution.
  • XPNPEP2 can be used to immunize a mammal, such as a mouse, rat, rabbit, guinea pig, monkey, or human, to produce polyclonal antibodies. If desired, XPNPEP2 can be conjugated to a carrier protein, such as bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin. Depending on the host species, various adjuvants can be used to increase the immunological response.
  • a carrier protein such as bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin.
  • various adjuvants can be used to increase the immunological response.
  • Such adjuvants include, but are not limited to, Freund's adjuvant, mineral gels (e.g., aluminum hydroxide), and surface active substances (e.g., lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol).
  • mineral gels e.g., aluminum hydroxide
  • surface active substances e.g., lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol.
  • BCG Bacilli Calmette-Guerin
  • Corynebacterium parvum are especially useful.
  • Monoclonal antibodies which specifically bind to XPNPEP2 can be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These techniques include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBN- hybridoma technique [Roberge, (1995)]. In addition, techniques developed for the production of "chimeric antibodies", the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used.
  • Monoclonal and other antibodies also can be "humanized" to prevent a patient from mounting an immune r esponse a gainst t he a ntibody w hen i t i s u sed t herapeutically.
  • Such antibodies may be sufficiently similar in sequence to human antibodies to be used directly in therapy or may require alteration of a few key residues. Sequence differences between rodent antibodies and human sequences can b e m inimized b y replacing residues which differ from those in the human sequences by site directed mutagenesis of individual residues or by grating of entire complementarity determining regions.
  • Antibodies which specifically bind to XPNPEP2 can contain antigen binding sites which are either partially or fully humanized, as disclosed in U.S. 5,565,332.
  • single chain antibodies can be adapted using methods known in the art to produce single chain antibodies which specifically b ind t o XPNPEP2.
  • Antibodies with related specificity, but of distinct idiotypic composition can be generated by chain shuffling from random combinatorial immunoglobin libraries.
  • Single-chain antibodies also can be constructed using a DNA amplification method, such as PCR, using hybridoma cDNA as a template.
  • Single-chain antibodies can be mono- or bispecific, and can be bivalent or tetravalent. Construction of tetravalent, bispecific single-chain antibodies is taught.
  • a nucleotide sequence encoding a single-chain antibody can be constructed using manual or automated nucleotide synthesis, cloned into an expression construct using standard recombinant DNA methods, and introduced into a cell to express the coding sequence, as described below.
  • single-chain antibodies can be produced directly using, for example, filamentous phage technology.
  • Antibodies which specifically bind to XPNPEP2 also can be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents.
  • Other types of antibodies can be constructed and used therapeutically in methods of the invention.
  • chimeric antibodies can be constructed as disclosed in WO 93/03151.
  • Antibodies according to the invention can be purified by methods well known in the art. For example, antibodies can be affinity purified by passage over a column to which XPNPEP2 is bound. The bound antibodies can then be eluted from the column using a buffer with a high salt concentration.
  • Antisense oligonucleotides are nucleotide sequences which are complementary to a specific DNA or RNA sequence. Once introduced into a cell, the complementary nucleotides combine with natural sequences produced by the cell to form complexes and block either transcription or translation. Preferably, an antisense oligonucleotide is at least 11 nucleotides in length, but can be at least 12, 15, 20, 25, 30, 35, 40, 45, or 50 or more nucleotides long. Longer sequences also can be used. Antisense oligonucleotide molecules can be provided in a DNA construct and introduced into a cell as described above to decrease the level of XPNPEP2 gene products in the cell.
  • Antisense oligonucleotides can be deoxyribonucleotides, ribonucleotides, or a combi- nation of both. Oligonucleotides can be synthesized manually or by an automated synthesizer, by covalently linking the 5' end of one nucleotide with the 3' end of another nucleotide with non-phosphodiester internucleotide linkages such alkyl- phosphonates, phosphorothioates, phosphorodithioates, alkylphosphonothioates, alkylphosphonates, phosphoramidates, phosphate esters, carbamates, acetamidate, carboxymethyl esters, carbonates, and phosphate triesters.
  • Modifications of XPNPEP2 gene expression can be obtained by designing antisense oligonucleotides which will form duplexes to the control, 5', or regulatory regions of the XPNPEP2 gene. Oligonucleotides derived from the transcription initiation site, e.g., between 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 chaperons. Therapeutic advances using triplex DNA have been described in the literature [Nicholls, (1993)]. An antisense oligonucleotide also can be designed to block translation of mRNA by preventing the transcript from binding to ribosomes.
  • Antisense oligonucleotides which comprise, for example, 2, 3, 4, or 5 or more stretches of contiguous nucleotides which are precisely complementary to a
  • XPNPEP2 polynucleotide each separated by a stretch of contiguous nucleotides which are not complementary to adjacent XPNPEP2 nucleotides, can provide sufficient targeting specificity for XPNPEP2 mRNA.
  • each stretch of complementary contiguous nucleotides is at least 4, 5, 6, 7, or 8 or more nucleotides in length.
  • Non-complementary intervening sequences are preferably 1, 2, 3, or 4 nucleotides in length.
  • One skilled in the art can easily use the calculated melting point of an antisense-sense pair to determine the degree of mismatching which will be tolerated between a particular antisense oligonucleotide and a particular XPNPEP2 polynucleotide sequence.
  • Antisense oligonucleotides can be modified without affecting their ability to hybridize to a XPNPEP2 polynucleotide. These modifications can be internal or at one or both ends of the antisense molecule.
  • internucleoside phosphate linkages can be modified by adding cholesteryl or diamine moieties with varying numbers of carbon residues between the amino groups and terminal ribose.
  • modified antisense oligonucleotide 5' phosphate group are substituted, also can be employed in a modified antisense oligonucleotide.
  • modified oligonucleotides can be prepared by methods well known in the art.
  • Ribozymes are RNA molecules with catalytic activity [Uhlmann, (1987)]. Ribozymes can be used to inhibit gene function by cleaving an RNA sequence, as is known in the art. The mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage. Examples include engineered hammerhead motif ribozyme molecules that can specifically and efficiently catalyze endonucleolytic cleavage of specific nucleotide sequences.
  • the coding sequence of a XPNPEP2 polynucleotide can be used to generate ribozymes which will specifically bind to mRNA transcribed from a XPNPEP2 polynucleotide.
  • Methods of designing and constructing ribozymes which can cleave other RNA molecules in trans in a highly sequence specific manner have been developed and described in the art.
  • the cleavage activity of ribozymes can be targeted to specific RNAs by engineering a discrete "hybridization" region into the ribozyme.
  • the hybridization region contains a sequence complementary to the target RNA and thus specifically hybridizes with the target RNA.
  • Specific ribozyme cleavage sites within a XPNPEP2 RNA target can be identified by scanning the target molecule for ribozyme cleavage sites which include 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 RNA containing the cleavage site can be evaluated for secondary structural features which may render the target inoperable. Suitability of candidate XPNPEP2 RNA targets also can be evaluated by testing accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays.
  • the nucleotide sequences shown in SEQ JJD NO: 1 and its complement provide sources of suitable hybridization region sequences. Longer complementary sequences can be used to increase the affinity of the hybridization sequence for the target.
  • the hybridizing and cleavage regions of the ribozyme can be integrally related such that upon hybridizing to the target RNA through the complementary regions, the catalytic region of the ribozyme can cleave the target.
  • Ribozymes can be introduced into cells as part of a DNA construct. Mechanical methods, such as microinj ection, liposome-mediated transfection, electroporation, or calcium phosphate precipitation, can be used to introduce a ribozyme-containing DNA construct into cells in which it is desired to decrease XPNPEP2 expression. Alternatively, if it is desired that the cells stably retain the DNA construct, the construct can be supplied on a plasmid and maintained as a separate element or integrated into the genome of the cells, as is known in the art.
  • a ribozyme-encoding DNA construct can include transcriptional regulatory elements, such as a promoter element, an enhancer or UAS element, and a transcriptional terminator signal, for controlling transcription of ribozymes in the cells (U.S. 5,641,673). Ribozymes also can be engineered to provide an additional level of regulation, so that destruction of mRNA occurs only when both a ribozyme and a target gene are induced in the cells.
  • Regulators as used herein, refer to compounds that affect the activity of XPNPEP2 in vivo and/or in vitro. Regulators can be agonists and antagonists of XPNPEP2 polypeptide and can be compounds that exert their effect on the XPNPEP2 activity via the enzymatic activity, expression, post-translational modifications or by other means.
  • Agonists of XPNPEP2 are molecules which, when bound to XPNPEP2, increase or prolong the activity of XPNPEP2.
  • Agonists of XPNPEP2 include proteins, nucleic acids, carbohydrates, small molecules, or any other molecule which activate XPNPEP2.
  • Antagonists of XPNPEP2 are molecules which, when bound to
  • Antagonists include proteins, nucleic acids, carbohydrates, antibodies, small molecules, or any other molecule which decrease the activity of XPNPEP2.
  • modulate refers to a change in the activity of XPNPEP2 polypeptide. For example, modulation may cause an increase or a decrease in enzymatic activity, binding characteristics, or any other biological, functional, or immunological properties of XPNPEP2.
  • telomere binding refers 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.
  • the invention provides methods (also referred to herein as "screening assays") for identifying compounds which can be used for the treatment of diseases related to XPNPEP2.
  • the methods entail the identification of candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other molecules) which bind to XPNPEP2 and/or have a s timulatory o r i nhibitory e ffect o n t he b iological activity of XPNPEP2 or its expression and then determining which of these compounds have an effect on symptoms or diseases related to XPNPEP2 in an in vivo assay.
  • candidate or test compounds or agents e.g., peptides, peptidomimetics, small molecules or other molecules
  • Candidate or test compounds or agents which bind to XPNPEP2 and/or have a stimulatory or inhibitory effect on the activity or the expression of XPNPEP2 are identified either in assays that employ cells which express XPNPEP2 (cell-based assays) or in assays with isolated XPNPEP2 (cell-free assays).
  • the various assays can employ a variety of variants of XPNPEP2 (e.g., full-length XPNPEP2, a biologically active fragment of XPNPEP2, or a fusion protein which includes all or a portion of XPNPEP2).
  • XPNPEP2 can be derived from any suitable mammalian species (e.g., human XPNPEP2, rat XPNPEP2 or murine XPNPEP2).
  • the assay can be a binding assay entailing direct or indirect measurement of the binding of a test compound or a known XPNPEP2 ligand to XPNPEP2.
  • the assay can also be an activity assay entailing direct or indirect measurement of the activity of XPNPEP2.
  • the assay can also be an expression assay entailing direct or indirect measurement of the expression of XPNPEP2 mRNA or XPNPEP2 protein.
  • the various screening assays are combined with an in vivo assay entailing measuring the effect of the test compound on the symptoms of diseases related to XPNPEP2.
  • the present invention includes biochemical, cell free assays that allow the identification of inhibitors and agonists of proteases suitable as lead structures for pharmacological drug development.
  • Such assays involve contacting a form of XPNPEP2 (e.g., full-length XPNPEP2, a biologically active fragment of XPNPEP2, or a fusion protein comprising all or a portion of XPNPEP2) with a test compound and determining the ability of the test compound to act as an antagonist (preferably) or an agonist of the enzymatic activity of XPNPEP2.
  • XPNPEP2 e.g., full-length XPNPEP2, a biologically active fragment of XPNPEP2, or a fusion protein comprising all or a portion of XPNPEP2
  • XPNPEP2 molecules of the present invention can be measured using a variety of assays that measure XPNPEP2 activity.
  • XPNPEP2 enzyme activity can be assessed by a standard in vitro serine/metallo/... protease assay (see, for example, [U.S. 5 ,057,414]).
  • substrates suitable for in vitro assays such as SucAla-Ala-Pro-Phe-pNA, fluorescein mono-p-guanidinobenzoate hydrochloride, benzyloxycarbonyl-L-Arginyl-S-benzyl- ester, Nalpha-Benzoyl-L-arginine ethyl ester hydrochloride, and the like.
  • protease assay kits available from commercial sources, such as CalbiochemTM (San Diego, Calif.).
  • Solution in vitro assays can be used to identify a XPNPEP2 substrate or inhibitor.
  • Solid phase systems can also be used to identify a substrate or inhibitor of a XPNPEP2 polypeptide.
  • a XPNPEP2 polypeptide or XPNPEP2 fusion protein c an b e i mmobilized o nto t he s urface o f a r eceptor c hip o f a commercially available biosensor instrument (BIACORE, Biacore AB; Uppsala, Sweden). The use of this instrument is disclosed, for example, by [Karlsson, (1991), and Cunningham and Wells, (1993)].
  • a XPNPEP2 polypeptide or fusion protein is covalently attached, using amine or sulfhydryl chemistry, to dextran fibers that are attached to gold film within a flow cell.
  • a test sample is then passed through the cell. If a XPNPEP2 substrate or inhibitor is present in the sample, it will bind to the immobilized polypeptide or fusion protein, causing a change in the refractive index of the medium, which is detected as a change in surface plasmon resonance of the gold film.
  • This system allows the determination on- and off-rates, from which binding affinity can be calculated, and assessment of the stoichiometry of binding, as well as the kinetic effects of XPNPEP2 mutation.
  • This system can also be used to examine antibody- antigen interactions, and the interactions of other complement/anti-complement pairs.
  • the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of XPNPEP2.
  • assays can employ full-length XPNPEP2, a biologically active fragment of XPNPEP2, or a fusion protein which includes all or a portion of XPNPEP2.
  • the test compound can be obtained by any suitable means, e.g., from conventional compound libraries.
  • Determining the ability of the test compound to modulate the activity of XPNPEP2 can be accomplished, for example, by determining the ability of XPNPEP2 to bind to or interact with a target molecule.
  • the target molecule can be a molecule with which
  • the target molecule can be a component of a signal transduction pathway which facilitates transduction of an extracellular signal.
  • the target XPNPEP2 molecule can be, for example, a second intracellular protein which has catalytic activity or a protein which facilitates the association of downstream signaling molecules with XPNPEP2.
  • Determining the ability of XPNPEP2 to bind to or interact with a target molecule can be accomplished by one of the methods described above for determining direct binding. In one embodiment, determining the ability of a polypeptide of the invention to bind to or interact with a target molecule can be accomplished by determining the activity of the target molecule.
  • the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (e.g., intracellular Ca 2+ , diacylglycerol, IP 3 , etc.), detecting catalytic/enzymatic activity of the target on an appropriate substrate, detecting the induction of a reporter gene (e.g., a regulatory element that is responsive to a polypeptide of the invention operably linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a cellular response.
  • a reporter gene e.g., a regulatory element that is responsive to a polypeptide of the invention operably linked to a nucleic acid encoding a detectable marker, e.g., luciferase
  • XPNPEP2 or a XPNPEP2 target molecule
  • Examples of such vessels include microtitre plates, test tubes, and micro-centrifuge tubes.
  • a fusion protein can be provided which adds a domain that allows one or both of the proteins to be bound to a matrix.
  • GST glutathione-S-transferase
  • gluta- thione-S-transferase fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical; St. Louis, Mo.) or glutathione derivatized microtitre plates, which are then combined with the test compound or the test compound and either the non-adsorbed target protein or XPNPEP2, and the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH).
  • the b eads or microtitre plate wells are washed to remove any unbound components and complex formation is measured either directly or indirectly, for example, as described above.
  • the complexes can be dissociated from the matrix, and the level of binding or activity of XPNPEP2 can be determined using standard techniques.
  • XPNPEP2 or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin.
  • Biotinylated polypeptide of the invention or target molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals; Rockford, 111.), and immobilized in the wells of streptavidin-coated plates (Pierce Chemical).
  • biotinylation kit Pierce Chemicals; Rockford, 111.
  • streptavidin-coated plates Piereptavidin-coated plates
  • antibodies reactive with XPNPEP2 or target molecules but which do not interfere with binding of the polypeptide of the invention to its target molecule can be derivatized to the wells of the plate, and unbound target or polypeptide of the invention trapped in the wells by antibody conjugation.
  • Methods for detecting such complexes include immunodetection of complexes using antibodies reactive with XPNPEP2 or target molecule, as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with XPNPEP2 or target molecule.
  • 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.
  • 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 XPNPEP2, or fragments thereof, and washed.
  • Bound XPNPEP2 is then detected by methods well known in the art.
  • Purified XPNPEP2 can also be coated directly onto plates for use in the aforementioned drug screening techniques.
  • non- neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support.
  • the screening assay can also involve monitoring the expression of XPNPEP2.
  • regulators of expression of XPNPEP2 can be identified in a method in which a cell is contacted with a candidate compound and the expression of XPNPEP2 protein or mRNA in the cell is determined. The level of expression of XPNPEP2 protein or mRNA the presence of the candidate compound is compared to the level of expression of XPNPEP2 protein or mRNA in the absence of the candidate compound. The candidate compound can then be identified as a regulator of expression of XPNPEP2 based on this comparison.
  • the candidate compound when expression of XPNPEP2 protein or mRNA protein is greater (statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of XPNPEP2 protein or mRNA expression.
  • the candidate compound when expression of XPNPEP2 protein or mRNA is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of XPNPEP2 protein or mRNA expression.
  • the level of XPNPEP2 protein or mRNA expression in the cells can be determined by methods described below.
  • the test compound is preferably a small molecule which binds to and occupies the active site of XPNPEP2 polypeptide, thereby making the ligand binding site inaccessible to substrate such that normal biological activity is prevented.
  • small molecules include, but are not limited to, small peptides or peptide-like molecules.
  • Potential ligands which bind to a polypeptide of the invention include, but are not limited to, the natural ligands of known XPNPEP2 proteases and analogues or derivatives thereof.
  • either the test compound or the XPNPEP2 polypeptide can comprise a detectable label, such as a fluorescent, radioisotopic, chemiluminescent, or enzymatic label, such as horseradish peroxidase, alkaline phosphatase, or luciferase. Detection of a test compound which is bound to XPNPEP2 polypeptide can then be accomplished, for example, by direct counting of radioemmission, b y scintillation counting, or by determining conversion of an appropriate substrate to a detectable product. Alternatively, binding of a test compound to a XPNPEP2 polypeptide can be determined without labeling either of the interactants.
  • a detectable label such as a fluorescent, radioisotopic, chemiluminescent, or enzymatic label, such as horseradish peroxidase, alkaline phosphatase, or luciferase.
  • a microphysiometer can be used to detect binding of a test compound with a XPNPEP2 polypeptide.
  • a microphysiometer e.g., CytosensorTM
  • LAPS light-addressable p otentiometric s ensor
  • C hanges i n t his acidification rate can be used as an indicator of the interaction between a test compound and XPNPEP2 [Haseloff, (1988)].
  • BIA Bimolecular Interaction Analysis
  • a XPNPEP2-like polypeptide can be used as a "bait protein" in a two-hybrid assay or three-hybrid assay [Szabo, (1995); U.S. 5,283,317), to identify other proteins which bind to or interact with XPNPEP2 and modulate its activity.
  • the two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constructs.
  • polynucleotide encoding XPNPEP2 can be fused to a polynucleotide encoding the DNA binding domain of a known transcription factor (e.g., GAL-4).
  • a DNA sequence that encodes an unidentified protein (“prey” or “sample") can be fused to a polynucleotide that codes for the activation domain of the known transcription factor. If the "bait” and the "prey” proteins are able to interact in vivo to form an protein- dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity.
  • reporter gene e.g., LacZ
  • a reporter gene e.g., LacZ
  • Expression of the reporter gene can be detected, and cell colonies containing the functional transcription factor can be isolated and used to obtain the DNA sequence encoding the protein which interacts with XPNPEP2.
  • either the XPNPEP2 (or polynucleotide) or the test compound can be bound to a solid support.
  • Suitable solid supports include, but are not limited to, glass or plastic slides, tissue culture plates, microtiter wells, tubes, silicon chips, or particles such as beads (including, but not limited to, latex, polystyrene, or glass beads).
  • Any method known in the art can be used to attach XPNPEP2-like polypeptide (or polynucleotide) or test compound to a solid support, including use of covalent and non-covalent linkages, passive abso ⁇ tion, or pairs of binding moieties attached respectively to the polypeptide (or polynucleotide) or test compound and the solid support.
  • Test compounds are preferably bound to the solid support in an array, so that the location of individual test compounds can be tracked. Binding of a test compound to XPNPEP2 (or a polynucleotide encoding for XPNPEP2) can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and microcentrifuge tubes.
  • XPNPEP2 is a fusion protein comprising a domain that allows binding of XPNPEP2 to a solid support.
  • glutathione-S-transferase fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, Mo.) or glutathione derivatized microtiter plates, which are then combined with the test compound or the test compound and the non-adsorbed XPNPEP2; the mixture is then incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtiter plate wells are washed to remove any unbound components. Binding of the interactants can be determined either directly or indirectly, as described above. Alternatively, the complexes can be dissociated from the solid support before binding is determined.
  • XPNPEP2 or a polynucleotide encoding XPNPEP2
  • test compound can be immobilized utilizing conjugation of biotin and streptavidin.
  • Biotinylated XPNPEP2 (or a poly- nucleotide encoding biotinylated XPNPEP2) or test compounds can be prepared from biotin-NHS (N-hydroxysuccinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, 111.) and immobilized in the wells of streptavidin-coated plates (Pierce Chemical).
  • antibodies which specifically bind to XPNPEP2, polynucleotide, or a test compound, but which do not interfere with a desired binding site, such as the active site of XPNPEP2 can be derivatized to the wells of the plate. Unbound target or protein can be trapped in the wells by antibody conjugation.
  • Methods for detecting such complexes include immunodetection of complexes using antibodies which specifically bind to XPNPEP2 polypeptide or test compound, enzyme- linked assays which rely on detecting an activity of XPNPEP2 polypeptide, and SDS gel electrophoresis under non-reducing conditions.
  • Screening for test compounds which bind to a XPNPEP2 polypeptide or polynucleo- tide also can be carried out in an intact cell.
  • Any cell which comprises a XPNPEP2 polypeptide or polynucleotide can be used in a cell-based assay system.
  • a XPNPEP2 polynucleotide can be naturally occurring in the cell or can be introduced using techniques such as those described above. Binding of the test compound to XPNPEP2 or a polynucleotide encoding XPNPEP2 is determined as described above.
  • Test compounds can be tested for the ability to increase or decrease XPNPEP2 activity of a XPNPEP2 polypeptide.
  • the XPNPEP2 activity can be measured, for example, using methods described in the specific examples, below.
  • XPNPEP2 activity can be measured after contacting either a purified XPNPEP2 or an intact cell with a test compound.
  • a test compound which decreases XPNPEP2 activity by at least about 10, preferably about 50, more preferably about 75, 90, or 100% is identified as a potential agent for decreasing XPNPEP2 activity.
  • a test compound which increases XPNPEP2 activity by at least about 10, preferably about 50, more preferably about 75, 90, or 100% is identified as a potential agent for increasing XPNPEP2 activity.
  • test compounds which increase or decrease XPNPEP2 gene expression are identified.
  • the term "correlates with expression of a polynucleotide” indicates that the detection of the presence of nucleic acids, the same or r elated t o a n ucleic a cid s equence e ncoding X PNPEP2, b y n orthern a nalysis o r realtime PCR is indicative of the presence of nucleic acids encoding XPNPEP2 in a sample, and thereby correlates with expression of the transcript from the poly- nucleotide encoding XPNPEP2.
  • microarray 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.
  • a XPNPEP2 polynucleotide is contacted with a test compound, and the expression of an RNA or polypeptide product of XPNPEP2 polynucleotide is determined.
  • the level of expression of appropriate mRNA or polypeptide in the presence of the test compound is compared to the level of expression of mRNA or polypeptide in the absence of the test compound.
  • the test compound can then be identified as a regulator of expression based on this comparison.
  • test compound when expression of mRNA or polypeptide is greater in the presence of the test compound than in its absence, the test compound is identified as a stimulator or enhancer of the mRNA or polypeptide expression.
  • test compound when expression of the mRNA or polypeptide is less in the presence of the test compound than in its absence, the test compound is identified as an inhibitor of the mRNA or polypeptide expression.
  • the level of XPNPEP2 mRNA or polypeptide expression in the cells can be determined by methods well known in the art for detecting mRNA or polypeptide. Either qualitative or quantitative methods can be used.
  • the presence of polypeptide products of XPNPEP2 polynucleotide can be determined, for example, using a variety of techniques known in the art, including immunochemical methods such as radioimmunoassay, Western blotting, and immunohistochemistry.
  • polypeptide synthesis can be determined in vivo, in a cell culture, or in an in vitro translation system by detecting inco ⁇ oration of labelled amino acids into XPNPEP2.
  • Such screening can be carried out either in a cell-free assay system or in an intact cell.
  • Any cell which expresses XPNPEP2 polynucleotide can be used in a cell-based assay system.
  • the XPNPEP2 polynucleotide can be naturally occurring in the cell or can be introduced using techniques such as those described above. Either a primary culture or an established cell line can be used.
  • test compounds for use in the screening assays of the invention can be obtained from any suitable source, e.g., conventional compound libraries.
  • the test compounds can also be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound” library method; and synthetic library methods using affinity chromatography selection.
  • the biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds [Lam, (1997)]. Examples of methods for the synthesis of molecular libraries can be found in the art. Libraries of compounds may be presented in solution or on beads, bacteria, spores, plasmids or phage.
  • Computer modeling and searching technologies permit identification of compounds, or the improvement of already identified compounds, that can modulate XPNPEP2 expression or activity. Having identified such a compound or composition, the active sites or regions are identified. Such sites might typically be the enzymatic active site, regulator binding sites, or ligand binding sites.
  • the active site can be identified using methods known in the art including, for example, from the amino acid sequences of peptides, from the nucleotide sequences of nucleic acids, or from study of complexes of the relevant compound or composition with its natural ligand. In the latter case, chemical or X-ray crystallographic methods can b e used to find the active site by finding where on the factor the complexed ligand is found.
  • the three dimensional geometric structure of the active site is determined. This can be done by known methods, including X-ray crystallography, which can determine a complete molecular structure. On the other hand, solid or liquid phase NMR can be used to determine certain intramolecular distances. Any other experimental method of structure determination can be used to obtain partial or complete g eometric s nesttures. T he g eometric s nesttures mayb e m easured w ith a complexed ligand, natural or artificial, which may increase the accuracy of the active site structure determined.
  • the methods of computer based numerical modeling can be used to complete the structure or improve its accuracy.
  • Any recognized modeling method may be used, including parameterized models specific to particular biopolymers such as proteins or nucleic acids, molecular dynamics models based on computing molecular motions, statistical mechanics models based on thermal ensembles, or combined models.
  • standard molecular force fields representing the forces between constituent atoms and groups, are necessary, and can be selected from force fields known in physical chemistry.
  • the incomplete or less accurate experimental structures can serve as constraints on the complete and more accurate structures computed by these modeling methods.
  • candidate modulating compounds can be identified by searching databases containing compounds along with information on their molecular structure. Such a search seeks compounds having structures that match the determined active site structure and that interact with the groups defining the active site. Such a search can be manual, but is preferably computer assisted. These compounds found from this search are potential XPNPEP2 modulating compounds.
  • these methods can be used to identify improved modulating compounds from an already known modulating compound or ligand.
  • the composition of the known compound can be modified and the structural effects of modification can be determined using the experimental and computer modeling methods described above applied to the new composition.
  • the altered structure is then compared to the active site structure of the compound to determine if an improved fit or interaction results. In this manner systematic variations in composition, such as by varying side groups, can be quickly evaluated to obtain modified modulating compounds or ligands of improved specificity or activity.
  • XPNPEP2 is expressed in various human tissues.
  • CNS disorders include disorders of the central nervous system as well as disorders of the peripheral nervous system.
  • CNS disorders include, but are not limited to brain injuries, cerebrovascular diseases and their consequences, Parkinson's disease, corticobasal degeneration, motor neuron disease, dementia, including ALS, multiple sclerosis, traumatic brain injury, stroke, post-stroke, post-traumatic brain injury, and small-vessel cerebrovascular disease.
  • Dementias such as Alzheimer's disease, vascular dementia, dementia with Lewy bodies, frontotemporal dementia and Parkinsonism linked to chromosome 17, frontotemporal dementias, including Pick's disease, progressive nuclear palsy, corticobasal degeneration, Huntington's disease, thalamic degeneration, Creutzfeld- Jakob dementia, HIN dementia, schizophrenia with dementia, and Korsakoff s psychosis, within the meaning of the definition are also considered to be C ⁇ S disorders.
  • CNS disorders such as mild cognitive impairment, age-associated memory impairment, age-related cognitive decline, vascular cognitive impairment, attention deficit disorders, attention deficit hyperactivity disorders, and memory disturbances in children with learning disabilities are also considered to be CNS disorders.
  • Pain within the meaning of this definition, is also considered to be a CNS disorder. Pain can be associated with CNS disorders, such as multiple sclerosis, spinal cord injury, sciatica, failed back surgery syndrome, traumatic brain injury, epilepsy, Parkinson's disease, post-stroke, and vascular lesions in the brain and spinal cord (e.g., infarct, hemorrhage, vascular malformation).
  • CNS disorders such as multiple sclerosis, spinal cord injury, sciatica, failed back surgery syndrome, traumatic brain injury, epilepsy, Parkinson's disease, post-stroke, and vascular lesions in the brain and spinal cord (e.g., infarct, hemorrhage, vascular malformation).
  • Non-central neuropathic pain includes that associated with post mastectomy pain, phantom feeling, reflex sympathetic dystrophy (RSD), trigeminal neuralgiaradioculopathy, post-surgical pain,
  • H1N/AIDS related pain cancer pain
  • metabolic neuropathies e.g., diabetic neuropathy, vasculitic neuropathy secondary to connective tissue disease
  • paraneoplastic p olyneuropathy associated, for example, with carcinoma of lung, or leukemia, or lymphoma, or carcinoma of prostate, colon or stomach, trigeminal neuralgia, cranial neuralgias, and post-he ⁇ etic neuralgia. Pain associated with peripheral nerve damage, central pain (i.e. due to cerebral ischemia) and various chronic pain i.e., lumbago, back pain (low back pain), inflammatory and/or rheumatic pain.
  • Headache pain for example, migraine with aura, migraine without aura, and other migraine disorders
  • episodic and chronic tension-type headache tension-type like h eadache, c luster headache, and chronic paroxysmal hemicrania are also C ⁇ S disorders.
  • Visceral pain such as p ancreatits, intestinal cystitis, dysmenorrhea, irritable B owel syndrome, Crohn's disease, biliary colic, ureteral colic, myocardial infarction and pain syndromes of the pelvic cavity, e.g., vulvodynia, orchialgia, urethral syndrome and protatodynia are also C ⁇ S disorders.
  • the human XPNPEP2 is highly expressed in the following brain tissues: temporal lobe, neuroblastoma SK-N-MC cells, neuroblastoma SH-SY5Y cells.
  • the expression in brain tissues demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose nervous system diseases. Additionally the activity of the human XPNPEP2 can be modulated to treat nervous system diseases.
  • Heart failure is defined as a pathophysiological state in which an abnormality of cardiac function is responsible for the failure of the heart to pump blood at a rate commensurate with the requirement of the metabolizing tissue. It includes all forms of pumping failures such as high-output and low-output, acute and chronic, right- sided or left-sided, systolic or diastolic, independent of the underlying cause.
  • MI Myocardial infarction
  • Ischemic diseases are conditions in which the coronary flow is restricted resulting in a perfusion which is inadequate to meet the myocardial requirement for oxygen.
  • This group of diseases includes stable angina, unstable angina and asymptomatic ischemia.
  • Arrhythmias include all forms of atrial and ventricular tachyarrhythmias, atrial tachycardia, atrial flutter, atrial fibrillation, atrio-ventricular reentrant tachycardia, preexitation syndrome, ventricular tachycardia, ventricular flutter, ventricular fibrillation, as well as bradycardic forms of arrhythmias.
  • Hypertensive vascular diseases include primary as well as all kinds of secondary arterial hypertension, renal, endocrine, neurogenic, others.
  • the genes may be used as drug targets for the treatment of hypertension as well as for the prevention of all complications arising from cardiovascular diseases.
  • Peripheral vascular diseases are defined as vascular diseases in which arterial and/or venous flow is reduced resulting in an imbalance between blood supply and tissue oxygen demand. It includes chronic peripheral arterial occlusive disease (PAOD), acute arterial thrombosis and embolism, inflammatory vascular disorders, Raynaud's phenomenon and venous disorders.
  • PAOD peripheral arterial occlusive disease
  • acute arterial thrombosis and embolism inflammatory vascular disorders
  • Raynaud's phenomenon Raynaud's phenomenon
  • Atherosclerosis is a cardiovascular disease in which the vessel wall is remodeled, compromising the lumen of the vessel.
  • the atherosclerotic remodeling process involves accumulation of cells, both smooth muscle cells and monocyte/macrophage inflammatory cells, in the intima of the vessel wall. These cells take up lipid, likely from the circulation, to form a mature atherosclerotic lesion.
  • the formation of these lesions is a chronic process, occurring over decades of an adult human life, the majority of the morbidity associated with atherosclerosis occurs when a lesion ruptures, releasing thrombogenic debris that rapidly occludes the artery. When such an acute event occurs in the coronary artery, myocardial infarction can ensue, and in the worst case, can result in death.
  • the formation of the atherosclerotic lesion can be considered to occur in five overlapping stages such as migration, lipid accumulation, recruitment of inflammatory cells, proliferation of vascular smooth muscle cells, and extracellular matrix deposition.
  • stages such as migration, lipid accumulation, recruitment of inflammatory cells, proliferation of vascular smooth muscle cells, and extracellular matrix deposition.
  • Each of these processes can be shown to occur in man and in animal models of atherosclerosis, but the relative contribution of each to the pathology and clinical significance of the lesion is unclear.
  • Cardiovascular diseases include but are not limited to disorders of the heart and the vascular system like congestive heart failure, myocardial infarction, ischemic diseases of the heart, all kinds of atrial and ventricular arrhythmias, hypertensive vascular diseases, peripheral vascular diseases, and atherosclerosis.
  • the risk to develop atherosclerosis and coronary artery or carotid artery disease (and thus the risk of having a heart attack or stroke) increases with the total cholesterol level increasing. Nevertheless, extremely low cholesterol levels may not be healthy.
  • hyperlipidemia abnormally high levels of fats (cholesterol, triglycerides, or both) in the b lood, m ay b e c aused by family history of hyperlipidemia
  • obesity a high-fat diet
  • lack of exercise moderate to high alcohol consumption
  • cigarette smoking poorly controlled diabetes
  • an underactive thyroid gland hereditary hyperlipidemias
  • type I hyperlipoproteinemia familial hyperchylomicronemia
  • hyperlipoproteinemia familial hypercholesterolemia
  • type UI hyperlipoproteinemia type IN hyperlipoproteinemia
  • type N hyperlipoproteinemia type N hyperlipoproteinemia
  • hypo- lipoproteinemia lipidoses (caused by abnormalities in the enzymes that metabolize fats), Gaucher's disease, ⁇ iemann-Pick disease, Fabry's disease, Wolman's disease, cerebrotendinous xanthomatosis, sitosterolemia, Refsum's disease, or Tay-Sachs disease.
  • Kidney disorders may lead to hypertension or hypotension.
  • Examples for kidney problems p ossibly 1 eading to hypertension are renal artery stenosis, pyelonephritis, glomerulonephritis, kidney tumors, polycistic kidney disease, injury to the kidney, or radiation therapy affecting the kidney. Excessive urination may lead to hypotension.
  • the human XP ⁇ PEP2 is highly expressed in the following cardiovascular related tissues: heart atrium (left), HUVEC cells. Expression in the above mentioned tissues demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of cardio vascular diseases. Additionally the activity of the human XPNPEP2 can be modulated to treat cardiovascular diseases.
  • Gastrointestinal diseases comprise primary or secondary, acute or chronic diseases of the organs of the gastrointestinal tract which may be acquired or inherited, benign or malignant or metaplastic, and which may affect the organs of the gastrointestinal tract or the body as a whole. They comprise but are not limited to 1) disorders of the esophagus like achalasia, vigoruos achalasia, dysphagia, cricopharyngeal incoordination, pre-esophageal dysphagia, diffuse esophageal spasm, globus sensation, Barrett's metaplasia, gastroesophageal reflux, 2) disorders of the stomach and duodenum like functional dyspepsia, inflammation of the gastric mucosa, gastritis, stress gastritis, chronic erosive gastritis, atrophy of gastric glands, metaplasia of gastric tissues, gastric ulcers, duodenal ulcers, neoplasms of the stomach, 3) disorders of the pancreas like acute
  • Liver diseases comprise primary or secondary, acute or chronic diseases or injury of the liver which may be acquired or inherited, benign or malignant, and which may affect the liver or the body as a whole. They comprise but are not limited to disorders of the bilirubin metabolism, jaundice, syndroms of Gilbert's, Crigler-Najjar, Dubin- Johnson and Rotor; intrahepatic cholestasis, hepatomegaly, portal hypertension, ascites, Budd-Chiari syndrome, portal-systemic encephalopathy, fatty liver, steatosis, Reye's syndrome, liver diseases due to alcohol, alcoholic hepatitis or cirrhosis, fibrosis and cirrhosis, fibrosis and cirrhosis of the liver due to inborn errors of metabolism or exogenous substances, storage diseases, syndromes of Gaucher's,
  • Zellweger's Wilson's - disease, acute or chronic hepatitis, viral hepatitis and its variants, inflammatory conditions of the liver due to viruses, bacteria, fungi, protozoa, helminths; drug induced disorders of the liver, chronic liver diseases like primary sclerosing cholangitis, alpha ! -antitrypsin-deficiency, primary biliary cirrhosis, postoperative liver disorders like postoperative intrahepatic cholestasis, hepatic granulomas, vascular liver disorders associated with systemic disease, benign or malignant neoplasms of the liver, disturbance of liver metabolism in the new-born or prematurely born.
  • the human XPNPEP2 is highly expressed in the following tissues of the gastroenterological system: esophagus tumor, stomach, stomach tumor, small intestine, ileum, fetal liver.
  • the expression in the above mentioned tissues and in particular the differential expression between diseased tissue esophagus tumor and healthy tissue esophagus, between diseased tissue stomach tumor and healthy tissue stomach demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of gastroenterological disorders. Additionally the activity of the human XPNPEP2 can be modulated to treat gastroenterological disorders.
  • the skin serves several functions. It's an multi-layered organ system that builds an effective protective cover and regulates body temperature, senses painful and pleasant stimuli, keeps substances from entering the body, and provides a shield from the sun's harmful effects. Skin color, texture, and folds help mark people as individuals. Thus, skin disorders or diseases often have important consequences for physical and mental health. Skin disorders include, but are not limited to the conditions described in the following.
  • Itching is a sensation that instinctively demands scratching, which may be caused by a skin condition or a systemic diseas.
  • Superficial Skin Disorders affect the uppermost layer of the skin, the stratum corneum or the keratin layer, and it consists of many layers of flattened, dead cells and acts as a barrier to protect the underlying tissue from injury and infection.
  • Disorders of the superficial skin layers involve the stratum corneum and deeper layers of the epidermis. Examples of superficial skin disorders are provided in the following.
  • ichthyosis Dry skin often occurs in people past middle age, severe dry skin (ichthyosis) results from an inherited scaling disease, such as ichthyosis vulgaris or epidermolytic hyperkeratosis. Ichthyosis also results from nonhereditary disorders, such as leprosy, underactive thyroid, lymphoma, AIDS, and sarcoidosis.
  • Keratosis pilaris is a common disorder in which dead cells shed from the upper layer of skin and form plugs that fill the openings of hair follicles.
  • a callus is an area on the stratum corneum or keratin layer, that becomes abnormally thick in response to repeated rubbing.
  • a corn is a pea-sized, thickened area of keratin that occurs on the feet.
  • Psoriasis is a chronic, recurring disease recognizable by silvery scaling bumps and various-sized plaques (raised patches). An abnormally high rate of growth and turnover of skin cells causes the scaling. Pityriasis rosea is a mild disease that causes scaly, rose-colored, inflamed skin. Pityriasis rosea is possibly caused by an infectious agent, although none has b een identified.
  • Lichen planus a recurring itchy disease, starts as a rash of small discrete bumps that then combine and become rough, scaly plaques (raised patches).
  • Dermatitis is an inflammation of the upper layers of the skin, causing blisters, redness, swelling, oozing, scabbing, scaling, and usually itching.
  • dermatitis are contact dermatitis, or chronic dermatitis of the hands and feet, e.g. Pompholyx.
  • dermatitic disorders are atopic dermatitis, seborrheic dermatitis, nummular dermatitis, generalized exfoliative dermatitis, stasis dermatitis, or localized scratch dermatitis (lichen simplex chronicus, neurodermatitis).
  • Drug rashes are side effects of medications, mainly allergic reactions to medications.
  • Toxic epidermal necrolysis is a life-threatening skin disease in which the top layer of the skin peels off in sheets. This condition can be caused by a reaction to a drug, or by some other serious disease.
  • Erythema multiforme often caused by he ⁇ es simplex is a disorder characterized by patches of red, raised skin that often look like targets and usually are distributed symmetrically over the body.
  • Erythema nodosum is an inflammatory disorder that produces tender red bumps (nodules) under the skin, most often over the shins but occasionally on the arms and other areas.
  • Granuloma annulare is a chronic skin condition of unknown cause in which small, firm, raised bumps form a ring with normal or slightly sunken skin in the center.
  • Some skin disorders are characterized as blistering diseases.
  • Pemphigus is an uncommon, sometimes fatal, disease in which blisters (bullae) of varying sizes break out on the skin, the lining of the mouth, and other mucous membranes.
  • Bullous pemphigoid is an autoimmune disease that causes blistering.
  • Dermatitis he ⁇ etiformis is an autoimmune disease in which clusters of intensely itchy, small blisters and hive-like swellings break out and persist.
  • proteins in wheat, rye, barley, and oat products activate the immune system, which attacks parts of the skin and somehow causes the rash and itching.
  • Sweating disorders also belong to skin disorders.
  • Prickly heat is an itchy skin rash caused by trapped sweat.
  • Excessive sweating may affect the entire surface of the skin, but often it's limited to the palms, soles, armpits, or groin.
  • the affected area is often pink or bluish white, and in severe cases the skin may be cracked, scaly, and soft, especially on the feet.
  • Skin disorders can affect the sebaceous glands.
  • Acne is a common skin condition in which the skin pores become clogged, leading to pimples and inflamed, infected abscesses (collections of pus). Acne tends to develop in teenagers.
  • Acne is further subdivided in superficial acne or deep acne.
  • Rosacea is a persistent skin disorder that produces redness, tiny pimples, and broken blood vessels, usually on the central area of the face.
  • Perioral dermatitis is a red, often bumpy rash around the mouth and on the chin.
  • a sebaceous cyst (keratinous cyst) is a slow-growing bump containing dead skin, skin excretions, and other skin particles. These cysts may be small and can appear anywhere.
  • Hair disorders also are skin disorders. Hair disorders include excessive hairiness, baldness, and ingrown beard hairs.
  • the skin can be infected by bacteria.
  • Bacterial skin infections can range in seriousness from minor acne to a life-threatening condition, such as staphylococcal scalded skin syndrome.
  • the most common bacterial skin infections are caused b y Staphylococcus and Streptococcus.
  • Risk factors for skin infections are for example diabetes, ADDS or skin leasons.
  • Impetigo is a skin infection, caused by Staphylococcus or Streptococcus, leading to the formation of small pus-filled blisters (pustules).
  • Folliculitis is an inflammation of the hair follicles caused by infection with Staphylococcus. The infection damages the hairs, which can be easily pulled out.
  • Boils are large, tender, swollen, raised areas caused by staphylococcal infection around hair follicles.
  • Carbuncles are clusters of boils that result in extensive sloughing of skin and scar formation. Carbuncles develop and heal more slowly than single boils and may lead to fever and fatigue.
  • Erysipelas is a skin infection caused by Streptococcus. A shiny, red, slightly swollen, tender rash develops, often with small blisters. Lymph nodes around the infected area may become enlarged and painful.
  • Cellulitis is a spreading infection in, and sometimes beneath, the deep layers of the skin. Cellulitis most often results from a streptococcal infection or a staphylococcal infection. However, many other bacteria can also cause cellulitis.
  • Paronychia is an infection around the edge of a fingernail or toenail. Paronychia can be caused by many different bacteria, including Pseudomonas and Proteus, and by fungi, such as Candida.
  • Staphylococcal scalded skin syndrome is a widespread skin infection that can lead to toxic shock syndrome, in which the skin peels off as though burned. Certain types of staphylococci produce a toxic substance that causes the top layer of skin (epidermis) to split from the rest of the skin.
  • Erythrasma is an infection of the top layers of the skin by the bacterium Corynebacterium minutissimum. Skin infections are often caused by fungi. Fungi that infect the skin (dermatophytes) live only in the dead, topmost layer (stratum corneum) and don't penetrate deeper. Some fungal infections cause no symptoms or produce only a small amount of irritation, scaling, and redness. Other fungal infections cause itching, swelling, blisters, and severe scaling.
  • Ringworm is a fungal skin infection caused by several different fungi and generally classified by its location on the body.
  • Athlete's foot foot ringworm, caused by either Trichophyton or
  • Epidermophyton can be caused by a variety of fungi and yeasts
  • scalp ringworm caused by Trichophyton or Microsporum
  • nail ringworm and body ringworm (caused by Trichophyton).
  • Candidiasis is an infection by the yeast Candida.
  • Candida usually infects the skin and mucous membranes, such as the lining of the mouth and vagina. Rarely, it invades deeper tissues as well as the blood, causing life- threatening systemic candidiasis.
  • the following types of Candida infections can be distinguished: Infections in skinfolds (intertriginous infections), vaginal and penile Candida infections (vulvovaginitis), thrush, Perleche (candida infection at the corners of the mouth), candidal paronychia (candida growing in the nail beds, produces painful swelling and pus).
  • Tinea versicolor is a fungal infection that causes white to light brown patches on the skin.
  • the skin can also be affected by parasites, mainly tiny insects or worms.
  • Scabies is a mite infestation that produces tiny reddish pimples and severe itching. Scabies is caused by the itch mite Sarcoptes scabiei. Lice infestation (pediculosis) causes intense itching and can affect almost any area of the skin. Head lice and pubic lice are two different species.
  • Creeping eruption (cutaneous larva migrans) is a hookworm infection transmitted from warm, moist soil to exposed skin. The infection is caused by a hookworm that normally inhabits dogs and cats.
  • Warts are caused by the papillomavirus
  • cold sores are caused by the he ⁇ es simplex virus.
  • Another important group of viruses that infect the skin belongs to the poxvirus family. C hickenpox r emains a common childhood infection.
  • a poxvirus also causes molluscum contagiosum, which is an infection of the skin by a poxvirus that causes skin-colored, smooth, waxy bumps.
  • UVB ultraviolet B
  • Drugs can cause skin photosensitivity reactions which can occur after only a few minutes of sun exposure. These reactions include redness, peeling, hives, blisters, and thickened, scaling patches (photosensitivity).
  • Pigment Disorders Some skin disorders are characterized as Pigment Disorders.
  • Nitiligo is a condition in which a loss of melanocytes results in smooth, whitish patches of skin, which may occur after unusual physical trauma and tends to occur with certain other diseases, including Addison's disease, diabetes, pernicious anemia, and thyroid disease.
  • Tinea versicolor is a fungal infection of the skin that sometimes results in hype ⁇ igmentation.
  • Melasma appears on the face (usually the forehead, cheeks, temples, and jaws) as a roughly symmetric group of dark brown patches of pigmentation that are often clearly delineated.
  • Skin growths which are abnormal accumulations of different types of cells, may be present at birth or develop later. ⁇ oncancerous (benign) growth and cancerous
  • Moles are small, usually dark, skin growths that develop from pigment- producing cells in the skin (melanocytes). Most moles are harmless. However, noncancerous moles can develop into malignant melanoma.
  • Skin tags are soft, small, flesh-colored or slightly darker skin flaps that appear mostly on the neck, in the armpits, or in the groin.
  • Lipomas are soft deposits of fatty material that grow under the skin, causing round or oval lumps.
  • Angiomas are collections of abnormally dense blood or lymph vessels that are usually located in and below the skin and that cause red or pu ⁇ le discolorations. Examples of angiomas are port-wine stains, strawberry marks, cavernous hemangiomas, spider angiomas, and lymphangiomas.
  • Pyogenic granulomas are scarlet, brown, or blue-black slightly raised areas caused by increased growth of capillaries (the smallest blood vessels) and swelling of the surrounding tissue.
  • Seborrheic keratoses (sometimes called seborrheic warts) are flesh-colored, brown, or black growths that can appear anywhere on the skin.
  • Dermatofibromas are small, red-to-brown bumps (nodules) that result from an accumulation of fibroblasts, the cells that populate the soft tissue under the skin.
  • Keratoacanthomas are round, firm, usually flesh-colored growths that have an unusual central crater containing a pasty material.
  • Keloids are smooth, shiny, slightly pink, often dome-shaped, proliferative growths of fibrous tissue that form over areas of injury or over surgical wounds.
  • Skin cancer is the most common form of cancer, but most types of skin cancers are curable.
  • Basal cell carcinoma is a cancer that originates in the lowest layer of the epidermis.
  • Squamous cell carcinoma is cancer that originates in the middle layer of the epidermis.
  • Bowen's disease is a form of squamous cell carcinoma that's confined to the epidermis and hasn't yet invaded the underlying dermis.
  • Melanoma is a cancer that originates in the pigment-producing cells of the skin (melanocytes).
  • Kaposi's sarcoma is a cancer that originates in the blood vessels, usually of the skin.
  • Paget's disease i s a r are type o f skin c ancer that 1 ooks 1 ike an inflamed, reddened patch of skin (dermatitis); it originates in glands in or under the skin.
  • the human XPNPEP2 is highly expressed in the following dermatological tissues: skin.
  • the expression in the above mentioned tissues demonstrates that the human
  • XPNPEP2 or mRNA can be utilized to diagnose of dermatological diseases. Additionally the activity of t he h uman X PNPEP2 c an b e m odulated to t reat t hose diseases.
  • Cancer disorders within the scope of this definition comprise any disease of an organ or tissue in mammals characterized by poorly controlled or uncontrolled multiplication of normal or abnormal cells in that tissue and its effect on the body as a whole.
  • Cancer diseases within the scope of the definition comprise benign neoplasms, dysplasias, hype ⁇ lasias as well as neoplasms showing metastatic growth or any other transformations like e.g. leukoplakias which often precede a breakout of cancer.
  • C ells and tissues are c ancerous when they grow more rapidly than normal cells, displacing or spreading into the surrounding healthy tissue or any other tissues of the body described as metastatic growth, assume abnormal shapes and sizes, show changes in their nucleocytoplasmatic ratio, nuclear polychromasia, and finally may cease.
  • Cancerous cells and tissues may affect the body as a whole when causing paraneoplastic syndromes or if cancer occurs within a vital organ or tissue, normal function will be impaired or halted, with possible fatal results.
  • the ultimate involvement of a vital organ by cancer, either primary or metastatic, may lead to the death of the mammal affected. Cancer tends to spread, and the extent of its spread is usually related to an individual's chances of surviving the disease.
  • Cancers are generally said to be in one of three stages of growth: early, or localized, when a tumor is still confined to the tissue of origin, or primary site; direct extension, where cancer cells from the tumour have invaded adjacent tissue or have spread only to regional lymph nodes; or metastasis, in which cancer cells have migrated to distant parts of the body from the primary site, via the blood or lymph systems, and have established secondary sites of infection.
  • Cancer is said to be malignant because of its tendency to cause death if not treated. Benign tumors usually do not c ause death, although they may if they interfere with a normal body function by virtue of their location, size, or paraneoplastic side effects. Hence benign tumors fall under the definition of cancer within the scope of this definition as well.
  • cancer cells divide at a higher rate than do normal cells, but the distinction between the growth of cancerous and normal tissues is not so much the rapidity of cell division in the former as it is the partial or complete loss of growth restraint in cancer cells and their failure to differentiate into a useful, limited tissue of the type that characterizes the functional equilibrium of growth of normal tissue.
  • Cancer tissues may express certain molecular receptors and probably are influenced by the host's susceptibility and immunity and it is known that certain cancers of the breast and prostate, for example, are considered dependent on specific hormones for their existence.
  • cancer under the scope of the definition is not limited to simple benign neoplasia but comprises any other benign and malign neoplasia like 1) Carcinoma, 2) Sarcoma, 3) Carcinosarcoma, 4) Cancers of the blood-forming tissues, 5) tumors of nerve tissues including the brain, 6) cancer of skin cells.
  • Cancer according to 1) occurs in epithelial tissues, which cover the outer body (the skin) and line mucous membranes and the inner cavitary structures of organs e.g. such as the breast, lung, the respiratory and gastrointestinal tracts, the endocrine glands, and the genitourinary system.
  • Ductal or glandular elements may persist in epithelial tumors, as in adenocarcinomas like e.g. thyroid adenocarcmoma, gastric adenocarcmoma, uterine adenocarcmoma.
  • Cancers of the pavement-cell epithelium of the skin and of certain mucous membranes, such as e .g. cancers of the tongue, lip, larynx, urinary bladder, uterine cervix, or penis, may be termed epidermoid or squamous-cell carcinomas of the respective tissues and are in the scope of the definition of cancer as well.
  • Cancer according to 2) develops in connective tissues, including fibrous tissues, adipose (fat) tissues, muscle, blood vessels, bone, and cartilage like e.g. osteogenic sarcoma; liposarcoma, fibrosarcoma, synovial sarcoma.
  • Cancer according to 3) is cancer that develops in both epithelial and connective tissue.
  • Cancer disease within the scope of this definition may be primary or secondary, whereby primary indicates that the cancer originated in the tissue where it is found rather than was established as a secondary site through metastasis from another lesion.
  • Cancers and tumor diseases within the scope of this definition may be benign or malign and may affect all anatomical structures of the body of a mammal.
  • the bone marrow and bone marrow derived cells comprise cancers and tumor diseases of I) the bone marrow and bone marrow derived cells (leukemias), IT) the endocrine and exocrine glands like e.g. thyroid, parathyroid, pituitary, adrenal glands, salivary glands, pancreas ILT) the breast, like e.g.
  • malignant osteogenic sarcoma benign osteoma, cartilage tumors; like malignant chondrosarcoma or benign chondroma; bone marrow tumors like malignant myeloma or b enign e osinophilic granuloma, as well as metastatic tumors from bone tissues at other locations of the body;
  • XIN) the lymphatic tissue like lymphomas and other tumors of lymphoid origin, XV) the skin, XVI) cancers and tumor diseases of all anatomical structures belonging to the respiration and respiratory systems including
  • the human XPNPEP2 is highly expressed in the following cancer tissues: esophagus tumor, stomach tumor, ovary tumor, breast tumor, kidney tumor.
  • the expression in the above mentioned tissues and in particular the differential expression between diseased tissue esophagus tumor and healthy tissue esophagus, between diseased tissue stomach tumor and healthy tissue stomach, between diseased tissue ovary tumor and healthy tissue ovary, between diseased tissue breast tumor and healthy tissue breast, between diseased tissue kidney tumor and healthy tissue kidney demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of cancer. Additionally the activity of the human XPNPEP2 can be modulated to treat cancer.
  • Asthma is thought to arise as a result of interactions between multiple genetic and environmental factors and is characterized by three major features: 1) intermittent and reversible airway obstruction caused by bronchoconstriction, increased mucus production, and thickening of the walls of the airways that leads to a narrowing of the airways, 2) airway hyperresponsiveness, and 3) airway inflammation.
  • Certain cells are critical to the inflammatory reaction of asthma and they include T cells and antigen presenting cells, B cells that produce IgE, and mast cells, basophils, eosinophils, and other cells that bind IgE. These effector cells accumulate at the site of allergic reaction in the airways and release toxic products that contribute to the acute pathology and eventually to tissue destruction related to the disorder.
  • Other resident c ells such as smooth muscle cells, lung epithelial cells, mucus-producing cells, and nerve cells may also be abnormal in individuals with asthma and may contribute to its pathology. While the airway obstruction of asthma, presenting clinically as an intermittent wheeze and shortness of breath, is generally the most pressing symptom of the disease requiring immediate treatment, the inflammation and tissue destruction associated with the disease can lead to irreversible changes that eventually make asthma a chronic and disabling disorder requiring long-term management.
  • COPD chronic obstructive pulmonary (or airways) disease
  • COPD chronic obstructive pulmonary (or airways) disease
  • Emphysema is characterised by destruction of alveolar walls leading to abnormal enlargement of the air spaces of the lung.
  • Chronic bronchitis is defined clinically as the presence of chronic productive cough for three months in each of two successive years.
  • airflow obstruction is usually progressive and i s only partially reversible. By far the most important risk factor for development of COPD is cigarette smoking, although the disease does also occur in non-smokers.
  • the human XPNPEP2 is highly expressed in the following tissues of the respiratory system: fetal lung, lung right upper lobe, trachea.
  • the expression in the above mentioned tissues demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of respiratory diseases. Additionally the activity of the human XPNPEP2 can be modulated to treat those diseases.
  • Genitourinary disorders comprise benign and malign disorders of the organs constituting the genitourinary system of female and male, renal diseases like acute or chronic renal failure, immunologically mediated renal diseases like renal transplant rejection, lupus nephritis, immune complex renal diseases, glomerulopathies, nephritis, toxic nephropathy, obstructive uropathies like benign prostatic hype ⁇ lasia (BPH), neurogenic bladder syndrome, urinary incontinence like urge-, stress-, or overflow incontinence, pelvic pain, and erectile dysfunction.
  • the human XPNPEP2 is highly expressed in the following urological tissues: fetal kidney, kidney. The expression in the above mentioned tissues demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of urological disorders. Additionally the activity of the human XPNPEP2 can be modulated to treat urological disorders.
  • Metabolic diseases are defined as conditions which result from an abnormality in any of the chemical or biochemical transformations and their regulating systems essential to producing energy, to regenerating cellular constituents, to eliminating unneeded products arising from these processes, and to regulate and maintain homeostasis in a mammal regardless of whether acquired or the result of a genetic transformation.
  • a single defective trans- formation or disturbance of its regulation may produce consequences that are narrow, involving a single body function, or broad, affecting many organs, organ-systems or the body as a whole.
  • Diseases resulting from abnormalities related to the fine and coarse mechanisms that affect each individual transformation, its rate and direction or the availability of substrates like amino acids, fatty acids, carbohydrates, minerals, cofactors, hormones, regardless whether they are inborn or acquired, are well within the scope of the definition of a metabolic disease according to this application.
  • Metabolic diseases often are caused by single defects in particular biochemical pathways, defects that are due to the deficient activity of individual enzymes or molecular receptors leading to the regulation of such enzymes. Hence in a broader sense disturbances of the underlying genes, their products and their regulation lie well within the scope of this definition of a metabolic disease.
  • metabolic diseases may affect 1) biochemical processes and tissues ubiquitous all over the body, 2) the bone, 3) the nervous system, 4) the endocrine system, 5) the muscle including the heart, 6) the skin and nervous tissue, 7) the urogenital system, 8) the homeostasis o f body systems like water and electrolytes.
  • metabolic diseases according to 1) comprise obesity, amyloidosis, disturbances of the amino acid metabolism like branched chain disease, hyperaminoacidemia, hyperaminoaciduria, disturbances of the metabolism of urea, hyperammonemia, mucopolysaccliaridoses e.g.
  • Maroteaux-Lamy syndrom storage diseases like glycogen storage diseases and lipid storage diseases, glycogenosis diseases like Cori's disease, malabso ⁇ tion diseases like intestinal carbohydrate malabso ⁇ tion, oligosaccharidase deficiency like maltase-, lactase-, sucrase- insufficiency, disorders of the metabolism of fructose, disorders of the metabolism of galactose, galactosaemia, disturbances of carbohydrate utilization like diabetes, hypoglycemia, disturbances of pyruvate metabolism, hypolipidemia, hypolipo- proteinemia, hyperlipidemia, hyperlipoproteinemia, carnitine or carnitine acyl- transferase deficiency, disturbances of the po ⁇ hyrin metabolism, po ⁇ hyrias, disturbances of the purine metabolism, lysosomal diseases, metabolic diseases of nerves and nervous systems like gangliosidoses, sphingolipidoses, sulfatidose
  • metabolic diseases according to 2) comprise osteoporosis, osteomalacia like osteoporosis, osteopenia, osteogenesis imperfecta, osteopefrosis, osteonecrosis, Paget's disease of bone, hypophosphatemia.
  • metabolic diseases according to 3) comprise cerebellar dysfunction, disturbances of brain metabolism like dementia, Alzheimer's disease, Huntington's chorea, Parkinson's disease, Pick's disease, toxic encephalopathy, demyelinating neuropathies like inflammatory neuropathy, Guillain-Barre syndrome.
  • metabolic diseases comprise primary and secondary metabolic disorders associated with hormonal defects like any disorder stemming from either an hyperfunction or hypofunction of some hormone-secreting endocrine gland and any combination thereof. They comprise Sipple's syndrome, pituitary gland dysfunction and its effects on other endocrine glands, such as the thyroid, adrenals, ovaries, and testes, acromegaly, hyper- and hypothyroidism, euthyroid goiter, euthyroid sick syndrome, thyroiditis, and thyroid cancer, over- or unde ⁇ roduction of the adrenal steroid hormones, adrenogenital syndrome, Cushing's syndrome, Addison's disease of the adrenal cortex, Addison's pernicious anemia, primary and secondary aldosteronism, diabetes insipidus, carcinoid syndrome, disturbances caused by the dysfunction of the parathyroid glands, pancreatic islet cell dysfunction, diabetes, disturbances of the endocrine system of the female like estrogen defic
  • metabolic diseases comprise muscle weakness, myotonia, Duchenne's and other muscular dystrophies, dystrophia myotonica of Steinert, mitochondrial myopathies like disturbances of the catabolic metabolism in the muscle, carbohydrate and lipid storage myopathies, glycogenoses, myoglobinuria, malignant hyperthermia, polymyalgia rheumatica, dermatomyositis, primary myocardial disease, cardiomyopathy.
  • metabolic diseases according to 5 comprise muscle weakness, myotonia, Duchenne's and other muscular dystrophies, dystrophia myotonica of Steinert, mitochondrial myopathies like disturbances of the catabolic metabolism in the muscle, carbohydrate and lipid storage myopathies, glycogenoses, myoglobinuria, malignant hyperthermia, polymyalgia rheumatica, dermatomyositis, primary myocardial disease, cardiomyopathy.
  • t o, m etabolic diseases according to 6) comprise disorders of the ectoderm, neurofibromatosis, scleroderma and polyarteritis, Louis-Bar syndrome, von Hippel- Lindau disease, Sturge- Weber syndrome, tuberous sclerosis, amyloidosis, po ⁇ hyria.
  • metabolic diseases according to 7) comprise sexual dysfunction of the male and female.
  • metabolic diseases according to 8 comprise confused states and seizures due to inappropriate secretion of antidiuretic hormone from the pituitary gland, Liddle's syndrome, Bartter's syndrome, Fanconi's syndrome, renal electrolyte wasting, diabetes insipidus.
  • the human XPNPEP2 is highly expressed in the following metabolic disease related tissues: liver liver cirrhosis and adipose.
  • liver liver cirrhosis and adipose The expression in the above mentioned tissues and in particular the differential expression between diseased tissue liver liver cirrhosis and healthy tissue liver demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of metabolic diseases. Additionally the activity of the human XPNPEP2 can be modulated to treat metabolic diseases.
  • the present invention provides for both prophylactic and therapeutic methods for cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders.
  • the regulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of XPNPEP2.
  • An agent that modulates activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring cognate ligand of the polypeptide, a peptide, a peptidomimetic, or any small molecule.
  • the agent stimulates one or more of the biological activities of XPNPEP2.
  • stimulatory agents include the active XPNPEP2 and nucleic acid molecules encoding a portion of XPNPEP2.
  • agent i nhibits o ne o r m ore o f t he b iological a ctivities o f XPNPEP2.
  • inhibitory agents include antisense nucleic acid molecules and antibodies.
  • the present invention provides methods of treating an individual afflicted with a disease or disorder characterized by unwanted expression or activity of XPNPEP2 or a protein in the XPNPEP2 signaling pathway.
  • the method involves administering an agent like any agent identified or being identifiable by a screening assay as described herein, or combination of such agents that modulate say upregulate or downregulate the expression or activity of XPNPEP2 or of any protein in the XPNPEP2 signaling pathway.
  • the method involves administering a regulator of
  • XPNPEP2 as therapy to compensate for reduced or undesirably low expression or activity of XPNPEP2 or a protein in the XPNPEP2 signaling pathway.
  • Stimulation of activity or expression of XPNPEP2 is desirable in situations in which enzymatic activity or expression is abnormally low and in which increased activity is likely to have a beneficial effect. Conversely, inhibition of enzymatic activity or expression of XPNPEP2 is desirable in situations in which activity or expression of XPNPEP2 is abnormally high and in which decreasing its activity is likely to have a beneficial effect.
  • This invention is further illustrated by the following examples which should not be construed as limiting. The contents of all references, patents and published patent applications cited throughout this application are hereby inco ⁇ orated by reference.
  • This invention further pertains to novel agents identified by the above-described screening assays and uses thereof for treatments as described herein.
  • nucleic acid molecules also referred to herein as antibodies
  • active compounds of the invention can be inco ⁇ orated into pharmaceutical compositions suitable for administration.
  • Such compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and abso ⁇ tion delaying agents, and the like, compatible with pharmaceutical administration.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be inco ⁇ orated into the compositions.
  • the invention includes pharmaceutical compositions comprising a regulator of XPNPEP2 expression or activity (and/or a regulator of the activity or expression of a protein in the XPNPEP2 signaling pathway) as well as methods for preparing such compositions by combining one or more such regulators and a pharmaceutically acceptable carrier.
  • pharmaceutical compositions comprising a regulator identified using the screening assays of the invention packaged with instructions for use.
  • the instructions would specify use of the pharmaceutical composition for treatment of hematological and cardiovascular diseases, disorders of the peripheral and central nervous system, COPD, asthma, genito-urological disorders and inflammation diseases.
  • regulators that are agonists of XPNPEP2 activity or increase XPNPEP2 expression the instructions would specify use of the pharmaceutical composition for treatment of hematological and cardiovascular diseases, disorders-of the peripheral and central nervous system,
  • An inhibitor of XPNPEP2 may be produced using methods which are generally known in the art.
  • purified XPNPEP2 may be used to produce antibodies or to screen libraries of pharmaceutical agents to identify those which specifically bind XPNPEP2.
  • Antibodies to XPNPEP2 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, single chain antibodies, Fab fragments, and fragments produced by a Fab expression library. N eutralizing antibodies like those which inhibit dimer formation are especially preferred for therapeutic use.
  • the polynucleotides encoding XPNPEP2, or any fragment or complement thereof may be used for therapeutic pu ⁇ oses.
  • the complement of the polynucleotide encoding XPNPEP2 may be used in situations in which it would be desirable to block the transcription of the mRNA.
  • cells may be transformed with sequences complementary to polynucleotides encoding XPNPEP2.
  • complementary molecules or fragments may be used to modulate XPNPEP2 activity, or to achieve regulation of gene function.
  • sense or antisense oligonucleotides or larger fragments can be designed from various locations along the coding or control regions of sequences encoding XPNPEP2.
  • Expression vectors derived from retroviruses, adenoviruses, or he ⁇ es 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 s equence c omplementary t o t he p olynucleotides o f t he g ene e ncoding XPNPEP2. These techniques are described, for example, in [Scott and Smith (1990)].
  • 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, c ats, c ows, horses, rabbits, monkeys, and most preferably, humans.
  • An additional embodiment of the invention relates to the administration of a pharmaceutical composition containing XPNPEP2 in conjunction with a pharmaceutically acceptable carrier, for any of the therapeutic effects discussed above.
  • Such pharmaceutical compositions may consist of XPNPEP2, antibodies to XPNPEP2, and mimetics, agonists, antagonists, or inhibitors of XPNPEP2.
  • 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.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic s olvents; antibacterial a gents s uch a s b enzyl a lcohol o r m ethyl p arabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • suitable carriers include physiological saline, bacterio- static water, Cremophor EMTM (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, a pharmaceutically acceptable polyol like glycerol, propylene glycol, liquid polyetheylene glycol, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition.
  • Prolonged abso ⁇ tion of the injectable compositions can be brought about by including in the composition an agent which delays abso ⁇ tion, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by inco ⁇ orating the active compound (e.g., a polypeptide or antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by inco ⁇ orating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the pu ⁇ ose of oral therapeutic administration, the active compound can be inco ⁇ orated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed.
  • compositions can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any o f the following i ngredients, or e ompounds o f a s imilar n ature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • the compounds are delivered in the form of an aerosol spray from a pressurized container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the a rt, a nd i nclude, f or e xample, f or t ransmucosal a dministration, d etergents, b ile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound a nd t he p articular t herapeutic e ffect t o b e a chieved, a nd t he 1 imitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • instructions for administration will specify use of the composition for hematological and cardiovascular diseases, disorders of the peripheral and central nervous system, COPD, asthma, genito-urological disorders and inflammation diseases.
  • compositions which include an agonist of XPNPEP2 activity, a compound which increases expression of XPNPEP2, or a compound which increases expression or activity of a protein in the XPNPEP2 signaling pathway or any combination thereof
  • the instructions for administration will s pecify use o f the composition for cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders.
  • antibodies which specifically bind XPNPEP2 may be used for the diagnosis of disorders characterized by the expression of XPNPEP2, or i n assays to monitor patients being treated with XPNPEP2 or agonists, antagonists, and inhibitors of XPNPEP2.
  • Antibodies useful for diagnostic pu ⁇ oses may be prepared in the same manner as those described above for therapeutics. Diagnostic assays for
  • XPNPEP2 include methods which utilize the antibody and a label to detect XPNPEP2 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.
  • reporter molecules A wide variety of reporter molecules, several of which are described above, are known in the art and may be used.
  • Normal or standard values for XPNPEP2 expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, preferably human, with antibody to XPNPEP2 under conditions suitable for complex formation. The amount of standard complex formation may be quantified by various methods, preferably by photometric means. Quantities of XPNPEP2 expressed in subject samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease.
  • the polynucleotides encoding XPNPEP2 may be used for diagnostic pu ⁇ oses.
  • the polynucleotides which may be used include o ligonucleotide s equences, 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 XPNPEP2 may be conelated with disease.
  • the diagnostic assay may be used to distinguish between absence, presence, and excess expression of XPNPEP2, and to monitor regulation of XPNPEP2 levels during therapeutic intervention.
  • Polynucleotide sequences encoding XPNPEP2 may be used for the diagnosis of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders associated with expression o f XPNPEP2.
  • T he p olynucleotide sequences encoding XPNPEP2 may be used in Southern, Northern, or dot-blot analysis, o r o ther m embrane-based t echnologies; i n P CR t echnologies; i n d ipstick, pin, and ELISA assays; and in microarrays utilizing fluids or tissues from patient biopsies to detect altered XPNPEP2 expression.
  • Such qualitative or quantitative methods are well known in the art.
  • nucleotide sequences encoding XPNPEP2 may be useful in assays that detect the presence of associated disorders, particularly those mentioned above.
  • the nucleotide sequences encoding XPNPEP2 may be labelled 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.
  • nucleotide sequences have hybridized with nucleotide sequences in the sample, and the presence of altered levels of nucleotide sequences encoding XPNPEP2 in the sample indicates the presence of the associated disorder.
  • 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.
  • 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 XPNPEP2, 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 nonnal 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.
  • a therapeutically effective dose refers to that amount of active ingredient which increases or decreases XPNPEP2 activity relative to XPNPEP2 activity which occurs in the absence of the therapeutically effective dose.
  • the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rabbits, dogs, or pigs. The animal model also can 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.
  • Therapeutic efficacy and toxicity e.g., ED 50 (the dose therapeutically effective in 50% of the population) and LD 50 (the dose lethal to 50% of the population), can be determined b y standard pharmaceutical procedures in cell cultures or experimental animals.
  • the dose ratio of toxic to therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD 5 o/ED 5 o.
  • 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 ED 50 with little or no toxicity.
  • the dosage varies within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration. The exact dosage will be determined by the practitioner, in light of factors related to the subject that requires treatment. Dosage and administration are adjusted to provide sufficient levels of the active ingredient or to maintain the desired effect. Factors which can be taken into account include the severity of the disease state, general health of the subject, 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 can 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.
  • Normal dosage amounts can vary from 0.1 micrograms to 100,000 micrograms, up to a total dose of about 1 g, 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.
  • polynucleotides encoding the antibody can be constructed and introduced into a cell either ex vivo or in vivo using well-established techniques including, but not limited to, transferrin-polycation- mediated DNA transfer, transfection with naked or encapsulated nucleic acids, liposome-mediated cellular fusion, intracellular transportation of DNA-coated latex beads, protoplast fusion, viral infection, electroporation, "gene gun", and DEAE- or calcium phosphate-mediated transfection.
  • the reagent is preferably an antisense oligonucleotide or a ribozyme.
  • Polynucleotides which express antisense oligonucleotides or ribozymes can be introduced into cells by a variety of methods, as described above.
  • a reagent reduces expression of XPNPEP2 gene or the activity of
  • XPNPEP2 by at least about 10, preferably about 50, more preferably about 75, 90, or 100% relative t o t he a bsence o f t he r eagent.
  • T he e ffectiveness o f t he m echanism chosen to decrease the level of expression of XPNPEP2 gene or the activity of XPNPEP2 can be assessed using methods well known in the art, such as hybridization of nucleotide probes to XPNPEP2-specific mRNA, quantitative RT-
  • PCR immunologic detection of XPNPEP2, or measurement of XPNPEP2 activity.
  • any of the pharmaceutical compositions of the invention can be administered in combination with other appropriate therapeutic agents. Selection of the appropriate agents for use in combination therapy can be made by one of ordinary skill in the art, according to conventional pharmaceutical principles.
  • the combination of therapeutic agents can 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.
  • Any of the therapeutic methods described above can 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.
  • Nucleic acid molecules of the invention are those nucleic acid molecules which are contained in a group of nucleic acid molecules consisting of (i) nucleic acid molecules encoding a polypeptide comprising the amino acid sequence of SEQ ID NO: 2, (ii) nucleic acid molecules comprising the sequence of SEQ ID NO: 1, (iii) nucleic acid molecules having the sequence of SEQ ID NO: 1, (iv)nucleic acid molecules the complementary strand of which hybridizes under stringent conditions to a nucleic acid molecule of (i), (ii), or (iii); and (v) nucleic acid molecules the sequence of which differs from the sequence of a nucleic acid molecule of (iii) due to the degeneracy of the genetic code, wherein the polypeptide encoded by said nucleic acid molecule has XPNPEP2 activity.
  • Polypeptides of the invention are those polypeptides which are contained in a group of polypeptides consisting of (i) polypeptides having the sequence of SEQ ID NO: 2, (ii) polypeptides comprising the sequence of SEQ ID NO: 2, (iii) polypeptides encoded by nucleic acid molecules of the invention and (iv) polypeptides which show at least 99%, 98%, 95%, 90%, or 80% homology with a polypeptide of (i), (ii), or (iii), wherein said purified polypeptide has XPNPEP2 activity.
  • An object of the invention is a method of screening for therapeutic agents useful in the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) contacting a test compound with a XPNPEP2 polypeptide, (ii) detect binding of said test compound to said XPNPEP2 polypeptide.
  • compounds that bind to the XPNPEP2 polypeptide are identified potential therapeutic agents for such a disease.
  • Another object of the invention is a method of screening for therapeutic agents useful in the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) determining the activity of a XPNPEP2 polypeptide at a certain concentration of a test compound or in the absence of said test compound, (ii) determining the activity of said polypeptide at a different concentration of said test compound.
  • compounds that lead to a difference in the activity of the XPNPEP2 polypeptide in (i) and (ii) are identified potential therapeutic agents for such a disease.
  • Another object of the invention is a method of screening for therapeutic agents useful in the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) determining the activity of a XPNPEP2 polypeptide at a certain concentration of a test compound, (ii) determining the activity of a XPNPEP2 polypeptide at the presence of a compound known to be a regulator of a XPNPEP2 p olypeptide.
  • compounds that show similar effects on the activity of the XPNPEP2 polypeptide in (i) as compared to compounds used in (ii) are identified potential therapeutic agents for such a disease.
  • Another objects of the invention are methods of the above, wherein the compound is coupled to a detectable label.
  • Other objects of the invention are methods of the above, wherein the test compound displaces a ligand which is first bound to the polypeptide.
  • Another object of the invention is a method of screening for therapeutic agents useful in the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) contacting a test compound with a XPNPEP2 polynucleotide, (ii) detect binding of said test compound to said XPNPEP2 polynucleotide.
  • Compounds that, e.g., bind to the XPNPEP2 polynucleotide are potential therapeutic agents for the treatment of such diseases.
  • Another object of the invention is the method of the above, wherein the nucleic acid molecule is RNA.
  • Another object of the invention is a method of the above, wherein the contacting step is in or at the surface of a cell.
  • Another object of the invention is a method of the above, wherein the contacting step is in a cell-free system.
  • Another object of the invention is a method of the above, wherein the polynucleotide is coupled to a detectable label.
  • Another object of the invention is a method of the above, wherein the test compound is coupled to a detectable label.
  • Another object of the invention is a method of diagnosing a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) determining the amount of a XPNPEP2 polynucleotide in a sample taken from said mammal, (ii) determining the amount of XPNPEP2 polynucleotide in healthy and or diseased mammal.
  • a disease is diagnosed, e.g., if there is a substantial similarity in the amount of XPNPEP2 polynucleotide in said test mammal as compared to a diseased mammal.
  • Another object of the invention is a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a therapeutic agent which binds to a XPNPEP2 polypeptide.
  • Another object of the invention is a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a therapeutic agent which regulates the activity of a
  • Another object of the invention is a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a therapeutic agent which regulates the activity of a XPNPEP2 polypeptide, wherein said therapeutic agent is (i) a small molecule, (ii) an RNA molecule, (iii) an antisense oligonucleotide, (iv) a polypeptide, (v) an antibody, or (vi) a ribozyme.
  • Another object of the invention is a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a XPNPEP2 polynucleotide.
  • Another object of the invention is a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a XPNPEP2 polypeptide.
  • Another object of the invention is the use of regulators of a XPNPEP2 for the preparation of a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal.
  • Another object of the invention is a method for the preparation of a pharmaceutical composition useful for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) identifying a regulator of XPNPEP2, (ii) determining whether said regulator ameliorates the symptoms of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal; and (iii) combining of said regulator with an acceptable pharmaceutical carrier.
  • Another object of the invention is the use of a regulator of XPNPEP2 for the regulation of XPNPEP2 activity in a mammal having a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders.
  • the degree of homology can readily be calculated by known methods. Preferred methods to determine homology are designed to give the largest match between the sequences tested. Methods to determine homology are codified in publicly available computer programs such as BestFit, BLASTP, BLASTN, and FASTA. The BLAST programs are publicly available from NCBI and other sources in the internet.
  • RNA from each cell or tissue source was first reverse transcribed. 85 ⁇ g of total RNA was reverse transcribed using 1 ⁇ mole random hexamer primers, 0.5 mM each of dATP, dCTP, dGTP and dTTP (Qiagen, Hilden, Germany), 3000 U RnaseQut (Invitrogen,
  • the first strand synthesis buffer and Omniscript reverse transcriptase (2 u/ ⁇ l) were from (Qiagen, Hilden, Germany). The reaction was incubated at 37°C for 90 minutes and cooled on ice. The v olume w as a djusted t o 6800 ⁇ 1 w ith w ater, yielding a final c oncenfration of 12.5 ng ⁇ l of starting RNA.
  • XPNPEP2 For relative quantitation of the distribution of XPNPEP2 mRNA in cells and tissues the Perkin Elmer ABI Prism RTM. 7700 Sequence Detection system or Biorad iCycler was used according to the manufacturer's specifications and protocols. PCR reactions were set up to quantitate XPNPEP2 and the housekeeping genes HPRT (hypoxanthine phosphoribosyltransferase), GAPDH (glyceraldehyde-3 -phosphate dehydrogenase), ⁇ -actin, and others. Forward and reverse primers and probes for XPNPEP2 were designed using the Perkin Elmer ABI Primer ExpressTM software and were synthesized by TibMolBiol (Berlin, Germany). The XPNPEP2 forward primer sequence was: Primerl (SEQ ID NO: 3). The XPNPEP2 reverse primer sequence was Primer2 (SEQ ID NO: 4). Probel (SEQ ID NO: 5), labelled with
  • XPNPEP2 (carboxyfluorescein succinimidyl ester) as the reporter dye and TAMRA (carboxytetramethylrhodamine) as the quencher, is used as a probe for XPNPEP2.
  • the following reagents were prepared in a total of 25 ⁇ l : lx TaqMan buffer A, 5.5 mM MgCl 2 , 200 nM of dATP, dCTP, dGTP, and dUTP, 0.025 U/ ⁇ l AmpliTaq GoldTM, 0.01 U/ ⁇ l A pErase and Probel (SEQ ID NO: 4), XPNPEP2 forward and reverse primers each at 200 nM, 200 nM XPNPEP2 FAM/TAMRA-labelled probe, and 5 ⁇ l of template cDNA. Thermal cycling parameters were 2 min at 50°C, followed by 10 min at 95°C, followed by 40 cycles of melting at 95°C for 15 sec and annea
  • the CT (threshold cycle) value is calculated as described in the "Quantitative determination of nucleic acids" section.
  • the CF-value (factor for threshold cycle correction) is calculated as follows : 1. PCR reactions were set up to quantitate the housekeeping genes (HKG) for each cDNA sample.
  • CT H ⁇ G -values were calculated as described in the "Quantitative determination of nucleic acids" section.
  • CTu KG - n -mean value (CT HKGI -value + CTHKG2-value +... + CT HKG - I .- value) / n
  • CT panne i mean value (CT mean value of all HKG in all tested cDNAs)
  • CTcDNA-n CT value of the tested gene for the cDNA n
  • CF cD NA-n correction factor for cDNA n
  • highest CT cor -cDNA-n ⁇ 40 is defined as CT CO r-cDNA [high]
  • Table 1 Relative expression ofXPNPEP2 in various human tissues.
  • fetal heart 72 heart 209 pericardium 116 heart atrium (right) 286 heart atrium (left) 2557 heart ventricle (left) 161 interventricular septum 46 fetal aorta 50 aorta 22 artery 52 coronary artery 48 vein 7 coronary artery smooth muscle primary cells 34
  • liver cirrhosis 27 esophagus 251 esophagus tumor 54 stomach 1323 stomach tumor 734 colon 212 colon tumor 57 small intestine 41765 ileum 143431 ileum tumor 35 ileum chronic inflammation 0 rectum 474 salivary gland 33 fetal liver 8079 liver 160 liver liver cirrhosis 523 liver tumor 324
  • leukocytes peripheral blood
  • Alzheimer cerebral cortex 0 frontal lobe 9
  • HeLa cells (cervix tumor) 5 placenta 226 uterus 87 uterus tumor 20 ovary 1105 ovary tumor 10 breast 2180 breast tumor 172 MDA MB 231 cells (breast tumor) 7 mammary gland 781
  • cDNA sequence coding for XPNPEP2 enables its use as a tool for antisense technology in the investigation of gene function.
  • Oligonucleotides, cDNA or genomic fragments comprising the antisense strand of a polynucleotide coding for XPNPEP2 are used either in vitro or in vivo to inhibit translation of the mRNA.
  • antisense molecules can be designed at various locations along the nucleotide sequences.
  • the gene of interest is effectively turned off.
  • the function of the gene is ascertained by observing behavior at the intracellular, cellular, tissue or organismal level (e.g., lethality, loss of differentiated function, changes in morphology, etc.).
  • modifications of gene expression is obtained by designing antisense sequences to intron regions, promoter/enhancer elements, or even to transacting regulatory genes.
  • XPNPEP2 Expression of XPNPEP2 is accomplished by subcloning the cDNAs into appropriate expression vectors and transfecting the vectors into expression hosts such as, e.g., E. coli.
  • the vector is engineered such that it contains a promoter for ⁇ -galactosidase, upstream of the cloning site, followed by sequence containing the amino-terminal Methionine and the subsequent seven residues of ⁇ -galactosidase.
  • an engineered bacteriophage promoter useful for artificial priming and transcription and for providing a number of unique endonuclease restriction sites for cloning.
  • IPTG Isopropyl- ⁇ -D-thio- galactopyranoside
  • the cDNA is not in the proper reading frame, it is obtained by deletion or insertion of the appropriate number of bases using well known methods including in vitro mutagenesis, digestion with exonuclease m or mung bean nuclease, or the inclusion of an oligonucleotide linker of appropriate length.
  • the XPNPEP2 cDNA is shuttled into other vectors known to be useful for expression of proteins in specific hosts.
  • Oligonucleotide primers containing cloning sites as well as a segment of DNA (about 25 bases) sufficient to hybridize to stretches at both ends of the target cDNA is synthesized chemically by standard methods. These primers are then used to amplify the desired gene segment by PCR.
  • the r esulting g ene segment is digested with appropriate restriction enzymes under standard conditions and isolated by gel electrophoresis. Alternately, similar gene segments are produced by digestion of the cDNA with appropriate restriction enzymes.
  • segments of coding sequence from more than one gene are ligated together and cloned in appropriate vectors. It is possible to optimize expression by construction of such chimeric sequences.
  • Suitable expression hosts for such chimeric molecules include, but are not limited to, mammalian cells such as Chinese Hamster Ovary (CHO) and human 293 cells., insect cells such as Sf9 cells, yeast cells such as Saccharomyces cerevisiae and bacterial cells such as E. coli.
  • a useful expression vector also includes an origin of replication to allow propagation in bacteria, and a selectable marker such as the ⁇ -lactamase antibiotic resistance gene to allow plasmid selection in bacteria.
  • the vector may include a second selectable marker such as the neomycin phosphotransferase gene to allow selection in transfected eukaryotic host cells.
  • Vectors for use in eukaryotic expression hosts require RNA processing elements such as 3' polyadenylation sequences if such are not part of the cDNA of interest.
  • the vector contains promoters or enhancers which increase gene expression.
  • promoters are host specific and include MMTV, SN40, and metallothionine promoters for CHO cells; tip, lac, tac and T7 promoters for bacterial hosts; and alpha factor, alcohol oxidase and PGH promoters for yeast.
  • Transcription enhancers such as the rous sarcoma virus enhancer, are used in mammalian host cells. Once homogeneous cultures of recombinant cells are obtained through standard culture methods, large quantities of recombinantly produced XP ⁇ PEP2 are recovered from the conditioned medium and analyzed using chromatographic methods known in the art.
  • XPNPEP2 can be cloned into the expression vector pcDNA3, as exemplified herein.
  • This product can be used to transform, for example, HEK293 or COS by methodology standard in the art. Specifically, for example, using Lipofectamine (Gibco BRL c atolog no. 1 8324-020) mediated gene transfer.
  • XPNPEP2 is expressed as a chimeric protein with one or more additional polypeptide domains added to facilitate protein purification.
  • purification facilitating domains include, but are not limited to, metal chelating peptides such as histidine- tryptophan modules that allow purification on immobilized metals [Appa Rao, 1997] and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp., Seattle, Washington).
  • the inclusion of a cleavable linker sequence such as Factor Xa or enterokinase (Invitrogen, Groningen, The Netherlands) between the purification domain and the XPNPEP2 sequence is useful to facilitate expression ofXPNPEP2.
  • the following example provides a method for purifying XPNPEP2.
  • XPNPEP2 is generated using the baculovirus expression system BAC-TO-BAC (GIBCO BRL) based on Autographa calif ornica nuclear polyhedrosis virus (AcNPV) infection of Spodoptera frugiperda insect cells (Sf9 cells).
  • the recombinant plasmid is transformed into DH10BAC competent cells which contain the parent bacmid bMON14272 (AcNPV infectious DNA) and a helper plasmid.
  • the mini-Tn7 element on the pFASTBAC donor can transpose to the attTn7 attachment site on the bacmid thus introducing the protease gene into the viral genome. Colonies containing recombinant bacmids are identified by disruption of the lacZ gene.
  • protease/bacmid construct can then be isolated and infected into insect cells (Sf9 cells) resulting in the production of infectious recombinant baculovirus particles and expression of either unfused recombinant enzyme (pFastbacl) or XPNPEP2-His fusion protein (pFastbacHT).
  • insect cells Sf9 cells
  • pFastbacl unfused recombinant enzyme
  • pFastbacHT XPNPEP2-His fusion protein
  • XPNPEP2 Specific Antibodies
  • This denatured protein is used to immunize mice or rabbits using standard protocols; about 100 ⁇ g are adequate for immunization of a mouse, while up to 1 mg might be used to immunize a rabbit.
  • the denatured protein is radioiodinated and used to screen potential murine B-cell hybridomas for those which produce antibody. This procedure requires only small quantities of protein, such that 20 mg is sufficient for labeling and screening of several thousand clones.
  • the amino acid sequence of an appropriate XPNPEP2 domain is analyzed to determine regions of high antigenicity.
  • Ohgopeptides comprising appropriate hydrophilic regions are synthesized and used in suitable immunization protocols to raise antibodies.
  • the optimal amino acid sequences for immunization are usually at the C-terminus, the N- terminus and those intervening, hydrophilic regions of the polypeptide which are likely to be exposed to the external environment when the protein is in its natural conformation.
  • selected peptides typically, about 15 residues in length, are synthesized using an Applied Biosystems Peptide Synthesizer Model 431 A using frnoc-chemistry and coupled to keyhole limpet hemocyanin (EXH; Sigma, St. Louis, MO) by reaction with M-maleimidobenzoyl-N-hydroxysuccinimide ester, MBS. If necessary, a cysteine is introduced at the N-terminus of the peptide to permit coupling to KLH. Rabbits are immunized with the peptide-KLH complex in complete Freund's adjuvant. The resulting antisera are tested for antipeptide activity by binding the peptide to plastic, blocking with 1% bovine serum albumin, reacting with antisera, - Ill -
  • Hybridomas are prepared and screened using standard techniques. Hybridomas of interest are detected by screening with labeled XPNPEP2 to identify those fusions producing the monoclonal antibody with the desired specificity.
  • wells of plates FAST; Becton-Dickinson, Palo Alto, CA
  • affinity purified, specific rabbit anti-mouse (or suitable antispecies 1 g) antibodies at 10 mg/ml.
  • the coated wells are blocked with 1% bovine serum albumin, (BSA), washed and incubated with supernatants from hybridomas. After washing the wells are incubated with labeled XPNPEP2 at 1 mg/ml.
  • BSA bovine serum albumin
  • 10 8 M “1 preferably 10 9 to 10 10 M "1 or stronger, are typically made by standard procedures.
  • Particular XPNPEP2 antibodies are useful for investigating signal transduction and the diagnosis of infectious or hereditary conditions which are characterized by differences in the amount or distribution of XPNPEP2 or downstream products of an active signaling cascade.
  • Diagnostic tests for XPNPEP2 include methods utilizing antibody and a label to detect XPNPEP2 in human body fluids, membranes, cells, tissues or extracts of such.
  • polypeptides and antibodies of the present invention are used with or without modification. Frequently, the polypeptides and antibodies are labeled by joining them, either covalently or noncovalently, with a substance which provides for a detectable signal.
  • labels and conjugation techniques are known and have been reported extensively in both the scientific and patent literature. Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent agents, chemiluminescent agents, chromogenic agents, magnetic particles and the like.
  • a variety of protocols for measuring soluble or membrane-bound XPNPEP2, using either polyclonal or monoclonal antibodies specific for the protein, are known in the art. Examples include enzyme-linked immunosorbent assay (ELISA), radioimmuno- assay (RIA) and fluorescent activated cell sorting (FACS).
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmuno- assay
  • FACS fluorescent activated cell sorting
  • a two-site monoclonal- based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on XPNPEP2 is preferred, but a competitive binding assay may be employed.
  • Native or recombinant XPNPEP2 is purified by immunoaffinity chromatography using antibodies s pecific for X PNPEP2.
  • Polyclonal immunoglobulms are prepared from immune sera either by precipitation with ammonium sulfate or by purification on immobilized Protein A (Pharmacia).
  • monoclonal antibodies are prepared from mouse ascites fluid by ammonium sulfate precipitation or chromatography on immobilized Protein A. Partially purified immunoglobulin is covalently attached to a chromatographic resin such as CnBr-activated Sepharose (Pharmacia LKB Biotechnology). The antibody is coupled to the resin, the resin is blocked, and the derivative resin is washed according to the manufacturer's instructions.
  • a chromatographic resin such as CnBr-activated Sepharose (Pharmacia LKB Biotechnology).
  • Such immunoaffinity columns are utilized in the purification of XPNPEP2 by pre- paring a fraction from cells containing XPNPEP2 in a soluble form.
  • This preparation is derived by solubilization of whole cells or of a subcellular fraction obtained via differential centrifugation (with or without addition of detergent) or by other methods well known in the art.
  • soluble XPNPEP2 containing a signal sequence is secreted in useful quantity into the medium in which the cells are grown.
  • a soluble XPNPEP2-containing preparation is passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of XPNPEP2 (e.g., high ionic strength buffers in the presence of detergent). Then, the column is eluted under conditions that disrupt antibody/protein binding (e.g., a buffer of pH 2-3 or a high concentration of a chaofrope such as urea or thiocyanate ion), and XPNPEP2 is collected.
  • a buffer of pH 2-3 or a high concentration of a chaofrope such as urea or thiocyanate ion
  • This invention is particularly useful for screening therapeutic compounds by using
  • the following example provides a system for drug screening measuring the protease activity.
  • the recombinant protease-His fusion protein can be purified from the crude lysate by metal-affinity chromatography using Ni-NTA agarose. This allows the specific retention of the recombinant material (since this is fused to the His-tag) whilst the endogenous insect proteins are washed off. The recombinant material is then eluted by competition with imidazol.
  • the activity of XPNPEP2 molecules of the present invention can be measured using a variety of assays that measure XPNPEP2 activity. For example, XPNPEP2 enzyme activity can be assessed by a standard in vitro serine/metallo/... protease assay (see, for example, [U.S.
  • the goal of rational drug design is to produce structural analogs of biologically active polypeptides of interest or of small molecules with which they interact, agonists, antagonists, or inhibitors. Any of these examples are used to fashion drugs which are more active or stable forms of the polypeptide or which enhance or interfere with the function of a polypeptide in vivo.
  • the three-dimensional structure of a protein of interest, or of a protein-inhibitor complex is determined by x-ray crystallography, by computer modeling or, most typically, by a combination of the two approaches. Both the shape and charges of the polypeptide must be ascertained to elucidate the structure and to determine active site(s) of the molecule. Less often, useful information regarding the structure of a polypeptide is gained by modeling based on the structure of homologous proteins. In both cases, relevant structural information is used to design efficient inhibitors. Useful examples of rational drug design include molecules which have improved activity or stability or which act as inhibitors, agonists, or antagonists of native peptides.
  • anti-ids to a functional, pharmacologically active antibody.
  • the binding site of the anti-ids is expected to be an analog of the original receptor.
  • the anti-id is then used to identify and isolate peptides from banks of chemically or biologically produced peptides. The isolated peptides then act as the pharmacore.
  • Labeled XPNPEP2 is useful as a reagent for the purification of molecules with which it interacts.
  • XPNPEP2 is covalently coupled to a chromatography column.
  • XPNPEP2-complex is recovered from the column, and the XPNPEP2-binding ligand disassociated and subjected to N-terminal protein sequencing.
  • the amino acid sequence information is then used to identify the captured molecule or to design degenerate oligonucleotide probes for cloning the relevant gene from an appropriate cDNA library.
  • antibodies are raised against XPNPEP2, specifically monoclonal antibodies. The monoclonal antibodies are screened to identify those which inhibit the binding of labeled XPNPEP2. These monoclonal antibodies are then used therapeutically.
  • Example 12 Use and Administration of Antibodies, Inhibitors, or Antagonists
  • LSTs are formulated in a nontoxic, inert, pharmaceutically acceptable aqueous carrier medium preferably at a pH of about 5 to 8, more preferably 6 to 8, although pH may vary according to the characteristics of the antibody, inhibitor, or antagonist being formulated and the condition to be treated. Characteristics of LSTs include solubility of the molecule, its half-life and antigenicity/immunogenicity. These and other characteristics aid in defining an effective carrier. Native human proteins are preferred as LSTs, but organic or synthetic molecules resulting from drug screens are equally effective in particular situations.
  • LSTs are delivered by known routes of administration including but not limited to topical creams and gels; transmucosal spray and aerosol; transdermal patch and bandage; injectable, intravenous and lavage formulations; and orally administered liquids and p ills p articularly formulated to r esist s tomach a cid a nd e nzymes.
  • T particular formulation, exact dosage, and route of administration is determined by the attending physician and varies according to each specific situation.
  • Such determinations are made by considering multiple variables such as the condition to be treated, the LST to be administered, and the pharmacokinetic profile of a particular LST. Additional factors which are taken into account include severity of the disease state, patient's age, weight, gender and diet, time and frequency of LST administration, possible combination with other drugs, reaction sensitivities, and tolerance/response to therapy. Long acting LST formulations might be administered every 3 to 4 days, every week, or once every two weeks depending on half-life and clearance rate of the particular LST.
  • Normal dosage amounts vary from 0.1 to 10 5 ⁇ g, up to a total dose of about 1 g, depending upon the route of administration.
  • Guidance as to particular dosages and methods o f d elivery i s p rovided i n t he 1 iterature; s ee U .S. P at. Nos. 4,657,760; 5,206,344; or 5,225,212.
  • Those skilled in the art employ different formulations for different LSTs.
  • Administration to cells such as nerve cells necessitates delivery in a manner different from that to other cells such as vascular endothelial cells.
  • abnormal signal transduction, trauma, or diseases which trigger XPNPEP2 activity are treatable with LSTs. These conditions or diseases are specifically diagnosed by the tests discussed above, and such testing should be performed in suspected cases of viral, bacterial or fungal infections, allergic responses, mechanical injury associated with trauma, hereditary diseases, lymphoma or carcinoma, or other conditions which activate the genes of lymphoid or neuronal tissues.
  • Animal model systems which elucidate the physiological and behavioral roles of the XPNPEP2 are produced by creating nonhuman transgenic animals in which the activity of the XPNPEP2 is either increased or decreased, or the amino acid sequence of the expressed XPNPEP2 is altered, by a variety of techniques.
  • Examples of these techniques include, but are not limited to: 1) Insertion of normal or mutant versions of DNA encoding a XPNPEP2, by microinj ection, electroporation, retro viral transfection or other means well known to those skilled in the art, into appropriately fertilized embryos in order to produce a transgenic animal or 2) homologous recombination of mutant or normal, human or animal versions of these genes with the native gene locus in transgenic animals to alter the regulation of expression or the structure of these XPNPEP2 sequences.
  • the technique of homologous recombination is well known in the art.
  • transgenic animal One means available for producing a transgenic animal, with a mouse as an example, is as follows: Female mice are mated, and the resulting fertilized eggs are dissected out of their oviducts. The eggs are stored in an appropriate medium such as cesiumchloride M2 medium. DNA or cDNA encoding XPNPEP2 is purified from a vector by methods well known to the one skilled in the art. Inducible promoters may be fused with the coding region of the DNA to provide an experimental means to regulate expression of the transgene. Alternatively or in addition, tissue specific regulatory elements may be fused with the coding region to permit tissue-specific expression of the transgene.
  • microinj ection is not the only method for inserting DNA into the egg but is used here only for exemplary purposes. References

Abstract

The invention provides a human XPNPEP2 which is associated with the cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders. The invention also provides assays for the identification of compounds useful in the treatment or prevention of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders. The invention also features compounds which bind to and/or activate or inhibit the activity of XPNPEP2 as well as pharmaceutical compositions comprising such compounds.

Description

Diagnostics and Therapeutics for Diseases Associated with X-PROLYL AMINOPEPTIDASE 2 (XPNPEP2)
Technical field of the invention
The present invention is in the field of molecular biology, more particularly, the present invention relates to nucleic acid sequences and amino acid sequences of a human XP PEP2 and its regulation for the treatment of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in mammals.
Background of the invention
Proteases play a role in carefully controlled processes, such as blood coagulation, fibrinolysis, complement activation, fertilization, and hormone production. These enzymes are also used in a variety of diagnostic, therapeutic, and industrial contexts. XP PEP2 is a member of the group of protease enzymes [WO200261131, Nenema et al. (1997), Prueitt et al. (2000)].
Proteases were recognized very early in the history of biochemistry. In the nineteenth century, one primary focus of research was on digestive proteases, like pepsin and trypsin. Proteases belong systematically to the C-Ν Hydrolases. More specifically, proteases catalyze the hyprolytic cleavage of a peptide bond and are therefore called peptidases as well.
Proteases can be classified according to several criteria, e.g. by localisation. Digestive proteases are located in the gastro-intestinal tract. These proteases are responsible for the digestion of food proteins. Peptidases located extracellularly in the blood or other extracellular compartments of the body play often regulatory roles in processes like for example blood clotting, fibrinolysis, or activation of complement constituents.
Intracellularly 1 ocated p roteases e xhibit a w ide v ariety o f r oles. T hey are found in compartiments like the ER, the Golgi apparatus, or the lysomes. Their functions include for example activation of peptide hormons, ubiquitin mediated proteolysis, among others.
Proteases are most commonly classified according to their mechanism of action, or to specific active groups that are present in the active center. The following groups can be distinguished:
1. Serin-peptidases, 2. cystein-peptidases, 3. aspartyl- or acidic-peptidases, 4. metallo-peptidases, or 5. peptidases with yet unclear reaction mechanism.
Serine peptidases
Serine p roteases e xhibit a s erine i n t he c atalytic s ite w hich forms a covalent ester intermediate during the catalytic reaction pathway by a nucleophilic attack on the carboxy carbon of the peptide bond. In the active site of serine proteases a catalytic triad comprised of an aspartate, a histidine and the above mentioned serine is found. This triad functions in the reaction mechanism as a charge relay system.
To t he 1 arge family o f s erine p rotease b elong, for e xample, t he digestive enzymes trypsin and chymotrypsin, components of the complement cascade, enzymes involved in the blood clotting cascade, as well as enzymes that function in degradation, rebuilding and maintenace of constituents of the extracellular matrix.
One feature of the serine protease family is the broad range of substrate specificity. Members of the trypase subgroup cleave after arginine or lysine, chymases after phenylalanine or leucine, aspases after aspartate, metases after methionine and serases after serine.
Cysteine proteases
During the catalytic reaction of cysteine proteases a covalent thioester intermediate is formed by a nucleophilic attack of the cysteine on the caboxy carbon of the peptide bond. Similar to the serine peptidases a catalytic triad comprised of the cysteine, a histidine and an asparagine is found which functions as a charge relay system to facilitate the formation of the thioester intermediate.
Members of the Cysteine protease family have roles in many different cellular processes, e.g. processing of precursers or intracellular degradation. Examples for cysteine proteases include lysosomal cathepsines, and cytosolic calpains.
Aspartyl- or acidic peptidase
The catalytic site of aspartyl proteases is composed of two aspartate residues. At the pH optimum of aspartyl proteases (2-3) one of the aspartyl carboxy groups is ionized and the other is neutral, which is important for the catalytic reaction to occur.
Examples for aspartyl proteases are gastric pepsins A and C, chymosin, as well as mammalian renin.
Metallo-peptidases
Metallo-peptidases are proteases, whose proteolytic activity depends on the presence of divalent cations in the active center. Examples of members of this class are carboxypeptidase A, which represents a pancreatic digestive enzyme, the Angiotension Converting Enzymes (ACE), which are responsible for the conversion of angiotensin I to angiotensin π, or the Extracellular Matrix Metalloprotienases. hi summary, a huge number of proteases play a central role in several important cellular and intracellular processes. Furthermore, the value as pharmaceutical targets has been proven for several proteases. For example, the protease encoded by the HIN genome is used as a target for drugs for the treatment of HIN infections, the proteasom complex h as b een discovered as an anti-cancer target, o r C ys-proteases have been implemented as drug targets for inflammatory disorders. Selective inhibitors have been developed as therapeutic agents for diseases such as HIN. Thus, the identification of further disease implications of protease species and their splice variants may lead to the development of specific inhibitors or modulators, or suggest new utilities for known compounds affecting proteases. That in turn will provide additional pharmacological approaches to treat diseases and conditions in which protease activities are involved. This diseases may include, but are not limited to, infections such as bacterial, fungal, protozoan, and viral infections, particularly those caused by HIN viruses, cancers, allergies including asthma, cardiovascular diseases including acute heart failure, hypotension, hypertension, angina pectoris, myocardial infarction, hematological diseases, genito-urinary diseases including urinary incontinence and benign prostate hyperplasia, osteoporosis, and peripheral and central nervous system disorders including pain, Alzheimer's disease and Parkinson's disease.
TaqMan-Technology / expression profiling
TaqMan is a recently developed technique, in which the release of a fluorescent reporter dye from a hybridisation probe in real-time during a polymerase chain reaction (PCR) is proportional to the accumulation of the PCR product.
Quantification is based on the early, linear part of the reaction, and by determining the threshold cycle (CT), at which fluorescence above background is first detected.
Gene expression technologies may be useful in several areas of drug discovery and development, such as target identification, lead optimization, and identification of mechanisms of action. The TaqMan technology can be used to compare differences between expression profiles of normal tissue and diseased tissue. Expression profiling has b een used in identifying genes, which are up- or downregulated in a variety of diseases. An interesting application of expression pro filing is temporal monitoring of changes in gene expression during disease progression and drug treatment or in patients versus healthy individuals. The premise in this approach is that changes in pattern of gene expression in response to physiological or environmental stimuli (e.g., drugs) may serve as indirect clues about disease-causing genes or drug targets. Moreover, the effects of drugs with established efficacy on global gene expression patterns may provide a guidepost, or a genetic signature, against which a new drug candidate can be compared.
XPNPEP2
The nucleotide sequence of XPNPEP2 is accessible in public databases by the accession number NM_003399 and is given in SEQ ID NO:l. The amino acid sequence of XP PEP2 is depicted in SEQ ID NO:2.
Aminopeptidase P is a hydrolase specific for N-terminal imido bonds, which are common to several collagen degradation products, neuropeptides, vasoactive peptides, and cytokines. Structurally, the enzyme is a member of the 'pita bread fold' family and occurs in mammalian tissues in both soluble and GPI-anchored membrane-bound forms. A membrane-bound and soluble form of this enzyme have been identified as products of two separate genes.
By RT-PCR u sing d egenerate o ligonucleotides b ased o n the pig aminopeptidase P amino acid sequence, and by 5-prime and 3-prime RACE, Nenema et al. (1997) isolated human kidney and lung cDΝAs encoding XPΝPEP2. The deduced XPNPEP2 protein has 673 amino acids and an estimated molecular mass of 75,490 Da. The authors stated that the human and pig XPNPEP2 amino acid sequences show significant evolutionary divergence, with 83% identity; 5 of 6 potential N- glycosylation sites, and 5 of 6 cysteine residues that are potentially involved in disulfide bond formation, are conserved. Northern blot analysis detected a 3.5-kb XPNPEP2 transcript in human kidney, lung, heart, placenta, liver, small intestine, and colon, but not in brain, skeletal muscle, pancreas, spleen, thymus, prostate, testis, ovary, or leukocytes. Based on these results and on RT-PCR studies, Nenema et al. (1997) suggested that the membrane-bound form and the soluble form, which is found in brain and leukocytes, are the products of 2 different genes or of alternative splicing of a single primary transcript
Women with balanced translocations b etween the 1 ong arm o f the X chromosome and an autosome frequently suffer premature ovarian failure (POF). Two critical regions for P OF which extend from Xql3 to Xq22 and from Xq22 to Xq26 have been identified by cytogenetic studies. To gain insight into the mechanism(s) responsible for ovarian failure in women with X/autosome translocations, Prueitt et al. (2000) molecularly characterized the translocation breakpoints of 9 X chromosomes. They mapped the breakpoints using somatic cell hybrids retaining the derivative autosome and densely spaced markers from the X-chromosome physical map. One of the POF-associated breakpoints in a critical region (Xq25) mapped to a sequenced PAC clone. The translocation disrupts XPΝPEP2. XPNPEP2 mRNA was detected in fibroblasts that carried the translocation, suggesting that this gene at least partially escapes X inactivation. Although the physiologic substrates for the enzyme were not known, Prueitt et al. (2000) suggested that XPNPEP2 is a candidate gene for POF.
XPNPEP2 is published in WO200261131. XPNPEP2 shows the highest homology (42%) to the human X-Pro aminopeptidase 1 as shown in example 1.
Summary of the invention
The invention relates to novel disease associations of XPNPEP2 polypeptides and polynucleotides. The invention also relates to novel methods of screening for therapeutic agents for the treatment of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal. The invention also relates to pharmaceutical compositions for the treatment of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a XPNPEP2 polypeptide, a XPNPEP2 polynucleotide, or regulators of XPNPEP2 or modulators of XPNPEP2 activity. The invention further comprises methods of diagnosing cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal.
Brief Description of the Drawings
Fig. 1 shows the nucleotide sequence of a XPNPEP2 polynucleotide (SEQ ID NO: 1).
Fig. 2 shows the amino acid sequence of a XPNPEP2 polypeptide (SEQ ID NO:2).
Fig. 3 shows the nucleotide sequence of a primer useful for the invention (SEQ ID
NO:3).
Fig. 4 shows the nucleotide sequence of a primer useful for the invention (SEQ ID NO:4).
Fig. 5 shows a nucleotide sequence useful as a probe to detect proteins of the invention (SEQ ID NO:5).
Detailed description of the invention
Definition of terms
An "oligonucleotide" is a stretch of nucleotide residues which has a sufficient number of bases to be used as an oligomer, amplimer or probe in a polymerase chain reaction (PCR). Oligonucleotides are prepared from genomic or cDNA sequence and are used to amplify, reveal, or confirm the presence of a similar DNA or RNA in a particular cell or tissue. Oligonucleotides or oligomers comprise portions of a DNA sequence having at least about 10 nucleotides and as many as about 35 nucleotides, preferably about 25 nucleotides.
"Probes" may be derived from naturally occurring or recombinant single- or double- stranded nucleic acids or may be chemically synthesized. They are useful in detecting the presence of identical or similar sequences. Such probes may be labeled with reporter molecules using nick translation, Klenow fill-in reaction, PCR or other methods well known in the art. Nucleic acid probes may be used in southern, northern or in situ hybridizations to determine whether DNA or RNA encoding a certain protein is present in a cell type, tissue, or organ.
A "fragment of a polynucleotide" is a nucleic acid that comprises all or any part of a given nucleotide molecule, the fragment having fewer nucleotides than about 6 kb, preferably fewer than about 1 kb.
"Reporter molecules" are radionuclides, enzymes, fluorescent, chemiluminescent, or chromogenic agents which associate with a particular nucleotide or amino acid sequence, thereby establishing the presence of a certain sequence, or allowing for the quantification of a certain sequence.
"Chimeric" molecules may be constructed by introducing all or part of the nucleotide sequence of this invention into a vector containing additional nucleic acid sequence which might be expected to change any one or several of the following XPNPEP2 characteristics: cellular location, distribution, ligand-binding affinities, interchain affinities, degradation/turnover rate, signaling, etc.
"Active", with respect to a XPNPEP2 polypeptide, refers to those forms, fragments, or domains of a XPNPEP2 polypeptide which retain the biological and/or antigenic activity of a XPNPEP2 polypeptide. "Naturally occurring XPNPEP2 polypeptide" refers to a polypeptide produced by cells which have not been genetically engineered and specifically contemplates various polypeptides arising from post-translational modifications of the polypeptide including but not limited to acetylation, carboxylation, glycosylation, phos- phorylation, lipidation and acylation.
"Derivative" refers to polypeptides which have been chemically modified by techniques such as ubiquitination, labeling (see above), pegylation (derivatization with polyethylene glycol), and chemical insertion or substitution of amino acids such as ornithine which do not normally occur in human proteins.
"Conservative amino acid substitutions" result from replacing one amino acid with another having similar structural and/or chemical properties, such as the replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, or a threonine with a serine.
"Insertions" or "deletions" are typically in the range of about 1 to 5 amino acids. The variation allowed may be experimentally determined by producing the peptide synthetically while systematically making insertions, deletions, or substitutions of nucleotides in the sequence using recombinant DNA techniques.
A "signal sequence" or "leader sequence" can be used, when desired, to direct the polypeptide through a membrane of a cell. Such a sequence may be naturally present on the polypeptides of the present invention or provided from heterologous sources by recombinant DNA techniques.
An " oligopeptide" i s a s hort s tretch of amino acid residues and may be expressed from an oligonucleotide. Ohgopeptides comprise a stretch of amino acid residues of at least 3, 5, 10 amino acids and at most 10, 15, 25 amino acids, typically of at least 9 to 13 amino acids, and of sufficient length to display biological and/or antigenic activity. "Inhibitor" i s any substance which retards or prevents a chemical or physiological reaction or response. Common inhibitors include but are not limited to antisense molecules, antibodies, and antagonists.
"Standard expression" is a quantitative or qualitative measurement for comparison. It is based on a statistically appropriate number of normal samples and is created to use as a basis of comparison when performing diagnostic assays, running clinical trials, or following patient treatment profiles.
"Animal" as used herein may be defined to include human, domestic (e.g., cats, dogs, etc.), agricultural (e.g., cows, horses, sheep, etc.) or test species (e.g., mouse, rat, rabbit, etc.).
A "XPNPEP2 polynucleotide", within the meaning of the invention, shall be understood as being a nucleic acid molecule selected from a group consisting of
(i) nucleic acid molecules encoding a polypeptide comprising the amino acid sequence of SEQ ID NO: 2,
(ii) nucleic acid molecules comprising the sequence of SEQ ID NO: 1,
(iii) nucleic acid molecules having the sequence of SEQ ID NO: 1,
(iv) nucleic acid molecules the complementary strand of which hybridizes under stringent conditions to a nucleic acid molecule of (i), (ii), or (iii); and
(v) nucleic acid molecules the sequence of which differs from the sequence of a nucleic acid molecule of (iii) due to the degeneracy of the genetic code; wherein the polypeptide encoded by said nucleic acid molecule has XPNPEP2 activity.
A "XPNPEP2 polypeptide", within the meaning of the invention, shall be understood as being a polypeptide selected from a group consisting of
(i) polypeptides having the sequence of SEQ ID NO: 2,
(ii) polypeptides comprising the sequence of SEQ LD NO: 2,
(iii) polypeptides encoded by XPNPEP2 polynucleotides; and
(iv) polypeptides which show at least 99%, 98%, 95%, 90%, or 80% homology with a polypeptide of (i), (ii), or (iii);
wherein said polypeptide has XPNPEP2 activity.
The nucleotide sequences encoding a XPNPEP2 (or their complement) have numerous applications in techniques known to those skilled in the art of molecular biology. These techniques include use as hybridization probes, use in the construction of oligomers for PCR, use for chromosome and gene mapping, use in the recombinant production of XPNPEP2, and use in generation of antisense DNA or RNA, their chemical analogs and the like. Uses of nucleotides encoding a XPNPEP2 disclosed h erein are exemplary o f known techniques and are not intended to limit their use in any technique known to a person of ordinary skill in the art.
Furthermore, the nucleotide sequences disclosed herein may be used in molecular biology techniques that have not yet been developed, provided the new techniques rely on properties of nucleotide sequences that are currently known, e.g., the triplet genetic code, specific base pair interactions, etc. It will be appreciated by those skilled in the art that as a result of the degeneracy of the genetic code, a multitude of XPNPEP2 - encoding nucleotide sequences may be produced. Some of these will only bear minimal homology to the nucleotide sequence of the known and naturally occurring XPNPEP2. The invention has specifically contemplated each and every possible variation of nucleotide 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 nucleotide sequence of naturally occurring XPNPEP2, and all such variations are to be considered as being specifically disclosed.
Although the nucleotide sequences which encode a XPNPEP2, its derivatives or its variants are preferably capable of hybridizing to the nucleotide sequence of the naturally occurring XPNPEP2 polynucleotide under stringent conditions, it may be advantageous to produce nucleotide sequences encoding XPNPEP2 polypeptides or its derivatives possessing a substantially different codon usage. Codons can be selected to increase the rate at which expression of the peptide occurs in a particular prokaryotic or eukaryotic expression host in accordance with the frequency with which particular codons are utilized by the host. Other reasons for substantially altering the nucleotide sequence encoding a XPNPEP2 polypeptide and/or its derivatives without altering the encoded amino acid sequence include the production of RNA transcripts having more desirable properties, such as a greater half-life, than transcripts produced from the naturally occurring sequence.
Nucleotide sequences encoding a XPNPEP2 polypeptide may be joined to a variety of other nucleotide sequences by means of well established recombinant DNA techniques. U seful nucleotide sequences for j oining to XPNPEP2 polynucleotides include an assortment of cloning vectors such as plasmids, cosmids, lambda phage derivatives, phagemids, and the like. Vectors of interest include expression vectors, replication vectors, probe generation vectors, sequencing vectors, etc. In general, vectors of interest may contain an origin of replication functional in at least one organism, convenient restriction endonuclease sensitive sites, and selectable markers for one or more host cell systems.
Another aspect of the subject invention is to provide for XPNPEP2-specific hybridi- zation probes capable of hybridizing with naturally occurring nucleotide sequences encoding XPNPEP2. Such probes may also be used for the detection of similar protease encoding sequences and should preferably show at least 40% nucleotide identity to XPNPEP2 polynucleotides. The hybridization probes of the subject invention may be derived from the nucleotide sequence presented as SEQ ID NO: 1 or from genomic sequences including promoter, enhancers or introns of the native gene. Hybridization probes may be labelled by a variety of reporter molecules using techniques well known in the art.
It will be recognized that many deletional or mutational analogs of XPNPEP2 poly- nucleotides will be effective hybridization probes for XPNPEP2 polynucleotides.
Accordingly, the invention relates to nucleic acid sequences that hybridize with such XPNPEP2 encoding nucleic acid sequences under stringent conditions.
"Stringent conditions" refers to conditions that allow for the hybridization of substantially related nucleic acid sequences. For instance, such conditions will generally allow hybridization of sequence with at least about 85% sequence identity, preferably with at least about 90% sequence identity, more preferably with at least about 95% sequence identity. Hybridization conditions and probes can be adjusted in well-characterized ways to achieve selective hybridization of human-derived probes. Stringent conditions, within the meaning of the invention are 65°C in a buffer containing 1 mM EDTA, 0.5 MNaHPO4 (pH 7.2), 7 % (w/v) SDS.
Nucleic acid molecules that will hybridize to XPNPEP2 polynucleotides under stringent conditions can be identified functionally. Without limitation, examples of the uses for hybridization probes include: histochemical uses such as identifying tissues that express XPNPEP2; measuring mRNA levels, for instance to identify a sample's tissue type or to identify cells that express abnormal levels of XPNPEP2; and detecting polymorphisms of XPNPEP2.
PCR provides additional uses for oligonucleotides based upon the nucleotide sequence which encodes XPNPEP2. Such probes used in PCR may be of recombinant origin, chemically synthesized, or a mixture of both. Oligomers may comprise discrete nucleotide sequences employed under optimized conditions for identification of XPNPEP2 in specific tissues or diagnostic use. The same two oligomers, a nested set of oligomers, or even a degenerate pool of oligomers may be employed under less stringent conditions for identification of closely related DNAs or RNAs.
Rules for designing polymerase chain reaction (PCR) primers are now established, as reviewed by PCR Protocols. Degenerate primers, i.e., preparations of primers that are heterogeneous at given sequence locations, can be designed to amplify nucleic acid sequences that are highly homologous to, but not identical with XPNPEP2.
Strategies are now available that allow for only one of the primers to be required to specifically hybridize with a known sequence. For example, appropriate nucleic acid primers can be ligated to the nucleic acid sought to be amplified to provide the hybridization partner for one of the primers. In this way, only one of the primers need be based on the sequence of the nucleic acid sought to be amplified.
PCR methods for amplifying nucleic acid will utilize at least two primers. One of these primers will be capable of hybridizing to a first strand of the nucleic acid to be amplified and of priming enzyme-driven nucleic acid synthesis in a first direction. The other will be capable of hybridizing the reciprocal sequence of the first strand (if the sequence to be amplified is single stranded, this sequence will initially be hypothetical, but will be synthesized in the first amplification cycle) and of priming nucleic acid synthesis from that strand in the direction opposite the first direction and towards the site of hybridization for the first primer. Conditions for conducting such amplifications, particularly under preferred stringent hybridization conditions, are well known. Other means o f producing specific hybridization probes for XPNPEP2 include the cloning of nucleic acid sequences encoding XPNPEP2 or XPNPEP2 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 polymerase as T7 or SP6 RNA polymerase and the appropriate reporter molecules.
It is possible to produce a DNA sequence, or portions thereof, entirely by synthetic chemistry. After synthesis, the nucleic acid sequence can be inserted into any of the many available DNA vectors and their respective host cells using techniques which are well known in the art. Moreover, synthetic chemistry may be used to introduce mutations into the nucleotide sequence. Alternately, a portion of sequence in which a mutation is desired can be synthesized and recombined with longer portion of an existing genomic or recombinant sequence.
XPNPEP2 polynucleotides may be used to produce a purified oligo-or polypeptide using well known methods of recombinant DNA technology. The ohgopeptide may be expressed in a variety of host cells, either prokaryotic or eukaryotic. Host cells may be from the same species from which the nucleotide sequence was derived or from a different species. Advantages of producing an oligonucleotide by recombinant DNA technology include obtaining adequate amounts of the protein for purification and the availability of simplified purification procedures.
Quantitative determinations of nucleic acids
An important step in the molecular genetic analysis of human disease is often the enumeration of the copy number of a nucleis acid or the relative expression of a gene in particular tissues. Several different approaches are currently available to make quantitative determinations of nucleic acids. Chromosome-based techniques, such as comparative genomic hybridization (CGH) and fluorescent in situ hybridization (FISH) facilitate efforts to cytogenetically localize genomic regions that are altered in tumor cells. Regions of genomic alteration can be narrowed further using loss of heterozygosity analysis (LOH), in which disease DNA is analyzed and compared with normal DNA for the loss of a heterozygous polymorphic marker. The first experiments used restriction fragment length polymorphisms (RFLPs) [Johnson, (1989)], or hyper- variable minisatellite DNA [Barnes, 2000]. In recent years LOH has been performed primarily using PCR amplification of microsatellite markers and electrophoresis of the radio labelled [Jeffreys, (1985)] or fluorescently labelled PCR products [Weber, (1990)] and compared between paired normal and disease DNAs.
A number of other methods have also been developed to quantify nucleic acids [Gergen, (1992)]. More recently, PCR and RT-PCR methods have been developed which are capable of measuring the amount of a nucleic acid in a sample. One approach, for example, measures PCR product quantity in the log phase of the reaction before the formation of reaction products plateaus [Thomas, (1980)].
A gene sequence contained in all samples at relatively constant quantity is typically utilized for sample amplification efficiency normalization. This approach, however, suffers from several drawbacks.. The method requires that each s ample has equal input amounts of the nucleic acid and that the amplification efficiency between samples is identical until the time of analysis. Furthermore, it is difficult using the conventional methods of PCR quantitation such as gel electrophoresis or plate capture hybridization to determine that all samples are in fact analyzed during the log phase of the reaction as required by the method.
Another method called quantitative competitive (QC)-PCR, as the name implies, relies on the inclusion of an internal control competitor in each reaction [Piatak,
(1993), BioTechniques]. The efficiency of each reaction is normalized to the internal competitor. A known amount of internal competitor is typically added to each sample. The unknown target PCR product is compared with the known competitor PCR product to obtain relative quantitation. A difficulty with this general approach lies in developing an internal control that amplifies with the same efficiency than the target molecule.
5 ' Fluorogenic Nuclease Assays
Fluorogenic nuclease assays are a real time quantitation method that uses a probe to monitor formation of amplification product. The basis for this method of monitoring the formation of amplification product is to measure continuously PCR product accumulation u sing a dual-labelled fluorogenic oligonucleotide probe, an approach frequently referred to in the literature simply as the "TaqMan method" [Piatak,(1993), Science; Heid, (1996); Gibson, (1996); Holland. (1991)].
The probe used in such assays is typically a short (about 20-25 bases) oligonucleotide that is labeled with two different fluorescent dyes. The 5' terminus of the probe is attached to a reporter dye and the 3' terminus is attached to a quenching dye, although the dyes could be attached at other locations on the probe as well. The probe is designed to have at least substantial sequence complementarity with the probe binding site. Upstream and downstream PCR primers which bind to flanking regions of the locus are added to the reaction mixture. When the probe is intact, energy transfer between the two fluorophors occurs and the quencher quenches emission from the reporter. During the extension phase of PCR, the probe is cleaved by the 5' nuclease activity of a nucleic acid polymerase such as Taq polymerase, thereby releasing the reporter from the oligonucleotide-quencher and resulting in an increase of reporter emission intensity which can be measured by an appropriate detector.
One detector which is specifically adapted for measuring fluorescence emissions such as those created during a fluorogenic assay is the ABI 7700 or 4700 HT manufactured by Applied Biosystems, Inc. in Foster City, Calif. The ABI 7700 uses fiber o ptics c onnected w ith e ach w ell i n a 96-or 3 84 w ell P CR tube arrangement. The instrument includes a laser for exciting the labels and is capable of measuring the fluorescence spectra intensity from each tube with continuous monitoring during PCR amplification. Each tube is re-examined every 8.5 seconds.
Computer software provided with the instrument is capable of recording the fluorescence intensity of reporter and quencher over the course of the amplification. The recorded values will then be used to calculate the increase in normalized reporter emission intensity on a continuous basis. The increase in emission intensity is plotted versus time, i.e., the number of amplification cycles, to produce a continuous measure of amplification. To quantify the locus in each amplification reaction, the amplification plot is examined at a point during the log phase of product accumulation. This is accomplished by assigning a fluorescence threshold intensity above background and determining the point at which each amplification plot crosses the threshold (defined as the threshold cycle number or Ct). Differences in threshold cycle number are used to quantify the relative amount of PCR target contained within each tube. Assuming that each reaction functions at 100% PCR efficiency, a difference of one Ct represents a two-fold difference in the amount of starting template. The fluorescence value can be used in conjunction with a standard curve to determine the amount of amplification product present.
Non-Probe-Based Detection Methods
A variety of options are available for measuring the amplification products as they are formed. O ne m ethod u tilizes 1 abels, s uch a s d yes, w hich o nly b ind t o double stranded DNA. In this type of approach, amplification product (which is double stranded) binds dye molecules in solution to form a complex. With the appropriate dyes, it is possible to distinguish between dye molecules free in solution and dye molecules bound to amplification product. For example, certain dyes fluoresce only when bound to amplification product. Examples of dyes which can be used in methods of this general type include, but are not limited to, Syber Green.TM. and Pico Green from Molecular Probes, Inc. of Eugene, Oreg., ethidium bromide, propidium i odide, chromomycin, acridine orange, Hoechst 33258, Toto-1, Yoyo-1, DAPI (4',6-diamidino-2-phenylindole hydrochloride).
Another real time detection technique measures alteration in energy fluorescence energy transfer between fluorophors conjugated with PCR primers [Livak, (1995)].
Probe-Based Detection Methods
These detection methods involve some alteration to the structure or conformation of a probe hybridized to the locus between the amplification primer pair. In some instances, the alteration is caused by the template-dependent extension catalyzed by a nucleic acid polymerase during the amplification process. The alteration generates a detectable signal which is an indirect measure of the amount of amplification product formed.
For example, some methods involve the degradation or digestion of the probe during the extension reaction. These methods are a consequence of the 5'-3' nuclease activity associated with some nucleic acid polymerases. Polymerases having this activity cleave mononucleotides or small o ligonucleotides from an o ligonucleotide probe annealed to its complementary sequence located within the locus.
The 3' end of the upstream primer provides the initial binding site for the nucleic acid polymerase. As the polymerase catalyzes extension of the upstream primer and encounters the bound probe, the nucleic acid polymerase displaces a portion of the 5' end of the probe and through its nuclease activity cleaves mononucleotides or oligonucleotides from the probe.
The upstream primer and the probe can be designed such that they anneal to the complementary strand in close proximity to one another. In fact, the 3' end of the upstream primer and the 5' end of the probe may abut one another. In this situation, extension of the upstream primer is not necessary in order for the nucleic acid polymerase to begin cleaving the probe. In the case in which intervening nucleotides separate the upstream primer and the probe, extension of the primer is necessary before the nucleic acid polymerase encounters the 5' end of the probe. Once contact occurs and polymerization continues, the 5'-3' exonuclease activity of the nucleic acid polymerase begins cleaving mononucleotides or oligonucleotides from the 5' end of the probe. Digestion of the probe continues until the remaining portion of the probe dissociates from the complementary strand.
hi solution, the two end sections can hybridize with each other to form a hairpin loop.
In this conformation, the reporter and quencher dye are in sufficiently close proximity that fluorescence from the reporter dye is effectively quenched by the quencher dye. Hybridized probe, in contrast, results in a linearized conformation in which the extent of quenching is decreased. Thus, by monitoring emission changes for the two dyes, it is possible to indirectly monitor the formation of amplification product.
Probes
The labeled probe is selected so that its sequence is substantially complementary to a segment of the test locus or a reference locus. As indicated above, the nucleic acid site to which the probe binds should be located between the primer binding sites for the upstream and downstream amplification primers.
Primers
The primers used in the amplification are selected so as to be capable of hybridizing to sequences at flanking regions of the locus being amplified. The primers are chosen to have at least substantial complementarity with the different strands of the nucleic acid b eing amplified. When a probe is utilized to detect the formation of amplification products, the primers are selected in such that they flank the probe, i.e. are located upstream and downstream of the probe.
The primer must have sufficient length so that it is capable of priming the synthesis of extension products in the presence of an agent for polymerization. The length and composition of the primer depends on many parameters, including, for example, the temperature at which the annealing reaction is conducted, proximity of the probe binding site to that of the primer, relative concentrations of the primer and probe and the particular nucleic acid composition of the probe. Typically the primer includes
15-30 nucleotides. However, the length of the primer may be more or less depending on the complexity of the primer binding site and the factors listed above.
Labels for Probes andPrimeYS
The labels used for labeling the probes or primers of the current invention and which can provide the signal corresponding to the quantity of amplification product can take a variety o f forms. A s indicated above with regard to the 5' fluorogenic nuclease method, a fluorescent signal is one signal which can be measured. However, measurements may also be made, for example, by monitoring radioactivity, colorimetry, absorption, magnetic parameters, or enzymatic activity. Thus, labels which can be employed include, but are not limited to, fluorophors, chromophores, radioactive isotopes, electron dense reagents, enzymes, and ligands having specific binding partners (e.g., biotin-avidin). Monitoring changes in fluorescence is a particularly useful way to monitor the accumulation of amplification products. A number of labels useful for attachment to probes or primers are commercially available including fluorescein and various fluorescein derivatives such as FAM, HEX, TET and JOE (all which are available from Applied Biosystems, Foster City, Calif); lucifer yellow, and coumarin derivatives.
Labels may be attached to the probe or primer using a variety of techniques and can be attached at the 5' end, and or the 3' end and/or at an internal nucleotide. The label can also be attached to spacer arms of various sizes which are attached to the probe or primer. T hese spacer arms are useful for obtaining a desired distance between multiple labels attached to the probe or primer.
In some instances, a single label may be utilized; whereas, in other instances, such as with the 5' fluorogenic nuclease assays for example, two or more labels are attached to the probe. In cases wherein the probe includes multiple labels, it is generally advisable to maintain spacing between the labels which is sufficient to permit separation of the labels during digestion of the probe through the 5'-3' nuclease activity of the nucleic acid polymerase.
Patients Exhibiting Symptoms of Disease
A number of diseases are associated with changes in the copy number of a certain gene. For patients having symptoms of a disease, the real-time PCR method can be used to determine if the patient has copy number alterations which are known to be linked with diseases that are associated with the symptoms the patient has. XPNPEP2 expression
XPNPEP2 fusion proteins
Fusion proteins are useful for generating antibodies against XPNPEP2 polypeptides and for use in various assay systems. For example, fusion proteins can be used to identify proteins which interact with portions of XPNPEP2 polypeptides. Protein affinity chromatography or library-based assays for protein-protein interactions, such as the yeast two-hybrid or phage display systems, can be used for this purpose. Such methods are well known in the art and also can be used as drug screens.
A XPNPEP2 fusion protein comprises two polypeptide segments fused together by means of a peptide bond. The first polypeptide segment can comprise at least 54, 75, 100, 125, 139, 150, 175, 200, 225, 250, 275, 300, 325 or 350 contiguous amino acids of SEQ ID NO: 2 or of a biologically active variant, such as those described above.
The first polypeptide segment also can comprise full-length XPNPEP2.
The second polypeptide segment can be a full-length protein or a protein fragment. Proteins commonly used in fusion protein construction include, but are not limited to β galactosidase, β-glucuronidase, green fluorescent protein (GFP), autofluorescent proteins, including blue fluorescent protein (BFP), glutathione-S-transferase (GST), luciferase, horseradish peroxidase (HRP), and chloramphenicol acetyltransferase (CAT). Additionally, epitope tags are used in fusion protein constructions, including histidine (His) tags, FLAG tags, influenza hemagglutinin (HA) tags, Myc tags, NSN- G tags, and thioredoxin (Trx) tags. Other fusion constructions can include maltose binding protein (MBP), S -tag, Lex a DΝA binding domain (DBD) fusions, GAL4 DΝA binding domain fusions, and herpes simplex virus (HSN) BP16 protein fusions. A fusion protein also can be engineered to contain a cleavage site located adjacent to the XPΝPEP2. Preparation of Polynucleotides
A naturally occurring XPNPEP2 polynucleotide can be isolated free of other cellular components such as membrane components, proteins, and lipids. Polynucleotides can be made by a cell and isolated using standard nucleic acid purification techniques, or synthesized using an amplification technique, such as the polymerase chain reaction (PCR), or by using an automatic synthesizer. Methods for isolating polynucleotides are routine and are known in the art. Any such technique for obtaining a polynucleotide can be used to obtain isolated XPNPEP2 polynucleotides. For example, restriction enzymes and probes can be used to isolate polynucleotide fragments which comprise XPNPEP2 nucleotide sequences. Isolated polynucleotides are in preparations which are free or at least 70, 80, or 90% free of other molecules.
XPNPEP2 cDNA molecules can be made with standard molecular biology techniques, using XPNPEP2 mRNA as a template. XPNPEP2 cDNA molecules can thereafter be replicated using molecular biology techniques known in the art. A n amplification technique, such as PCR, can be used to obtain additional copies of polynucleotides of the invention, using either human genomic DNA or cDNA as a template.
Alternatively, synthetic chemistry techniques can be used to synthesizes XPNPEP2 polynucleotides. The degeneracy of the genetic code allows alternate nucleotide sequences to be synthesized which will encode XPNPEP2 having, for example, an amino acid sequence shown in SEQ ID NO: 2 or a biologically active variant thereof.
Extending Polynucleotides
Various PCR-based methods can be used to extend nucleic acid sequences encoding human XPNPEP2, for example to detect upstream sequences of XPNPEP2 gene such as promoters and regulatory elements. For example, restriction-site PCR uses universal primers to retrieve unknown sequence adjacent to a known locus. 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.
Inverse PCR also can be used to amplify or extend sequences using divergent primers based on a known region. P rimers c an b e d esigned u sing c ommercially available software, such as OLIGO 4.06 Primer Analysis software (National Biosciences Inc., Plymouth, Minn.), to be 22-30 nucleotides in length, to have a GC content of 50% or more, and to anneal to the target sequence at temperatures about 68-72°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.
Another method which can be used is capture PCR, which involves PCR amplification of DNA fragments adjacent to a known sequence in human and yeast artificial chromosome DNA. In this method, multiple restriction enzyme digestions and ligations also can be used to place an engineered double-stranded sequence into an unknown fragment of the DNA molecule before performing PCR.
When screening for full-length cDNAs, it is preferable to use libraries that have been size-selected to include larger cDNAs. Randomly-primed libraries are preferable, in that they will contain 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 can be useful for extension of sequence into 5' non-transcribed regulatory regions.
Commercially available capillary electrophoresis systems can be used to analyze the size or confirm the nucleotide sequence of PCR or sequencing products. For example, capillary sequencing can employ flowable polymers for electrophoretic separation, four different fluorescent dyes (one for each nucleotide) which are laser activated, and detection of the emitted wavelengths by a charge coupled device camera. Output/light intensity can be converted to electrical signal using appropriate equipment and software (e.g., GENOTYPER and Sequence NAVIGATOR, Perkin Elmer), and the entire process from loading of samples to computer analysis and electronic data display can 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.
Obtaining Polypeptides
XPNPEP2 can be obtained, for example, by purification from human cells, by expression of XPNPEP2 polynucleotides, or by direct chemical synthesis.
Protein Purification
XPNPEP2 can be purified from any human cell which expresses the enzyme, including those which have been transfected with expression constructs which express XPNPEP2. A purified XPNPEP2 is separated from other compounds which normally associate with XPNPEP2 in the cell, such as certain proteins, carbohydrates, or lipids, using methods well-known in the art. Such methods include, but are not limited to, size exclusion chromatography, ammonium sulfate fractionation, ion exchange chromatography, affinity chromatography, and preparative gel electrophoresis.
Expression ofXPNPEP2 Polynucleotides
To express XPNPEP2, XPNPEP2 polynucleotides can be inserted into an expression vector which contains the necessary elements for the transcription and translation of the inserted coding sequence. Methods which are well known to those skilled in the art can be used to construct expression vectors containing sequences encoding XPNPEP2 and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
A variety of expression vector/host systems can be utilized to contain and express sequences encoding XPNPEP2. 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., baculo virus), plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMN) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids), or animal cell systems.
The control elements or regulatory sequences are those non-translated regions of the vector - enhancers, promoters, 5' and 3' untranslated regions — which interact with host cellular proteins to carry out transcription and translation. Such elements can 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 i nducible p romoters, c an b e used. F or example, when cloning in bacterial systems, inducible promoters such as the hybrid lacZ promoter of the
BLUESCREPT phagemid (Stratagene, LaJolla, Calif.) or pSPORTl plasmid (Life Technologies) and the like can be used. The baculovirus polyhedrin promoter can 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) can 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 a nucleotide sequence encoding XPΝPEP2, vectors based on SN40 or EBN can be used with an appropriate selectable marker. Bacterial and Yeast Expression Systems
In bacterial systems, a number of expression vectors can be selected. For example, when a large quantity of XPNPEP2 is needed for the induction of antibodies, vectors which direct high level expression of fusion proteins that are readily purified can be used. Such vectors include, but are not limited to, multifunctional E. coli cloning and expression vectors such as BLUESCRIPT (Stratagene). hi a BLUESCRIPT vector, a sequence encoding XPNPEP2 can 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 or pGEX vectors (Promega, Madison, Wis.) also can 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 adsoφtion to glutathione-agarose beads followed by elution in the presence of free glutathione. Proteins made in such systems can 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.
Plant and Insect Expression Systems
If plant expression vectors are used, the expression of sequences encoding XPNPEP2 can be driven by any of a number of promoters. For example, viral promoters such as the 35S and 19S promoters of CaMV can be used alone or in combination with the omega leader sequence from TMN. Alternatively, plant promoters such as the small subunit of RUBISCO or heat shock promoters can be used. These constructs can be introduced into plant cells by direct DΝA transformation or by pathogen-mediated transfection.
An insect system also can be used to express XPΝPEP2. For example, in one such system Autographa californica nuclear polyhedrosis virus (AcNPN) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae. Sequences encoding XPΝPEP2 can 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 XPNPEP2 will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein. The recombinant viruses can then be used to infect S. frugiperda cells or Trichoplusia larvae in which XPNPEP2 can be expressed.
Mammalian Expression Systems
A number of viral-based expression systems can b e used to express XPNPEP2 in mammalian host cells. For example, if an adenovirus is used as an expression vector, sequences encoding XPNPEP2 can be ligated into an adenovirus transcription/- translation complex comprising the late promoter and tripartite leader sequence. Insertion in a non-essential El or E3 region of the viral genome can be used to obtain a viable virus which is capable of expressing XPNPEP2 in infected host cells [Engelhard, 1 994)]. I f desired, transcription enhancers, such as the Rous sarcoma virus (RSV) enhancer, can be used to increase expression in mammalian host cells.
Human artificial chromosomes (HACs) also can be used to deliver larger fragments of DNA than can be contained and expressed in a plasmid. HACs of 6M to 10M are constructed and delivered to cells via conventional delivery methods (e.g., liposomes, polycationic amino polymers, or vesicles). Specific initiation signals also can be used to achieve more efficient translation of sequences encoding XPNPEP2. Such signals include the ATG initiation codon and adjacent sequences. I n cases where sequences encoding XPNPEP2, 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. The initiation codon should be in the correct reading frame to ensure translation of the entire insert. Exogenous translational elements and initiation codons can be of various origins, both natural and synthetic. Host Cells
A host cell strain can be chosen for its ability to modulate the expression of the inserted sequences or to process the expressed XPNPEP2 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 polypeptide also can 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; 10801 University Boulevard, Manassas, VA 20110-2209) and can be chosen to ensure the correct modification and processing of the foreign protein.
Stable e xpression i s p referred for 1 ong-term. h igh-yield p roduction o f r ecombinant proteins. For example, cell lines which stably express XPNPEP2 can be transformed using expression vectors which can contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate v ector. F ollowing t he i ntroduction o f t he v ector, c ells can be allowed to grow for 1-2 days in an enriched medium before they are switched to a selective medium. 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 XPNPEP2 sequences. Resistant clones of stably transformed cells can be proliferated using tissue culture techniques appropriate to the cell type. Any number of selection systems can be used to recover transformed cell lines. These include, but are not limited to, the heφes simplex virus thymidine kinase [Logan, (1984)] and adenine phosphoribosyltransferase [Wigler, (1977)] genes which can be employed in tic or aprf cells, respectively. Also, antimetabolite, antibiotic, or herbicide resistance can be used as the basis for selection. For example, dhfr confers resistance to methotrexate [Lowy, (1980)], npt confers resistance to the aminoglycosides, neomycin and G-418 [Wigler, (1980)], and als and pat confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively [Colbere-Garapin, 1981]. Additional selectable genes have been described. For example, tYpB allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine. Visible markers such as anthocyanins, β-glucuronidase and its substrate GUS, and luciferase and its substrate luciferin, can be used to identify transformants and to quantify the amount of transient or stable protein expression attributable to a specific vector system
Detecting Polypeptide Expression
Although the p resence o f m arker g ene e pression s uggests that a XPNPEP2 polynucleotide is also present, its presence and expression may need to be confirmed. For example, if a sequence encoding XPNPEP2 is inserted within a marker gene sequence, transformed cells containing sequences which encode XPNPEP2 can b e identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a sequence encoding XPNPEP2 under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of XPNPEP2 polynucleotide.
Alternatively, host cells which contain a XPNPEP2 polynucleotide and which express XPNPEP2 can 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. For example, the presence of a polynucleotide s equence e ncoding X PNPEP2 c an b e d etected b y D NA-DNA or DNA- RNA hybridization or amplification using probes or fragments or fragments of polynucleotides encoding XPNPEP2. Nucleic acid amplification-based assays involve the use of oligonucleotides selected from sequences encoding XPNPEP2 to detect transformants which contain a XPNPEP2 polynucleotide. A variety of protocols for detecting and measuring the expression of XPNPEP2, using either polyclonal or monoclonal antibodies specific for the polypeptide, are known in the art. Examples include enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and fluorescence activated cell sorting (FACS). A two- site, monoclonal-based immunoassay using monoclonal antibodies reactive to two non-interfering epitopes on XPNPEP2 can be used, or a competitive binding assay can be employed.
A wide variety of labels and conjugation techniques are known by those skilled in the art and can 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 XPNPEP2 include oligolabeling, nick translation, end- labeling, or PCR amplification using a labeled nucleotide. Alternatively, sequences encoding XPNPEP2 can be cloned into a vector for the production of an mRNA probe. Such vectors are known in the art, are commercially available, and can be used t o s ynthesize RNA p robes i n v itro b y a ddition o f 1 abeled nucleotides and an appropriate RNA polymerase such as T7, T3, or SP6. These procedures can be conducted using a variety of commercially available kits (Amersham Pharmacia Biotech, Promega, and US Biochemical). Suitable reporter molecules or labels which can be used for ease of detection include radionuclides, enzymes, and fluorescent, chemiluminescent, or chromogenic agents, as well as substrates, cofactors, inhibitors, magnetic particles, and the like.
Expression and Purification of Polypeptides
Host cells transformed with XPNPEP2 polynucleotides can be cultured under conditions suitable for the expression and recovery of the protein from cell culture. The polypeptide produced by a transformed cell can 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 XPNPEP2 polynucleotides can be designed to contain signal sequences which direct secretion of soluble XPNPEP2 through a prokaryotic or eukaryotic cell membrane or which direct the membrane insertion of membrane-bound XPNPEP2.
5 As discussed above, other constructions can be used to join a sequence encoding
XPNPEP2 to a nucleotide sequence 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
10 purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (Immunex Coφ., Seattle, Wash.). Inclusion of cleavable linker sequences such as those specific for Factor XA or enterokinase (Invitrogen, San Diego, CA) between the purification domain and XPNPEP2 also can be used to facilitate purification. One such expression vector provides for expression
15. of a fusion protein containing XPNPEP2 and 6 histidine residues preceding a thioredoxin or an enterokinase cleavage site. The histidine residues facilitate purification by -MAC (immobilized metal ion affinity chromatography) Maddox, (1983)], while the enterokinase cleavage site provides a means for purifying XPNPEP2 from the fusion protein [Porath, (1992)].
20
Chemical Synthesis
Sequences encoding XPNPEP2 can be synthesized, in whole or in part, using chemical methods well known in the art. Alternatively, XPNPEP2 itself can be
25 produced using chemical methods to synthesize its amino acid sequence, such as by direct peptide synthesis using solid-phase techniques. Protein synthesis can either be performed using manual techniques or by automation. Automated synthesis can be achieved, for example, using Applied Biosystems 431 A Peptide Synthesizer Perkin Elmer). Optionally, fragments of XPNPEP2 can be separately synthesized and 0 combined using chemical methods to produce a full-length molecule. The newly synthesized peptide can be substantially purified by preparative high performance liquid chromatography. The composition of a synthetic XPNPEP2 can be confirmed by amino acid analysis or sequencing. Additionally, any portion of the amino acid sequence of XPNPEP2 can be altered during direct synthesis and/or combined using chemical methods with sequences from other proteins to produce a variant polypeptide or a fusion protein.
Production of Altered Polypeptides
As will be understood by those of skill in the art, it may be advantageous to produce
XPNPEP2 polynucleotides possessing non-narurally 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.
The nucleotide sequences referred to herein can be engineered using methods generally known in the art to alter XPNPEP2 polynucleotides for a variety of reasons, including but not limited to, alterations which modify the cloning, processing, and/or expression of the polypeptide or mRNA product. DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides can be used to engineer the nucleotide sequences. For example, site-directed mutagenesis can be used to insert new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, introduce mutations, and so forth.
XPNPEP2 Analogs
One general class of XPNPEP2 analogs are variants having an amino acid sequence that is a mutation of the amino acid sequence disclosed herein. Another general class of XPNPEP2 analogs is provided by anti-idiotype antibodies, and fragments thereof, as described below. Moreover, recombinant antibodies comprising anti-idiotype variable domains can be used as analogs (see, for example, [Monfardini et al., (1996)]). Since the variable domains of anti-idiotype XPNPEP2 antibodies mimic XPNPEP2, these domains can provide XPNPEP2 enzymatic activity. Methods of producing anti-idiotypic catalytic antibodies are known to those of skill in the art [Joron et al., (1992), Friboulet et al. (1994), Avalle et al., (1998)].
Another approach to identifying XPNPEP2 analogs is provided by the use of combinatorial libraries. Methods for constructing and screening phage display and other combinatorial libraries are provided, for example, by [Kay et al., Phage Display of Peptides and Proteins (Academic Press 1996), U.S. 5,783,384, U.S. 5,747,334, and
U.S. 5,723,323.
One illustrative in vitro use of XPNPEP2 and its analogs is the production of labeled peptides from a labeled protein substrate. Proteases can also be used in detergents and cleaning solutions. For example, serine proteases are used in solutions to clean and to disinfect contact lenses (see, for example, [U.S. 5,985,629]). Another use for a serine protease is in the formulation of vaccines (see, for example, [U.S. 5,885,814]). Those of skill in the art can devise other uses for molecules having XPNPEP2 activity.
Antibodies
Any type of antibody known in the art can be generated to bind specifically to an epitope of XPNPEP2.
"Antibody" as used herein includes intact immunoglobulin molecules, as well as fragments thereof, such as Fab, F(ab')2, and Fv, which are capable of binding an epitope of XPNPEP2. Typically, at least 6, 8, 10, or 12 contiguous amino acids are required to form an epitope. However, epitopes which involve non-contiguous amino acids may require more, e.g., at least 15, 25, or 50 amino acid. An antibody which specifically binds to an epitope of XPNPEP2 can be used therapeutically, as well as in immunochemical assays, such as Western blots, ELISAs, radioimmunoassays, immunohistochemical assays, immunoprecipitations, or other immunochemical assays known i n t he a rt. N arious i mmunoassays c an b e u sed t o identify antibodies having the desired specificity. Numerous protocols for competitive binding or immunoradiometric assays are well known in the art. Such immunoassays typically involve the measurement of complex formation between an immunogen and an antibody which specifically binds to the XPNPEP2 immunogen.
Typically, an antibody which specifically binds to XPNPEP2 provides a d etection signal at least 5-, 10-, or 20-fold higher than a detection signal provided with other proteins when used in an immunochemical assay. Preferably, antibodies which specifically bind to XPNPEP2 do not detect other proteins in immunochemical assays and can immunoprecipitate XPNPEP2 from solution.
XPNPEP2 can be used to immunize a mammal, such as a mouse, rat, rabbit, guinea pig, monkey, or human, to produce polyclonal antibodies. If desired, XPNPEP2 can be conjugated to a carrier protein, such as bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin. Depending on the host species, various adjuvants can be used to increase the immunological response. Such adjuvants include, but are not limited to, Freund's adjuvant, mineral gels (e.g., aluminum hydroxide), and surface active substances (e.g., lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol). Among adjuvants used in humans, BCG (bacilli Calmette-Guerin) and Corynebacterium parvum are especially useful.
Monoclonal antibodies which specifically bind to XPNPEP2 can be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These techniques include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBN- hybridoma technique [Roberge, (1995)]. In addition, techniques developed for the production of "chimeric antibodies", the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used. Monoclonal and other antibodies also can be "humanized" to prevent a patient from mounting an immune r esponse a gainst t he a ntibody w hen i t i s u sed t herapeutically. Such antibodies may be sufficiently similar in sequence to human antibodies to be used directly in therapy or may require alteration of a few key residues. Sequence differences between rodent antibodies and human sequences can b e m inimized b y replacing residues which differ from those in the human sequences by site directed mutagenesis of individual residues or by grating of entire complementarity determining regions. Antibodies which specifically bind to XPNPEP2 can contain antigen binding sites which are either partially or fully humanized, as disclosed in U.S. 5,565,332.
Alternatively, techniques described for the production of single chain antibodies can be adapted using methods known in the art to produce single chain antibodies which specifically b ind t o XPNPEP2. Antibodies with related specificity, but of distinct idiotypic composition, can be generated by chain shuffling from random combinatorial immunoglobin libraries. Single-chain antibodies also can be constructed using a DNA amplification method, such as PCR, using hybridoma cDNA as a template. Single-chain antibodies can be mono- or bispecific, and can be bivalent or tetravalent. Construction of tetravalent, bispecific single-chain antibodies is taught. A nucleotide sequence encoding a single-chain antibody can be constructed using manual or automated nucleotide synthesis, cloned into an expression construct using standard recombinant DNA methods, and introduced into a cell to express the coding sequence, as described below. Alternatively, single-chain antibodies can be produced directly using, for example, filamentous phage technology.
Antibodies which specifically bind to XPNPEP2 also can be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents. Other types of antibodies can be constructed and used therapeutically in methods of the invention. For example, chimeric antibodies can be constructed as disclosed in WO 93/03151. Binding proteins which are derived from immunoglobulms and which are multivalent and multispecific, such as the "diabodies" described in WO 94/13804, also can be prepared.
Antibodies according to the invention can be purified by methods well known in the art. For example, antibodies can be affinity purified by passage over a column to which XPNPEP2 is bound. The bound antibodies can then be eluted from the column using a buffer with a high salt concentration.
Antisense Oligonucleotides
Antisense oligonucleotides are nucleotide sequences which are complementary to a specific DNA or RNA sequence. Once introduced into a cell, the complementary nucleotides combine with natural sequences produced by the cell to form complexes and block either transcription or translation. Preferably, an antisense oligonucleotide is at least 11 nucleotides in length, but can be at least 12, 15, 20, 25, 30, 35, 40, 45, or 50 or more nucleotides long. Longer sequences also can be used. Antisense oligonucleotide molecules can be provided in a DNA construct and introduced into a cell as described above to decrease the level of XPNPEP2 gene products in the cell.
Antisense oligonucleotides can be deoxyribonucleotides, ribonucleotides, or a combi- nation of both. Oligonucleotides can be synthesized manually or by an automated synthesizer, by covalently linking the 5' end of one nucleotide with the 3' end of another nucleotide with non-phosphodiester internucleotide linkages such alkyl- phosphonates, phosphorothioates, phosphorodithioates, alkylphosphonothioates, alkylphosphonates, phosphoramidates, phosphate esters, carbamates, acetamidate, carboxymethyl esters, carbonates, and phosphate triesters. Modifications of XPNPEP2 gene expression can be obtained by designing antisense oligonucleotides which will form duplexes to the control, 5', or regulatory regions of the XPNPEP2 gene. Oligonucleotides derived from the transcription initiation site, e.g., between 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 chaperons. Therapeutic advances using triplex DNA have been described in the literature [Nicholls, (1993)]. An antisense oligonucleotide also can be designed to block translation of mRNA by preventing the transcript from binding to ribosomes.
Precise complementarity is not required for successful complex formation between an antisense oligonucleotide and the complementary sequence of a XPNPEP2 polynucleotide. Antisense oligonucleotides which comprise, for example, 2, 3, 4, or 5 or more stretches of contiguous nucleotides which are precisely complementary to a
XPNPEP2 polynucleotide, each separated by a stretch of contiguous nucleotides which are not complementary to adjacent XPNPEP2 nucleotides, can provide sufficient targeting specificity for XPNPEP2 mRNA. Preferably, each stretch of complementary contiguous nucleotides is at least 4, 5, 6, 7, or 8 or more nucleotides in length. Non-complementary intervening sequences are preferably 1, 2, 3, or 4 nucleotides in length. One skilled in the art can easily use the calculated melting point of an antisense-sense pair to determine the degree of mismatching which will be tolerated between a particular antisense oligonucleotide and a particular XPNPEP2 polynucleotide sequence. Antisense oligonucleotides can be modified without affecting their ability to hybridize to a XPNPEP2 polynucleotide. These modifications can be internal or at one or both ends of the antisense molecule. For example, internucleoside phosphate linkages can be modified by adding cholesteryl or diamine moieties with varying numbers of carbon residues between the amino groups and terminal ribose. Modified bases and/or sugars, such as arabinose instead of ribose, or a 3', 5'-substituted oligonucleotide in which the 3' hydroxyl group or the
5' phosphate group are substituted, also can be employed in a modified antisense oligonucleotide. These modified oligonucleotides can be prepared by methods well known in the art.
Ribozymes
Ribozymes are RNA molecules with catalytic activity [Uhlmann, (1987)]. Ribozymes can be used to inhibit gene function by cleaving an RNA sequence, as is known in the art. The mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage. Examples include engineered hammerhead motif ribozyme molecules that can specifically and efficiently catalyze endonucleolytic cleavage of specific nucleotide sequences. The coding sequence of a XPNPEP2 polynucleotide can be used to generate ribozymes which will specifically bind to mRNA transcribed from a XPNPEP2 polynucleotide. Methods of designing and constructing ribozymes which can cleave other RNA molecules in trans in a highly sequence specific manner have been developed and described in the art. For example, the cleavage activity of ribozymes can be targeted to specific RNAs by engineering a discrete "hybridization" region into the ribozyme. The hybridization region contains a sequence complementary to the target RNA and thus specifically hybridizes with the target RNA.
Specific ribozyme cleavage sites within a XPNPEP2 RNA target can be identified by scanning the target molecule for ribozyme cleavage sites which include 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 RNA containing the cleavage site can be evaluated for secondary structural features which may render the target inoperable. Suitability of candidate XPNPEP2 RNA targets also can be evaluated by testing accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays. The nucleotide sequences shown in SEQ JJD NO: 1 and its complement provide sources of suitable hybridization region sequences. Longer complementary sequences can be used to increase the affinity of the hybridization sequence for the target. The hybridizing and cleavage regions of the ribozyme can be integrally related such that upon hybridizing to the target RNA through the complementary regions, the catalytic region of the ribozyme can cleave the target.
Ribozymes can be introduced into cells as part of a DNA construct. Mechanical methods, such as microinj ection, liposome-mediated transfection, electroporation, or calcium phosphate precipitation, can be used to introduce a ribozyme-containing DNA construct into cells in which it is desired to decrease XPNPEP2 expression. Alternatively, if it is desired that the cells stably retain the DNA construct, the construct can be supplied on a plasmid and maintained as a separate element or integrated into the genome of the cells, as is known in the art. A ribozyme-encoding DNA construct can include transcriptional regulatory elements, such as a promoter element, an enhancer or UAS element, and a transcriptional terminator signal, for controlling transcription of ribozymes in the cells (U.S. 5,641,673). Ribozymes also can be engineered to provide an additional level of regulation, so that destruction of mRNA occurs only when both a ribozyme and a target gene are induced in the cells.
Screening / Screening Assays
Regulators
Regulators as used herein, refer to compounds that affect the activity of XPNPEP2 in vivo and/or in vitro. Regulators can be agonists and antagonists of XPNPEP2 polypeptide and can be compounds that exert their effect on the XPNPEP2 activity via the enzymatic activity, expression, post-translational modifications or by other means. Agonists of XPNPEP2 are molecules which, when bound to XPNPEP2, increase or prolong the activity of XPNPEP2. Agonists of XPNPEP2 include proteins, nucleic acids, carbohydrates, small molecules, or any other molecule which activate XPNPEP2. Antagonists of XPNPEP2 are molecules which, when bound to
XPNPEP2, decrease the amount or the duration of the activity of XPNPEP2. Antagonists include proteins, nucleic acids, carbohydrates, antibodies, small molecules, or any other molecule which decrease the activity of XPNPEP2.
The term "modulate", as it appears herein, refers to a change in the activity of XPNPEP2 polypeptide. For example, modulation may cause an increase or a decrease in enzymatic activity, binding characteristics, or any other biological, functional, or immunological properties of XPNPEP2.
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.
The invention provides methods (also referred to herein as "screening assays") for identifying compounds which can be used for the treatment of diseases related to XPNPEP2. The methods entail the identification of candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other molecules) which bind to XPNPEP2 and/or have a s timulatory o r i nhibitory e ffect o n t he b iological activity of XPNPEP2 or its expression and then determining which of these compounds have an effect on symptoms or diseases related to XPNPEP2 in an in vivo assay.
Candidate or test compounds or agents which bind to XPNPEP2 and/or have a stimulatory or inhibitory effect on the activity or the expression of XPNPEP2 are identified either in assays that employ cells which express XPNPEP2 (cell-based assays) or in assays with isolated XPNPEP2 (cell-free assays). The various assays can employ a variety of variants of XPNPEP2 (e.g., full-length XPNPEP2, a biologically active fragment of XPNPEP2, or a fusion protein which includes all or a portion of XPNPEP2). Moreover, XPNPEP2 can be derived from any suitable mammalian species (e.g., human XPNPEP2, rat XPNPEP2 or murine XPNPEP2). The assay can be a binding assay entailing direct or indirect measurement of the binding of a test compound or a known XPNPEP2 ligand to XPNPEP2. The assay can also be an activity assay entailing direct or indirect measurement of the activity of XPNPEP2. The assay can also be an expression assay entailing direct or indirect measurement of the expression of XPNPEP2 mRNA or XPNPEP2 protein. The various screening assays are combined with an in vivo assay entailing measuring the effect of the test compound on the symptoms of diseases related to XPNPEP2.
The present invention includes biochemical, cell free assays that allow the identification of inhibitors and agonists of proteases suitable as lead structures for pharmacological drug development. Such assays involve contacting a form of XPNPEP2 (e.g., full-length XPNPEP2, a biologically active fragment of XPNPEP2, or a fusion protein comprising all or a portion of XPNPEP2) with a test compound and determining the ability of the test compound to act as an antagonist (preferably) or an agonist of the enzymatic activity of XPNPEP2.
The activity of XPNPEP2 molecules of the present invention can be measured using a variety of assays that measure XPNPEP2 activity. For example, XPNPEP2 enzyme activity can be assessed by a standard in vitro serine/metallo/... protease assay (see, for example, [U.S. 5 ,057,414]). Those o f skill in the art are aware of a variety of substrates suitable for in vitro assays, such as SucAla-Ala-Pro-Phe-pNA, fluorescein mono-p-guanidinobenzoate hydrochloride, benzyloxycarbonyl-L-Arginyl-S-benzyl- ester, Nalpha-Benzoyl-L-arginine ethyl ester hydrochloride, and the like. In addition, protease assay kits available from commercial sources, such as Calbiochem™ (San Diego, Calif.). For general references, see Barrett (Ed.), Methods in Enzymology, Proteolytic Enzymes: Serine and Cysteine Peptidases (Academic Press Inc. 1994), and Barrett et al., (Eds.), Handbook of Proteolytic Enzymes (Academic Press Inc.
1998). Solution in vitro assays can be used to identify a XPNPEP2 substrate or inhibitor. Solid phase systems can also be used to identify a substrate or inhibitor of a XPNPEP2 polypeptide. For example, a XPNPEP2 polypeptide or XPNPEP2 fusion protein c an b e i mmobilized o nto t he s urface o f a r eceptor c hip o f a commercially available biosensor instrument (BIACORE, Biacore AB; Uppsala, Sweden). The use of this instrument is disclosed, for example, by [Karlsson, (1991), and Cunningham and Wells, (1993)].
In brief, a XPNPEP2 polypeptide or fusion protein is covalently attached, using amine or sulfhydryl chemistry, to dextran fibers that are attached to gold film within a flow cell. A test sample is then passed through the cell. If a XPNPEP2 substrate or inhibitor is present in the sample, it will bind to the immobilized polypeptide or fusion protein, causing a change in the refractive index of the medium, which is detected as a change in surface plasmon resonance of the gold film. This system allows the determination on- and off-rates, from which binding affinity can be calculated, and assessment of the stoichiometry of binding, as well as the kinetic effects of XPNPEP2 mutation. This system can also be used to examine antibody- antigen interactions, and the interactions of other complement/anti-complement pairs.
In one embodiment, the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of XPNPEP2. Such assays can employ full-length XPNPEP2, a biologically active fragment of XPNPEP2, or a fusion protein which includes all or a portion of XPNPEP2. As described in greater detail below, the test compound can be obtained by any suitable means, e.g., from conventional compound libraries.
Determining the ability of the test compound to modulate the activity of XPNPEP2 can be accomplished, for example, by determining the ability of XPNPEP2 to bind to or interact with a target molecule. The target molecule can be a molecule with which
XPNPEP2 binds or interacts with in nature. The target molecule can be a component of a signal transduction pathway which facilitates transduction of an extracellular signal. The target XPNPEP2 molecule can be, for example, a second intracellular protein which has catalytic activity or a protein which facilitates the association of downstream signaling molecules with XPNPEP2.
Determining the ability of XPNPEP2 to bind to or interact with a target molecule can be accomplished by one of the methods described above for determining direct binding. In one embodiment, determining the ability of a polypeptide of the invention to bind to or interact with a target molecule can be accomplished by determining the activity of the target molecule. For example, the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (e.g., intracellular Ca2+, diacylglycerol, IP3, etc.), detecting catalytic/enzymatic activity of the target on an appropriate substrate, detecting the induction of a reporter gene (e.g., a regulatory element that is responsive to a polypeptide of the invention operably linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a cellular response.
In various embodiments of the above assay methods of the present invention, it may be desirable to immobilize XPNPEP2 (or a XPNPEP2 target molecule) to facilitate separation of complex ed from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. B inding of a test compound to XPNPEP2, or interaction of XPNPEP2 with a target molecule in the presence and absence of a candidate c ompound, c an b e accomplished i n any v essel s uitable for containing the reactants. Examples of such vessels include microtitre plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein can be provided which adds a domain that allows one or both of the proteins to be bound to a matrix. For example, glutathione-S-transferase (GST) fusion proteins or gluta- thione-S-transferase fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical; St. Louis, Mo.) or glutathione derivatized microtitre plates, which are then combined with the test compound or the test compound and either the non-adsorbed target protein or XPNPEP2, and the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). F ollowing i ncubation, the b eads or microtitre plate wells are washed to remove any unbound components and complex formation is measured either directly or indirectly, for example, as described above. Alternatively, the complexes can be dissociated from the matrix, and the level of binding or activity of XPNPEP2 can be determined using standard techniques.
Other techniques for immobilizing proteins on matrices can also be used in the screening assays of the invention. For example, either XPNPEP2 or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin.
Biotinylated polypeptide of the invention or target molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals; Rockford, 111.), and immobilized in the wells of streptavidin-coated plates (Pierce Chemical). Alternatively, antibodies reactive with XPNPEP2 or target molecules but which do not interfere with binding of the polypeptide of the invention to its target molecule can be derivatized to the wells of the plate, and unbound target or polypeptide of the invention trapped in the wells by antibody conjugation. Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with XPNPEP2 or target molecule, as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with XPNPEP2 or target molecule.
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 m ethod, 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 XPNPEP2, or fragments thereof, and washed. Bound XPNPEP2 is then detected by methods well known in the art. Purified XPNPEP2 can also be coated directly onto plates for use in the aforementioned drug screening techniques. Alternatively, non- neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support.
In another embodiment, one may use competitive drug screening assays in which neutralizing antibodies capable of binding XPNPEP2 specifically compete with a testcompound for binding XPNPEP2. hi this manner, antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants with XPNPEP2.
The screening assay can also involve monitoring the expression of XPNPEP2. For example, regulators of expression of XPNPEP2 can be identified in a method in which a cell is contacted with a candidate compound and the expression of XPNPEP2 protein or mRNA in the cell is determined. The level of expression of XPNPEP2 protein or mRNA the presence of the candidate compound is compared to the level of expression of XPNPEP2 protein or mRNA in the absence of the candidate compound. The candidate compound can then be identified as a regulator of expression of XPNPEP2 based on this comparison. For example, when expression of XPNPEP2 protein or mRNA protein is greater (statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of XPNPEP2 protein or mRNA expression. Alternatively, when expression of XPNPEP2 protein or mRNA is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of XPNPEP2 protein or mRNA expression. The level of XPNPEP2 protein or mRNA expression in the cells can be determined by methods described below.
Binding Assays
For binding assays, the test compound is preferably a small molecule which binds to and occupies the active site of XPNPEP2 polypeptide, thereby making the ligand binding site inaccessible to substrate such that normal biological activity is prevented. Examples of such small molecules include, but are not limited to, small peptides or peptide-like molecules. Potential ligands which bind to a polypeptide of the invention include, but are not limited to, the natural ligands of known XPNPEP2 proteases and analogues or derivatives thereof.
In binding assays, either the test compound or the XPNPEP2 polypeptide can comprise a detectable label, such as a fluorescent, radioisotopic, chemiluminescent, or enzymatic label, such as horseradish peroxidase, alkaline phosphatase, or luciferase. Detection of a test compound which is bound to XPNPEP2 polypeptide can then be accomplished, for example, by direct counting of radioemmission, b y scintillation counting, or by determining conversion of an appropriate substrate to a detectable product. Alternatively, binding of a test compound to a XPNPEP2 polypeptide can be determined without labeling either of the interactants. For example, a microphysiometer can be used to detect binding of a test compound with a XPNPEP2 polypeptide. A microphysiometer (e.g., Cytosensor™) is an analytical instrument that measures the rate at which a cell acidifies its environment using a light-addressable p otentiometric s ensor ( LAPS). C hanges i n t his acidification rate can be used as an indicator of the interaction between a test compound and XPNPEP2 [Haseloff, (1988)].
Determining the ability of a test compound to bind to XPNPEP2 also can be accomplished using a technology such as real-time Bimolecular Interaction Analysis (BIA) [McConnell, (1992); Sjolander, (1991)]. BIA is a technology for studying biospecific interactions in real time, without labeling any of the interactants (e.g., BIAcore™). Changes in the optical phenomenon surface plasmon resonance (SPR) can be used as an indication of real-time reactions between biological molecules.
In yet another aspect of the invention, a XPNPEP2-like polypeptide can be used as a "bait protein" in a two-hybrid assay or three-hybrid assay [Szabo, (1995); U.S. 5,283,317), to identify other proteins which bind to or interact with XPNPEP2 and modulate its activity. The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constructs. For example, in one construct, polynucleotide encoding XPNPEP2 can be fused to a polynucleotide encoding the DNA binding domain of a known transcription factor (e.g., GAL-4). In the other construct a DNA sequence that encodes an unidentified protein ("prey" or "sample") can be fused to a polynucleotide that codes for the activation domain of the known transcription factor. If the "bait" and the "prey" proteins are able to interact in vivo to form an protein- dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ), which is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected, and cell colonies containing the functional transcription factor can be isolated and used to obtain the DNA sequence encoding the protein which interacts with XPNPEP2.
It may be desirable to immobilize either the XPNPEP2 (or polynucleotide) or the test compound to facilitate separation of the bound form from unbound forms of one or both of the interactants, as well as to accommodate automation of the assay. Thus, either the XPNPEP2-like polypeptide (or polynucleotide) or the test compound can be bound to a solid support. Suitable solid supports include, but are not limited to, glass or plastic slides, tissue culture plates, microtiter wells, tubes, silicon chips, or particles such as beads (including, but not limited to, latex, polystyrene, or glass beads). Any method known in the art can be used to attach XPNPEP2-like polypeptide (or polynucleotide) or test compound to a solid support, including use of covalent and non-covalent linkages, passive absoφtion, or pairs of binding moieties attached respectively to the polypeptide (or polynucleotide) or test compound and the solid support. Test compounds are preferably bound to the solid support in an array, so that the location of individual test compounds can be tracked. Binding of a test compound to XPNPEP2 (or a polynucleotide encoding for XPNPEP2) can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and microcentrifuge tubes.
hi one embodiment, XPNPEP2 is a fusion protein comprising a domain that allows binding of XPNPEP2 to a solid support. For example, glutathione-S-transferase fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, Mo.) or glutathione derivatized microtiter plates, which are then combined with the test compound or the test compound and the non-adsorbed XPNPEP2; the mixture is then incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtiter plate wells are washed to remove any unbound components. Binding of the interactants can be determined either directly or indirectly, as described above. Alternatively, the complexes can be dissociated from the solid support before binding is determined.
Other techniques for immobilizing proteins or polynucleotides on a solid support also can be used in the screening assays of the invention. For example, either XPNPEP2 (or a polynucleotide encoding XPNPEP2) or a test compound can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated XPNPEP2 (or a poly- nucleotide encoding biotinylated XPNPEP2) or test compounds can be prepared from biotin-NHS (N-hydroxysuccinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, 111.) and immobilized in the wells of streptavidin-coated plates (Pierce Chemical). Alternatively, antibodies which specifically bind to XPNPEP2, polynucleotide, or a test compound, but which do not interfere with a desired binding site, such as the active site of XPNPEP2, can be derivatized to the wells of the plate. Unbound target or protein can be trapped in the wells by antibody conjugation.
Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies which specifically bind to XPNPEP2 polypeptide or test compound, enzyme- linked assays which rely on detecting an activity of XPNPEP2 polypeptide, and SDS gel electrophoresis under non-reducing conditions.
Screening for test compounds which bind to a XPNPEP2 polypeptide or polynucleo- tide also can be carried out in an intact cell. Any cell which comprises a XPNPEP2 polypeptide or polynucleotide can be used in a cell-based assay system. A XPNPEP2 polynucleotide can be naturally occurring in the cell or can be introduced using techniques such as those described above. Binding of the test compound to XPNPEP2 or a polynucleotide encoding XPNPEP2 is determined as described above.
Functional Assays
Test compounds can be tested for the ability to increase or decrease XPNPEP2 activity of a XPNPEP2 polypeptide. The XPNPEP2 activity can be measured, for example, using methods described in the specific examples, below. XPNPEP2 activity can be measured after contacting either a purified XPNPEP2 or an intact cell with a test compound. A test compound which decreases XPNPEP2 activity by at least about 10, preferably about 50, more preferably about 75, 90, or 100% is identified as a potential agent for decreasing XPNPEP2 activity. A test compound which increases XPNPEP2 activity by at least about 10, preferably about 50, more preferably about 75, 90, or 100% is identified as a potential agent for increasing XPNPEP2 activity.
Gene Expression
In another embodiment, test compounds which increase or decrease XPNPEP2 gene expression are identified. As used herein, the term "correlates with expression of a polynucleotide" indicates that the detection of the presence of nucleic acids, the same or r elated t o a n ucleic a cid s equence e ncoding X PNPEP2, b y n orthern a nalysis o r realtime PCR is indicative of the presence of nucleic acids encoding XPNPEP2 in a sample, and thereby correlates with expression of the transcript from the poly- nucleotide encoding XPNPEP2. 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. A XPNPEP2 polynucleotide is contacted with a test compound, and the expression of an RNA or polypeptide product of XPNPEP2 polynucleotide is determined. The level of expression of appropriate mRNA or polypeptide in the presence of the test compound is compared to the level of expression of mRNA or polypeptide in the absence of the test compound. The test compound can then be identified as a regulator of expression based on this comparison. For example, when expression of mRNA or polypeptide is greater in the presence of the test compound than in its absence, the test compound is identified as a stimulator or enhancer of the mRNA or polypeptide expression. Alternatively, when expression of the mRNA or polypeptide is less in the presence of the test compound than in its absence, the test compound is identified as an inhibitor of the mRNA or polypeptide expression.
The level of XPNPEP2 mRNA or polypeptide expression in the cells can be determined by methods well known in the art for detecting mRNA or polypeptide. Either qualitative or quantitative methods can be used. The presence of polypeptide products of XPNPEP2 polynucleotide can be determined, for example, using a variety of techniques known in the art, including immunochemical methods such as radioimmunoassay, Western blotting, and immunohistochemistry. Alternatively, polypeptide synthesis can be determined in vivo, in a cell culture, or in an in vitro translation system by detecting incoφoration of labelled amino acids into XPNPEP2.
Such screening can be carried out either in a cell-free assay system or in an intact cell. Any cell which expresses XPNPEP2 polynucleotide can be used in a cell-based assay system. The XPNPEP2 polynucleotide can be naturally occurring in the cell or can be introduced using techniques such as those described above. Either a primary culture or an established cell line can be used. Test Compounds
Suitable test compounds for use in the screening assays of the invention can be obtained from any suitable source, e.g., conventional compound libraries. The test compounds can also be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound" library method; and synthetic library methods using affinity chromatography selection. The biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds [Lam, (1997)]. Examples of methods for the synthesis of molecular libraries can be found in the art. Libraries of compounds may be presented in solution or on beads, bacteria, spores, plasmids or phage.
Modeling of Regulators
Computer modeling and searching technologies permit identification of compounds, or the improvement of already identified compounds, that can modulate XPNPEP2 expression or activity. Having identified such a compound or composition, the active sites or regions are identified. Such sites might typically be the enzymatic active site, regulator binding sites, or ligand binding sites. The active site can be identified using methods known in the art including, for example, from the amino acid sequences of peptides, from the nucleotide sequences of nucleic acids, or from study of complexes of the relevant compound or composition with its natural ligand. In the latter case, chemical or X-ray crystallographic methods can b e used to find the active site by finding where on the factor the complexed ligand is found.
Next, the three dimensional geometric structure of the active site is determined. This can be done by known methods, including X-ray crystallography, which can determine a complete molecular structure. On the other hand, solid or liquid phase NMR can be used to determine certain intramolecular distances. Any other experimental method of structure determination can be used to obtain partial or complete g eometric s tructures. T he g eometric s tructures mayb e m easured w ith a complexed ligand, natural or artificial, which may increase the accuracy of the active site structure determined.
If an incomplete or insufficiently accurate structure is determined, the methods of computer based numerical modeling can be used to complete the structure or improve its accuracy. Any recognized modeling method may be used, including parameterized models specific to particular biopolymers such as proteins or nucleic acids, molecular dynamics models based on computing molecular motions, statistical mechanics models based on thermal ensembles, or combined models. For most types of models, standard molecular force fields, representing the forces between constituent atoms and groups, are necessary, and can be selected from force fields known in physical chemistry. The incomplete or less accurate experimental structures can serve as constraints on the complete and more accurate structures computed by these modeling methods.
Finally, having determined the structure of the active site, either experimentally, by modeling, or by a combination, candidate modulating compounds can be identified by searching databases containing compounds along with information on their molecular structure. Such a search seeks compounds having structures that match the determined active site structure and that interact with the groups defining the active site. Such a search can be manual, but is preferably computer assisted. These compounds found from this search are potential XPNPEP2 modulating compounds.
Alternatively, these methods can be used to identify improved modulating compounds from an already known modulating compound or ligand. The composition of the known compound can be modified and the structural effects of modification can be determined using the experimental and computer modeling methods described above applied to the new composition. The altered structure is then compared to the active site structure of the compound to determine if an improved fit or interaction results. In this manner systematic variations in composition, such as by varying side groups, can be quickly evaluated to obtain modified modulating compounds or ligands of improved specificity or activity.
Therapeutic Indications and Methods
It was found by the present applicant that XPNPEP2 is expressed in various human tissues.
Neurology
CNS disorders include disorders of the central nervous system as well as disorders of the peripheral nervous system.
CNS disorders include, but are not limited to brain injuries, cerebrovascular diseases and their consequences, Parkinson's disease, corticobasal degeneration, motor neuron disease, dementia, including ALS, multiple sclerosis, traumatic brain injury, stroke, post-stroke, post-traumatic brain injury, and small-vessel cerebrovascular disease. Dementias, such as Alzheimer's disease, vascular dementia, dementia with Lewy bodies, frontotemporal dementia and Parkinsonism linked to chromosome 17, frontotemporal dementias, including Pick's disease, progressive nuclear palsy, corticobasal degeneration, Huntington's disease, thalamic degeneration, Creutzfeld- Jakob dementia, HIN dementia, schizophrenia with dementia, and Korsakoff s psychosis, within the meaning of the definition are also considered to be CΝS disorders.
Similarly, cognitive-related disorders, such as mild cognitive impairment, age- associated memory impairment, age-related cognitive decline, vascular cognitive impairment, attention deficit disorders, attention deficit hyperactivity disorders, and memory disturbances in children with learning disabilities are also considered to be CNS disorders.
Pain, within the meaning of this definition, is also considered to be a CNS disorder. Pain can be associated with CNS disorders, such as multiple sclerosis, spinal cord injury, sciatica, failed back surgery syndrome, traumatic brain injury, epilepsy, Parkinson's disease, post-stroke, and vascular lesions in the brain and spinal cord (e.g., infarct, hemorrhage, vascular malformation). Non-central neuropathic pain includes that associated with post mastectomy pain, phantom feeling, reflex sympathetic dystrophy (RSD), trigeminal neuralgiaradioculopathy, post-surgical pain,
H1N/AIDS related pain, cancer pain, metabolic neuropathies (e.g., diabetic neuropathy, vasculitic neuropathy secondary to connective tissue disease), paraneoplastic p olyneuropathy associated, for example, with carcinoma of lung, or leukemia, or lymphoma, or carcinoma of prostate, colon or stomach, trigeminal neuralgia, cranial neuralgias, and post-heφetic neuralgia. Pain associated with peripheral nerve damage, central pain (i.e. due to cerebral ischemia) and various chronic pain i.e., lumbago, back pain (low back pain), inflammatory and/or rheumatic pain. Headache pain (for example, migraine with aura, migraine without aura, and other migraine disorders), episodic and chronic tension-type headache, tension-type like h eadache, c luster headache, and chronic paroxysmal hemicrania are also CΝS disorders.
Visceral pain such as p ancreatits, intestinal cystitis, dysmenorrhea, irritable B owel syndrome, Crohn's disease, biliary colic, ureteral colic, myocardial infarction and pain syndromes of the pelvic cavity, e.g., vulvodynia, orchialgia, urethral syndrome and protatodynia are also CΝS disorders.
Also considered to be a disorder of the nervous system are acute pain, for example postoperative pain, and pain after trauma. The human XPNPEP2 is highly expressed in the following brain tissues: temporal lobe, neuroblastoma SK-N-MC cells, neuroblastoma SH-SY5Y cells. The expression in brain tissues demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose nervous system diseases. Additionally the activity of the human XPNPEP2 can be modulated to treat nervous system diseases.
Cardiovascular Disorders
Heart failure is defined as a pathophysiological state in which an abnormality of cardiac function is responsible for the failure of the heart to pump blood at a rate commensurate with the requirement of the metabolizing tissue. It includes all forms of pumping failures such as high-output and low-output, acute and chronic, right- sided or left-sided, systolic or diastolic, independent of the underlying cause.
Myocardial infarction (MI) is generally caused by an abrupt decrease in coronary blood flow that follows a thrombotic occlusion of a coronary artery previously narrowed by arteriosclerosis. MI prophylaxis (primary and secondary prevention) is included as well as the acute treatment of MI and the prevention of complications.
Ischemic diseases are conditions in which the coronary flow is restricted resulting in a perfusion which is inadequate to meet the myocardial requirement for oxygen. This group of diseases includes stable angina, unstable angina and asymptomatic ischemia.
Arrhythmias include all forms of atrial and ventricular tachyarrhythmias, atrial tachycardia, atrial flutter, atrial fibrillation, atrio-ventricular reentrant tachycardia, preexitation syndrome, ventricular tachycardia, ventricular flutter, ventricular fibrillation, as well as bradycardic forms of arrhythmias.
Hypertensive vascular diseases include primary as well as all kinds of secondary arterial hypertension, renal, endocrine, neurogenic, others. The genes may be used as drug targets for the treatment of hypertension as well as for the prevention of all complications arising from cardiovascular diseases.
Peripheral vascular diseases are defined as vascular diseases in which arterial and/or venous flow is reduced resulting in an imbalance between blood supply and tissue oxygen demand. It includes chronic peripheral arterial occlusive disease (PAOD), acute arterial thrombosis and embolism, inflammatory vascular disorders, Raynaud's phenomenon and venous disorders.
Atherosclerosis is a cardiovascular disease in which the vessel wall is remodeled, compromising the lumen of the vessel. The atherosclerotic remodeling process involves accumulation of cells, both smooth muscle cells and monocyte/macrophage inflammatory cells, in the intima of the vessel wall. These cells take up lipid, likely from the circulation, to form a mature atherosclerotic lesion. Although the formation of these lesions is a chronic process, occurring over decades of an adult human life, the majority of the morbidity associated with atherosclerosis occurs when a lesion ruptures, releasing thrombogenic debris that rapidly occludes the artery. When such an acute event occurs in the coronary artery, myocardial infarction can ensue, and in the worst case, can result in death.
The formation of the atherosclerotic lesion can be considered to occur in five overlapping stages such as migration, lipid accumulation, recruitment of inflammatory cells, proliferation of vascular smooth muscle cells, and extracellular matrix deposition. Each of these processes can be shown to occur in man and in animal models of atherosclerosis, but the relative contribution of each to the pathology and clinical significance of the lesion is unclear.
Thus, a need exists for therapeutic methods and agents to treat cardiovascular pathologies, such as atherosclerosis and other conditions related to coronary artery disease. Cardiovascular diseases include but are not limited to disorders of the heart and the vascular system like congestive heart failure, myocardial infarction, ischemic diseases of the heart, all kinds of atrial and ventricular arrhythmias, hypertensive vascular diseases, peripheral vascular diseases, and atherosclerosis.
Too high or too low levels of fats in the bloodstream, especially cholesterol, can cause long-term problems. The risk to develop atherosclerosis and coronary artery or carotid artery disease (and thus the risk of having a heart attack or stroke) increases with the total cholesterol level increasing. Nevertheless, extremely low cholesterol levels may not be healthy. Examples of disorders of lipid metabolism are hyperlipidemia (abnormally high levels of fats (cholesterol, triglycerides, or both) in the b lood, m ay b e c aused by family history of hyperlipidemia), obesity, a high-fat diet, lack of exercise, moderate to high alcohol consumption, cigarette smoking, poorly controlled diabetes, and an underactive thyroid gland), hereditary hyperlipidemias (type I hyperlipoproteinemia (familial hyperchylomicronemia), type
II hyperlipoproteinemia (familial hypercholesterolemia), type UI hyperlipoproteinemia, type IN hyperlipoproteinemia, or type N hyperlipoproteinemia), hypo- lipoproteinemia, lipidoses (caused by abnormalities in the enzymes that metabolize fats), Gaucher's disease, Νiemann-Pick disease, Fabry's disease, Wolman's disease, cerebrotendinous xanthomatosis, sitosterolemia, Refsum's disease, or Tay-Sachs disease.
Kidney disorders may lead to hypertension or hypotension. Examples for kidney problems p ossibly 1 eading to hypertension are renal artery stenosis, pyelonephritis, glomerulonephritis, kidney tumors, polycistic kidney disease, injury to the kidney, or radiation therapy affecting the kidney. Excessive urination may lead to hypotension.
The human XPΝPEP2 is highly expressed in the following cardiovascular related tissues: heart atrium (left), HUVEC cells. Expression in the above mentioned tissues demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of cardio vascular diseases. Additionally the activity of the human XPNPEP2 can be modulated to treat cardiovascular diseases.
Gastrointestinal and Liver Diseases
Gastrointestinal diseases comprise primary or secondary, acute or chronic diseases of the organs of the gastrointestinal tract which may be acquired or inherited, benign or malignant or metaplastic, and which may affect the organs of the gastrointestinal tract or the body as a whole. They comprise but are not limited to 1) disorders of the esophagus like achalasia, vigoruos achalasia, dysphagia, cricopharyngeal incoordination, pre-esophageal dysphagia, diffuse esophageal spasm, globus sensation, Barrett's metaplasia, gastroesophageal reflux, 2) disorders of the stomach and duodenum like functional dyspepsia, inflammation of the gastric mucosa, gastritis, stress gastritis, chronic erosive gastritis, atrophy of gastric glands, metaplasia of gastric tissues, gastric ulcers, duodenal ulcers, neoplasms of the stomach, 3) disorders of the pancreas like acute or chronic pancreatitis, insufficiency of the exocrinic or endocrinic tissues of the pancreas like steatorrhea, diabetes, neoplasms of the exocrine or endocrine pancreas like 3.1) multiple endocrine neoplasia syndrome, ductal adenocarcmoma, cystadenocarcinoma, islet cell tumors, insulinoma, gastrinoma, carcinoid tumors, glucagonoma, Zollinger-Ellison syndrome, Vipoma syndrome, malabsoφtion syndrome, 4) disorders of the bowel like chronic inflammatory diseases of the bowel, Crohn's disease, ileus, diarrhea and constipation, colonic inertia, megacolon, malabsoφtion syndrome, ulcerative colitis, 4.1) functional bowel disorders like irritable bowel syndrome, 4.2) neoplasms of the bowel like familial polyposis, adenocarcmoma, primary malignant lymphoma, carcinoid tumors, Kaposi's sarcoma, polyps, cancer of the colon and rectum.
Liver diseases comprise primary or secondary, acute or chronic diseases or injury of the liver which may be acquired or inherited, benign or malignant, and which may affect the liver or the body as a whole. They comprise but are not limited to disorders of the bilirubin metabolism, jaundice, syndroms of Gilbert's, Crigler-Najjar, Dubin- Johnson and Rotor; intrahepatic cholestasis, hepatomegaly, portal hypertension, ascites, Budd-Chiari syndrome, portal-systemic encephalopathy, fatty liver, steatosis, Reye's syndrome, liver diseases due to alcohol, alcoholic hepatitis or cirrhosis, fibrosis and cirrhosis, fibrosis and cirrhosis of the liver due to inborn errors of metabolism or exogenous substances, storage diseases, syndromes of Gaucher's,
Zellweger's, Wilson's - disease, acute or chronic hepatitis, viral hepatitis and its variants, inflammatory conditions of the liver due to viruses, bacteria, fungi, protozoa, helminths; drug induced disorders of the liver, chronic liver diseases like primary sclerosing cholangitis, alpha! -antitrypsin-deficiency, primary biliary cirrhosis, postoperative liver disorders like postoperative intrahepatic cholestasis, hepatic granulomas, vascular liver disorders associated with systemic disease, benign or malignant neoplasms of the liver, disturbance of liver metabolism in the new-born or prematurely born.
The human XPNPEP2 is highly expressed in the following tissues of the gastroenterological system: esophagus tumor, stomach, stomach tumor, small intestine, ileum, fetal liver. The expression in the above mentioned tissues and in particular the differential expression between diseased tissue esophagus tumor and healthy tissue esophagus, between diseased tissue stomach tumor and healthy tissue stomach demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of gastroenterological disorders. Additionally the activity of the human XPNPEP2 can be modulated to treat gastroenterological disorders.
Dermatologic Disorders
The skin serves several functions. It's an multi-layered organ system that builds an effective protective cover and regulates body temperature, senses painful and pleasant stimuli, keeps substances from entering the body, and provides a shield from the sun's harmful effects. Skin color, texture, and folds help mark people as individuals. Thus, skin disorders or diseases often have important consequences for physical and mental health. Skin disorders include, but are not limited to the conditions described in the following.
Itching (pruritus) is a sensation that instinctively demands scratching, which may be caused by a skin condition or a systemic diseas.
Superficial Skin Disorders affect the uppermost layer of the skin, the stratum corneum or the keratin layer, and it consists of many layers of flattened, dead cells and acts as a barrier to protect the underlying tissue from injury and infection. Disorders of the superficial skin layers involve the stratum corneum and deeper layers of the epidermis. Examples of superficial skin disorders are provided in the following.
Dry skin often occurs in people past middle age, severe dry skin (ichthyosis) results from an inherited scaling disease, such as ichthyosis vulgaris or epidermolytic hyperkeratosis. Ichthyosis also results from nonhereditary disorders, such as leprosy, underactive thyroid, lymphoma, AIDS, and sarcoidosis.
Keratosis pilaris is a common disorder in which dead cells shed from the upper layer of skin and form plugs that fill the openings of hair follicles.
A callus is an area on the stratum corneum or keratin layer, that becomes abnormally thick in response to repeated rubbing.
A corn is a pea-sized, thickened area of keratin that occurs on the feet.
Psoriasis is a chronic, recurring disease recognizable by silvery scaling bumps and various-sized plaques (raised patches). An abnormally high rate of growth and turnover of skin cells causes the scaling. Pityriasis rosea is a mild disease that causes scaly, rose-colored, inflamed skin. Pityriasis rosea is possibly caused by an infectious agent, although none has b een identified.
Lichen planus, a recurring itchy disease, starts as a rash of small discrete bumps that then combine and become rough, scaly plaques (raised patches).
Dermatitis (eczema) is an inflammation of the upper layers of the skin, causing blisters, redness, swelling, oozing, scabbing, scaling, and usually itching.
Forms of dermatitis are contact dermatitis, or chronic dermatitis of the hands and feet, e.g. Pompholyx.
Further examples of dermatitic disorders are atopic dermatitis, seborrheic dermatitis, nummular dermatitis, generalized exfoliative dermatitis, stasis dermatitis, or localized scratch dermatitis (lichen simplex chronicus, neurodermatitis).
Other skin disorders are caused by inflammation. The skin can break out in a variety of rashes, sores, and blisters. Some skin eruptions can even be life threatening.
Drug rashes are side effects of medications, mainly allergic reactions to medications.
Toxic epidermal necrolysis is a life-threatening skin disease in which the top layer of the skin peels off in sheets. This condition can be caused by a reaction to a drug, or by some other serious disease.
Erythema multiforme, often caused by heφes simplex is a disorder characterized by patches of red, raised skin that often look like targets and usually are distributed symmetrically over the body. Erythema nodosum is an inflammatory disorder that produces tender red bumps (nodules) under the skin, most often over the shins but occasionally on the arms and other areas.
Granuloma annulare is a chronic skin condition of unknown cause in which small, firm, raised bumps form a ring with normal or slightly sunken skin in the center.
Some skin disorders are characterized as blistering diseases. Three autoimmune diseases—pemphigus, bullous pemphigoid, and dermatitis heφetiformis—are among the most serious.
Pemphigus is an uncommon, sometimes fatal, disease in which blisters (bullae) of varying sizes break out on the skin, the lining of the mouth, and other mucous membranes.
Bullous pemphigoid is an autoimmune disease that causes blistering.
Dermatitis heφetiformis is an autoimmune disease in which clusters of intensely itchy, small blisters and hive-like swellings break out and persist. In people with the disease, proteins in wheat, rye, barley, and oat products activate the immune system, which attacks parts of the skin and somehow causes the rash and itching.
Sweating disorders also belong to skin disorders.
Prickly heat is an itchy skin rash caused by trapped sweat.
Excessive sweating (hyperhidrosis) may affect the entire surface of the skin, but often it's limited to the palms, soles, armpits, or groin. The affected area is often pink or bluish white, and in severe cases the skin may be cracked, scaly, and soft, especially on the feet. Skin disorders can affect the sebaceous glands. The sebaceous glands, which secrete oil onto the skin, lie in the dermis, the skin layer just below the surface layer (epidermis). Sebaceous gland disorders include acne, rosacea, perioral dermatitis, and sebaceous cysts.
Acne is a common skin condition in which the skin pores become clogged, leading to pimples and inflamed, infected abscesses (collections of pus). Acne tends to develop in teenagers.
Acne is further subdivided in superficial acne or deep acne.
Rosacea is a persistent skin disorder that produces redness, tiny pimples, and broken blood vessels, usually on the central area of the face.
Perioral dermatitis is a red, often bumpy rash around the mouth and on the chin.
A sebaceous cyst (keratinous cyst) is a slow-growing bump containing dead skin, skin excretions, and other skin particles. These cysts may be small and can appear anywhere.
Hair Disorders also are skin disorders. Hair disorders include excessive hairiness, baldness, and ingrown beard hairs.
The skin can be infected by bacteria. Bacterial skin infections can range in seriousness from minor acne to a life-threatening condition, such as staphylococcal scalded skin syndrome. The most common bacterial skin infections are caused b y Staphylococcus and Streptococcus. Risk factors for skin infections are for example diabetes, ADDS or skin leasons.
Impetigo is a skin infection, caused by Staphylococcus or Streptococcus, leading to the formation of small pus-filled blisters (pustules). Folliculitis is an inflammation of the hair follicles caused by infection with Staphylococcus. The infection damages the hairs, which can be easily pulled out.
Boils (furuncles) are large, tender, swollen, raised areas caused by staphylococcal infection around hair follicles.
Carbuncles are clusters of boils that result in extensive sloughing of skin and scar formation. Carbuncles develop and heal more slowly than single boils and may lead to fever and fatigue.
Erysipelas is a skin infection caused by Streptococcus. A shiny, red, slightly swollen, tender rash develops, often with small blisters. Lymph nodes around the infected area may become enlarged and painful.
Cellulitis is a spreading infection in, and sometimes beneath, the deep layers of the skin. Cellulitis most often results from a streptococcal infection or a staphylococcal infection. However, many other bacteria can also cause cellulitis.
Paronychia is an infection around the edge of a fingernail or toenail. Paronychia can be caused by many different bacteria, including Pseudomonas and Proteus, and by fungi, such as Candida.
Staphylococcal scalded skin syndrome is a widespread skin infection that can lead to toxic shock syndrome, in which the skin peels off as though burned. Certain types of staphylococci produce a toxic substance that causes the top layer of skin (epidermis) to split from the rest of the skin.
Erythrasma is an infection of the top layers of the skin by the bacterium Corynebacterium minutissimum. Skin infections are often caused by fungi. Fungi that infect the skin (dermatophytes) live only in the dead, topmost layer (stratum corneum) and don't penetrate deeper. Some fungal infections cause no symptoms or produce only a small amount of irritation, scaling, and redness. Other fungal infections cause itching, swelling, blisters, and severe scaling.
Ringworm is a fungal skin infection caused by several different fungi and generally classified by its location on the body.
Examples are Athlete's foot (foot ringworm, caused by either Trichophyton or
Epidermophyton), jock itch (groin ringworm, can be caused by a variety of fungi and yeasts), scalp ringworm, caused by Trichophyton or Microsporum), nail ringworm and body ringworm (caused by Trichophyton).
Candidiasis (yeast infection, moniliasis) is an infection by the yeast Candida.
Candida usually infects the skin and mucous membranes, such as the lining of the mouth and vagina. Rarely, it invades deeper tissues as well as the blood, causing life- threatening systemic candidiasis. The following types of Candida infections can be distinguished: Infections in skinfolds (intertriginous infections), vaginal and penile Candida infections (vulvovaginitis), thrush, Perleche (candida infection at the corners of the mouth), candidal paronychia (candida growing in the nail beds, produces painful swelling and pus).
Tinea versicolor is a fungal infection that causes white to light brown patches on the skin.
The skin can also be affected by parasites, mainly tiny insects or worms.
Scabies is a mite infestation that produces tiny reddish pimples and severe itching. Scabies is caused by the itch mite Sarcoptes scabiei. Lice infestation (pediculosis) causes intense itching and can affect almost any area of the skin. Head lice and pubic lice are two different species.
Creeping eruption (cutaneous larva migrans) is a hookworm infection transmitted from warm, moist soil to exposed skin. The infection is caused by a hookworm that normally inhabits dogs and cats.
Many types of viruses invade the skin. The medically important once cause warts and cold sores ( fever b listers) on the lip. Warts are caused by the papillomavirus, and cold sores are caused by the heφes simplex virus. Another important group of viruses that infect the skin belongs to the poxvirus family. C hickenpox r emains a common childhood infection. A poxvirus also causes molluscum contagiosum, which is an infection of the skin by a poxvirus that causes skin-colored, smooth, waxy bumps.
Sunlight can cause severe skin damage. Sunburn results from an overexposure to ultraviolet B (UVB) rays. Some sunburned people develop a fever, chills, and weakness, and those with very bad sunburns even may go into shock—low blood pressure, and fainting.
People who are in the sun a lot have an increased risk of skin cancers, including squamous cell carcinoma, basal cell carcinoma, and to some degree, malignant melanoma.
Drugs, among other causes, can cause skin photosensitivity reactions which can occur after only a few minutes of sun exposure. These reactions include redness, peeling, hives, blisters, and thickened, scaling patches (photosensitivity).
Some skin disorders are characterized as Pigment Disorders.
Albinism is a rare, inherited disorder in which no melanin is formed. Nitiligo is a condition in which a loss of melanocytes results in smooth, whitish patches of skin, which may occur after unusual physical trauma and tends to occur with certain other diseases, including Addison's disease, diabetes, pernicious anemia, and thyroid disease.
Tinea versicolor is a fungal infection of the skin that sometimes results in hypeφigmentation.
Melasma appears on the face (usually the forehead, cheeks, temples, and jaws) as a roughly symmetric group of dark brown patches of pigmentation that are often clearly delineated.
Skin growths, which are abnormal accumulations of different types of cells, may be present at birth or develop later. Νoncancerous (benign) growth and cancerous
(malignant) growth types are distinguished.
Moles (nevi) are small, usually dark, skin growths that develop from pigment- producing cells in the skin (melanocytes). Most moles are harmless. However, noncancerous moles can develop into malignant melanoma.
Skin tags are soft, small, flesh-colored or slightly darker skin flaps that appear mostly on the neck, in the armpits, or in the groin.
Lipomas are soft deposits of fatty material that grow under the skin, causing round or oval lumps.
Angiomas are collections of abnormally dense blood or lymph vessels that are usually located in and below the skin and that cause red or puφle discolorations. Examples of angiomas are port-wine stains, strawberry marks, cavernous hemangiomas, spider angiomas, and lymphangiomas.
Pyogenic granulomas are scarlet, brown, or blue-black slightly raised areas caused by increased growth of capillaries (the smallest blood vessels) and swelling of the surrounding tissue.
Seborrheic keratoses (sometimes called seborrheic warts) are flesh-colored, brown, or black growths that can appear anywhere on the skin.
Dermatofibromas are small, red-to-brown bumps (nodules) that result from an accumulation of fibroblasts, the cells that populate the soft tissue under the skin.
Keratoacanthomas are round, firm, usually flesh-colored growths that have an unusual central crater containing a pasty material.
Keloids are smooth, shiny, slightly pink, often dome-shaped, proliferative growths of fibrous tissue that form over areas of injury or over surgical wounds.
Skin cancer is the most common form of cancer, but most types of skin cancers are curable.
Basal cell carcinoma is a cancer that originates in the lowest layer of the epidermis.
Squamous cell carcinoma is cancer that originates in the middle layer of the epidermis.
Bowen's disease is a form of squamous cell carcinoma that's confined to the epidermis and hasn't yet invaded the underlying dermis. Melanoma is a cancer that originates in the pigment-producing cells of the skin (melanocytes).
Kaposi's sarcoma is a cancer that originates in the blood vessels, usually of the skin.
Paget's disease i s a r are type o f skin c ancer that 1 ooks 1 ike an inflamed, reddened patch of skin (dermatitis); it originates in glands in or under the skin.
The human XPNPEP2 is highly expressed in the following dermatological tissues: skin. The expression in the above mentioned tissues demonstrates that the human
XPNPEP2 or mRNA can be utilized to diagnose of dermatological diseases. Additionally the activity of t he h uman X PNPEP2 c an b e m odulated to t reat t hose diseases.
Cancer Disorder's
Cancer disorders within the scope of this definition comprise any disease of an organ or tissue in mammals characterized by poorly controlled or uncontrolled multiplication of normal or abnormal cells in that tissue and its effect on the body as a whole. Cancer diseases within the scope of the definition comprise benign neoplasms, dysplasias, hypeφlasias as well as neoplasms showing metastatic growth or any other transformations like e.g. leukoplakias which often precede a breakout of cancer. C ells and tissues are c ancerous when they grow more rapidly than normal cells, displacing or spreading into the surrounding healthy tissue or any other tissues of the body described as metastatic growth, assume abnormal shapes and sizes, show changes in their nucleocytoplasmatic ratio, nuclear polychromasia, and finally may cease. Cancerous cells and tissues may affect the body as a whole when causing paraneoplastic syndromes or if cancer occurs within a vital organ or tissue, normal function will be impaired or halted, with possible fatal results. The ultimate involvement of a vital organ by cancer, either primary or metastatic, may lead to the death of the mammal affected. Cancer tends to spread, and the extent of its spread is usually related to an individual's chances of surviving the disease. Cancers are generally said to be in one of three stages of growth: early, or localized, when a tumor is still confined to the tissue of origin, or primary site; direct extension, where cancer cells from the tumour have invaded adjacent tissue or have spread only to regional lymph nodes; or metastasis, in which cancer cells have migrated to distant parts of the body from the primary site, via the blood or lymph systems, and have established secondary sites of infection. Cancer is said to be malignant because of its tendency to cause death if not treated. Benign tumors usually do not c ause death, although they may if they interfere with a normal body function by virtue of their location, size, or paraneoplastic side effects. Hence benign tumors fall under the definition of cancer within the scope of this definition as well. In general, cancer cells divide at a higher rate than do normal cells, but the distinction between the growth of cancerous and normal tissues is not so much the rapidity of cell division in the former as it is the partial or complete loss of growth restraint in cancer cells and their failure to differentiate into a useful, limited tissue of the type that characterizes the functional equilibrium of growth of normal tissue. Cancer tissues may express certain molecular receptors and probably are influenced by the host's susceptibility and immunity and it is known that certain cancers of the breast and prostate, for example, are considered dependent on specific hormones for their existence. The term "cancer" under the scope of the definition is not limited to simple benign neoplasia but comprises any other benign and malign neoplasia like 1) Carcinoma, 2) Sarcoma, 3) Carcinosarcoma, 4) Cancers of the blood-forming tissues, 5) tumors of nerve tissues including the brain, 6) cancer of skin cells. Cancer according to 1) occurs in epithelial tissues, which cover the outer body (the skin) and line mucous membranes and the inner cavitary structures of organs e.g. such as the breast, lung, the respiratory and gastrointestinal tracts, the endocrine glands, and the genitourinary system. Ductal or glandular elements may persist in epithelial tumors, as in adenocarcinomas like e.g. thyroid adenocarcmoma, gastric adenocarcmoma, uterine adenocarcmoma. Cancers of the pavement-cell epithelium of the skin and of certain mucous membranes, such as e .g. cancers of the tongue, lip, larynx, urinary bladder, uterine cervix, or penis, may be termed epidermoid or squamous-cell carcinomas of the respective tissues and are in the scope of the definition of cancer as well. Cancer according to 2) develops in connective tissues, including fibrous tissues, adipose (fat) tissues, muscle, blood vessels, bone, and cartilage like e.g. osteogenic sarcoma; liposarcoma, fibrosarcoma, synovial sarcoma. Cancer according to 3) is cancer that develops in both epithelial and connective tissue. Cancer disease within the scope of this definition may be primary or secondary, whereby primary indicates that the cancer originated in the tissue where it is found rather than was established as a secondary site through metastasis from another lesion. Cancers and tumor diseases within the scope of this definition may be benign or malign and may affect all anatomical structures of the body of a mammal. By example but not limited to they comprise cancers and tumor diseases of I) the bone marrow and bone marrow derived cells (leukemias), IT) the endocrine and exocrine glands like e.g. thyroid, parathyroid, pituitary, adrenal glands, salivary glands, pancreas ILT) the breast, like e.g. benign or malignant tumors in the mammary glands of either a male or a female, the mammary ducts, adenocarcmoma, medullary carcinoma, comedo carcinoma, Paget's disease of the nipple, inflammatory carcinoma of the young woman, IV) the lung, V) the stomach, VI) the liver and spleen, VII) the small intestine, NET) the colon, IX) the bone and its supportive and connective tissues like malignant or benign bone tumour, e.g. malignant osteogenic sarcoma, benign osteoma, cartilage tumors; like malignant chondrosarcoma or benign chondroma; bone marrow tumors like malignant myeloma or b enign e osinophilic granuloma, as well as metastatic tumors from bone tissues at other locations of the body; X) the mouth, throat, larynx, and the esophagus, XI) the urinary bladder and the internal and external organs and structures of the urogenital system of male and female like ovaries, uterus, cervix of the uterus, testes, and prostate gland, XH) the prostate, XIII) the pancreas, like ductal carcinoma of the pancreas; XIN) the lymphatic tissue like lymphomas and other tumors of lymphoid origin, XV) the skin, XVI) cancers and tumor diseases of all anatomical structures belonging to the respiration and respiratory systems including thoracal muscles and linings, XVII) primary or secondary cancer of the lymph nodes XVIII) the tongue and of the bony structures of the hard palate or sinuses, XV-V) the mouth, cheeks, neck and salivary glands, XX) the blood vessels including the heart and their linings, XXI) the smooth or skeletal muscles and their ligaments and linings, X-XJJ) the peripheral, the autonomous, the central nervous system including the cerebellum, XXILT) the adipose tissue.
The human XPNPEP2 is highly expressed in the following cancer tissues: esophagus tumor, stomach tumor, ovary tumor, breast tumor, kidney tumor. The expression in the above mentioned tissues and in particular the differential expression between diseased tissue esophagus tumor and healthy tissue esophagus, between diseased tissue stomach tumor and healthy tissue stomach, between diseased tissue ovary tumor and healthy tissue ovary, between diseased tissue breast tumor and healthy tissue breast, between diseased tissue kidney tumor and healthy tissue kidney demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of cancer. Additionally the activity of the human XPNPEP2 can be modulated to treat cancer.
Disorders Related to Pulmology
Asthma is thought to arise as a result of interactions between multiple genetic and environmental factors and is characterized by three major features: 1) intermittent and reversible airway obstruction caused by bronchoconstriction, increased mucus production, and thickening of the walls of the airways that leads to a narrowing of the airways, 2) airway hyperresponsiveness, and 3) airway inflammation. Certain cells are critical to the inflammatory reaction of asthma and they include T cells and antigen presenting cells, B cells that produce IgE, and mast cells, basophils, eosinophils, and other cells that bind IgE. These effector cells accumulate at the site of allergic reaction in the airways and release toxic products that contribute to the acute pathology and eventually to tissue destruction related to the disorder. Other resident c ells, such as smooth muscle cells, lung epithelial cells, mucus-producing cells, and nerve cells may also be abnormal in individuals with asthma and may contribute to its pathology. While the airway obstruction of asthma, presenting clinically as an intermittent wheeze and shortness of breath, is generally the most pressing symptom of the disease requiring immediate treatment, the inflammation and tissue destruction associated with the disease can lead to irreversible changes that eventually make asthma a chronic and disabling disorder requiring long-term management.
Chronic obstructive pulmonary (or airways) disease (COPD) is a condition defined physiologically as airflow obstruction that generally results from a mixture of emphysema and peripheral airway obstruction due to chronic bronchitis [Botstein, 1980]. Emphysema is characterised by destruction of alveolar walls leading to abnormal enlargement of the air spaces of the lung. Chronic bronchitis is defined clinically as the presence of chronic productive cough for three months in each of two successive years. In COPD, airflow obstruction is usually progressive and i s only partially reversible. By far the most important risk factor for development of COPD is cigarette smoking, although the disease does also occur in non-smokers.
The human XPNPEP2 is highly expressed in the following tissues of the respiratory system: fetal lung, lung right upper lobe, trachea. The expression in the above mentioned tissues demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of respiratory diseases. Additionally the activity of the human XPNPEP2 can be modulated to treat those diseases.
Disorders Related to Urology
Genitourinary disorders comprise benign and malign disorders of the organs constituting the genitourinary system of female and male, renal diseases like acute or chronic renal failure, immunologically mediated renal diseases like renal transplant rejection, lupus nephritis, immune complex renal diseases, glomerulopathies, nephritis, toxic nephropathy, obstructive uropathies like benign prostatic hypeφlasia (BPH), neurogenic bladder syndrome, urinary incontinence like urge-, stress-, or overflow incontinence, pelvic pain, and erectile dysfunction. The human XPNPEP2 is highly expressed in the following urological tissues: fetal kidney, kidney. The expression in the above mentioned tissues demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of urological disorders. Additionally the activity of the human XPNPEP2 can be modulated to treat urological disorders.
Metabolic Disorders
Metabolic diseases are defined as conditions which result from an abnormality in any of the chemical or biochemical transformations and their regulating systems essential to producing energy, to regenerating cellular constituents, to eliminating unneeded products arising from these processes, and to regulate and maintain homeostasis in a mammal regardless of whether acquired or the result of a genetic transformation.
Depending on which metabolic pathway is involved, a single defective trans- formation or disturbance of its regulation may produce consequences that are narrow, involving a single body function, or broad, affecting many organs, organ-systems or the body as a whole. Diseases resulting from abnormalities related to the fine and coarse mechanisms that affect each individual transformation, its rate and direction or the availability of substrates like amino acids, fatty acids, carbohydrates, minerals, cofactors, hormones, regardless whether they are inborn or acquired, are well within the scope of the definition of a metabolic disease according to this application.
Metabolic diseases often are caused by single defects in particular biochemical pathways, defects that are due to the deficient activity of individual enzymes or molecular receptors leading to the regulation of such enzymes. Hence in a broader sense disturbances of the underlying genes, their products and their regulation lie well within the scope of this definition of a metabolic disease. For example, but not limited to, metabolic diseases may affect 1) biochemical processes and tissues ubiquitous all over the body, 2) the bone, 3) the nervous system, 4) the endocrine system, 5) the muscle including the heart, 6) the skin and nervous tissue, 7) the urogenital system, 8) the homeostasis o f body systems like water and electrolytes. For example, but not limited to, metabolic diseases according to 1) comprise obesity, amyloidosis, disturbances of the amino acid metabolism like branched chain disease, hyperaminoacidemia, hyperaminoaciduria, disturbances of the metabolism of urea, hyperammonemia, mucopolysaccliaridoses e.g. Maroteaux-Lamy syndrom, storage diseases like glycogen storage diseases and lipid storage diseases, glycogenosis diseases like Cori's disease, malabsoφtion diseases like intestinal carbohydrate malabsoφtion, oligosaccharidase deficiency like maltase-, lactase-, sucrase- insufficiency, disorders of the metabolism of fructose, disorders of the metabolism of galactose, galactosaemia, disturbances of carbohydrate utilization like diabetes, hypoglycemia, disturbances of pyruvate metabolism, hypolipidemia, hypolipo- proteinemia, hyperlipidemia, hyperlipoproteinemia, carnitine or carnitine acyl- transferase deficiency, disturbances of the poφhyrin metabolism, poφhyrias, disturbances of the purine metabolism, lysosomal diseases, metabolic diseases of nerves and nervous systems like gangliosidoses, sphingolipidoses, sulfatidoses, leucodystrophies, Lesch-Nyhan syndrome. For example, but not limited to, metabolic diseases according to 2) comprise osteoporosis, osteomalacia like osteoporosis, osteopenia, osteogenesis imperfecta, osteopefrosis, osteonecrosis, Paget's disease of bone, hypophosphatemia. For example, but not limited to, metabolic diseases according to 3) comprise cerebellar dysfunction, disturbances of brain metabolism like dementia, Alzheimer's disease, Huntington's chorea, Parkinson's disease, Pick's disease, toxic encephalopathy, demyelinating neuropathies like inflammatory neuropathy, Guillain-Barre syndrome. For example, but not limited to, metabolic diseases according to 4) comprise primary and secondary metabolic disorders associated with hormonal defects like any disorder stemming from either an hyperfunction or hypofunction of some hormone-secreting endocrine gland and any combination thereof. They comprise Sipple's syndrome, pituitary gland dysfunction and its effects on other endocrine glands, such as the thyroid, adrenals, ovaries, and testes, acromegaly, hyper- and hypothyroidism, euthyroid goiter, euthyroid sick syndrome, thyroiditis, and thyroid cancer, over- or undeφroduction of the adrenal steroid hormones, adrenogenital syndrome, Cushing's syndrome, Addison's disease of the adrenal cortex, Addison's pernicious anemia, primary and secondary aldosteronism, diabetes insipidus, carcinoid syndrome, disturbances caused by the dysfunction of the parathyroid glands, pancreatic islet cell dysfunction, diabetes, disturbances of the endocrine system of the female like estrogen deficiency, resistant ovary syndrome. For example, but not limited to, metabolic diseases according to 5) comprise muscle weakness, myotonia, Duchenne's and other muscular dystrophies, dystrophia myotonica of Steinert, mitochondrial myopathies like disturbances of the catabolic metabolism in the muscle, carbohydrate and lipid storage myopathies, glycogenoses, myoglobinuria, malignant hyperthermia, polymyalgia rheumatica, dermatomyositis, primary myocardial disease, cardiomyopathy. For example, but not limited t o, m etabolic diseases according to 6) comprise disorders of the ectoderm, neurofibromatosis, scleroderma and polyarteritis, Louis-Bar syndrome, von Hippel- Lindau disease, Sturge- Weber syndrome, tuberous sclerosis, amyloidosis, poφhyria. For example, but not limited to, metabolic diseases according to 7) comprise sexual dysfunction of the male and female. For example, but not limited to, metabolic diseases according to 8) comprise confused states and seizures due to inappropriate secretion of antidiuretic hormone from the pituitary gland, Liddle's syndrome, Bartter's syndrome, Fanconi's syndrome, renal electrolyte wasting, diabetes insipidus.
The human XPNPEP2 is highly expressed in the following metabolic disease related tissues: liver liver cirrhosis and adipose. The expression in the above mentioned tissues and in particular the differential expression between diseased tissue liver liver cirrhosis and healthy tissue liver demonstrates that the human XPNPEP2 or mRNA can be utilized to diagnose of metabolic diseases. Additionally the activity of the human XPNPEP2 can be modulated to treat metabolic diseases.
Applications
The present invention provides for both prophylactic and therapeutic methods for cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders. The regulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of XPNPEP2. An agent that modulates activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring cognate ligand of the polypeptide, a peptide, a peptidomimetic, or any small molecule. In one embodiment, the agent stimulates one or more of the biological activities of XPNPEP2. Examples of such stimulatory agents include the active XPNPEP2 and nucleic acid molecules encoding a portion of XPNPEP2. In another embodiment, the agent i nhibits o ne o r m ore o f t he b iological a ctivities o f XPNPEP2. Examples of such inhibitory agents include antisense nucleic acid molecules and antibodies. These regulatory methods can be performed in vitro (e.g., by culturing the cell with the agent) or, alternatively, in vivo (e.g, by administering the agent to a subject). As such, the present invention provides methods of treating an individual afflicted with a disease or disorder characterized by unwanted expression or activity of XPNPEP2 or a protein in the XPNPEP2 signaling pathway. hi one embodiment, the method involves administering an agent like any agent identified or being identifiable by a screening assay as described herein, or combination of such agents that modulate say upregulate or downregulate the expression or activity of XPNPEP2 or of any protein in the XPNPEP2 signaling pathway. In another embodiment, the method involves administering a regulator of
XPNPEP2 as therapy to compensate for reduced or undesirably low expression or activity of XPNPEP2 or a protein in the XPNPEP2 signaling pathway.
Stimulation of activity or expression of XPNPEP2 is desirable in situations in which enzymatic activity or expression is abnormally low and in which increased activity is likely to have a beneficial effect. Conversely, inhibition of enzymatic activity or expression of XPNPEP2 is desirable in situations in which activity or expression of XPNPEP2 is abnormally high and in which decreasing its activity is likely to have a beneficial effect. This invention is further illustrated by the following examples which should not be construed as limiting. The contents of all references, patents and published patent applications cited throughout this application are hereby incoφorated by reference.
Pharmaceutical Compositions
This invention further pertains to novel agents identified by the above-described screening assays and uses thereof for treatments as described herein.
The nucleic acid molecules, polypeptides, and antibodies (also referred to herein as
"active compounds") of the invention can be incoφorated into pharmaceutical compositions suitable for administration. Such compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier. As used herein the language "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absoφtion delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incoφorated into the compositions.
The invention includes pharmaceutical compositions comprising a regulator of XPNPEP2 expression or activity (and/or a regulator of the activity or expression of a protein in the XPNPEP2 signaling pathway) as well as methods for preparing such compositions by combining one or more such regulators and a pharmaceutically acceptable carrier. Also within the invention are pharmaceutical compositions comprising a regulator identified using the screening assays of the invention packaged with instructions for use. For regulators that are antagonists of XPNPEP2 activity or which reduce XPNPEP2 expression, the instructions would specify use of the pharmaceutical composition for treatment of hematological and cardiovascular diseases, disorders of the peripheral and central nervous system, COPD, asthma, genito-urological disorders and inflammation diseases. For regulators that are agonists of XPNPEP2 activity or increase XPNPEP2 expression, the instructions would specify use of the pharmaceutical composition for treatment of hematological and cardiovascular diseases, disorders-of the peripheral and central nervous system,
COPD, asthma, genito-urological disorders and 'inflammation diseases.
An inhibitor of XPNPEP2 may be produced using methods which are generally known in the art. In particular, purified XPNPEP2 may be used to produce antibodies or to screen libraries of pharmaceutical agents to identify those which specifically bind XPNPEP2. Antibodies to XPNPEP2 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, single chain antibodies, Fab fragments, and fragments produced by a Fab expression library. N eutralizing antibodies like those which inhibit dimer formation are especially preferred for therapeutic use.
In another embodiment of the invention, the polynucleotides encoding XPNPEP2, or any fragment or complement thereof, may be used for therapeutic puφoses. In one aspect, the complement of the polynucleotide encoding XPNPEP2 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 XPNPEP2. Thus, complementary molecules or fragments may be used to modulate XPNPEP2 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 XPNPEP2.
Expression vectors derived from retroviruses, adenoviruses, or heφes 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 s equence c omplementary t o t he p olynucleotides o f t he g ene e ncoding XPNPEP2. These techniques are described, for example, in [Scott and Smith (1990)].
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, c ats, c ows, horses, rabbits, monkeys, and most preferably, humans.
An additional embodiment of the invention relates to the administration of a pharmaceutical composition containing XPNPEP2 in conjunction with a pharmaceutically acceptable carrier, for any of the therapeutic effects discussed above. Such pharmaceutical compositions may consist of XPNPEP2, antibodies to XPNPEP2, and mimetics, agonists, antagonists, or inhibitors of XPNPEP2. 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.
A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic s olvents; antibacterial a gents s uch a s b enzyl a lcohol o r m ethyl p arabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. For intravenous administration, suitable carriers include physiological saline, bacterio- static water, Cremophor EM™ (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, a pharmaceutically acceptable polyol like glycerol, propylene glycol, liquid polyetheylene glycol, and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition. Prolonged absoφtion of the injectable compositions can be brought about by including in the composition an agent which delays absoφtion, for example, aluminum monostearate and gelatin. Sterile injectable solutions can be prepared by incoφorating the active compound (e.g., a polypeptide or antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incoφorating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the puφose of oral therapeutic administration, the active compound can be incoφorated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed.
Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any o f the following i ngredients, or e ompounds o f a s imilar n ature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
For administration by inhalation, the compounds are delivered in the form of an aerosol spray from a pressurized container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the a rt, a nd i nclude, f or e xample, f or t ransmucosal a dministration, d etergents, b ile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art. The compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid.
Methods for preparation of such formulations will be apparent to those skilled in the art. T he m aterials c an a lso b e o btained c ommercially from A lza C oφoration a nd Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. 4,522,811.
It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound a nd t he p articular t herapeutic e ffect t o b e a chieved, a nd t he 1 imitations inherent in the art of compounding such an active compound for the treatment of individuals.
The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration. For pharmaceutical compositions which include an antagonist of XPNPEP2 activity, a compound which reduces expression of XPNPEP2, or a compound which reduces expression or activity of a protein in the XPNPEP2 signaling pathway or any combination thereof, the instructions for administration will specify use of the composition for hematological and cardiovascular diseases, disorders of the peripheral and central nervous system, COPD, asthma, genito-urological disorders and inflammation diseases. For pharmaceutical compositions which include an agonist of XPNPEP2 activity, a compound which increases expression of XPNPEP2, or a compound which increases expression or activity of a protein in the XPNPEP2 signaling pathway or any combination thereof, the instructions for administration will s pecify use o f the composition for cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders.
Diagnostics
hi another embodiment, antibodies which specifically bind XPNPEP2 may be used for the diagnosis of disorders characterized by the expression of XPNPEP2, or i n assays to monitor patients being treated with XPNPEP2 or agonists, antagonists, and inhibitors of XPNPEP2. Antibodies useful for diagnostic puφoses may be prepared in the same manner as those described above for therapeutics. Diagnostic assays for
XPNPEP2 include methods which utilize the antibody and a label to detect XPNPEP2 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.
A variety of protocols for measuring XPNPEP2, including ELISAs, RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of XPNPEP2 expression. Normal or standard values for XPNPEP2 expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, preferably human, with antibody to XPNPEP2 under conditions suitable for complex formation. The amount of standard complex formation may be quantified by various methods, preferably by photometric means. Quantities of XPNPEP2 expressed in subject samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease.
In another embodiment of the invention, the polynucleotides encoding XPNPEP2 may be used for diagnostic puφoses. The polynucleotides which may be used include o ligonucleotide s equences, 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 XPNPEP2 may be conelated with disease. The diagnostic assay may be used to distinguish between absence, presence, and excess expression of XPNPEP2, and to monitor regulation of XPNPEP2 levels during therapeutic intervention.
Polynucleotide sequences encoding XPNPEP2 may be used for the diagnosis of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders associated with expression o f XPNPEP2. T he p olynucleotide sequences encoding XPNPEP2 may be used in Southern, Northern, or dot-blot analysis, o r o ther m embrane-based t echnologies; i n P CR t echnologies; i n d ipstick, pin, and ELISA assays; and in microarrays utilizing fluids or tissues from patient biopsies to detect altered XPNPEP2 expression. Such qualitative or quantitative methods are well known in the art.
In a particular aspect, the nucleotide sequences encoding XPNPEP2 may be useful in assays that detect the presence of associated disorders, particularly those mentioned above. The nucleotide sequences encoding XPNPEP2 may be labelled 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 XPNPEP2 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.
In order to provide a basis for the diagnosis of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders associated with expression of XPNPEP2, 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 XPNPEP2, 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 nonnal 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.
Determination of a Therapeutically Effective Dose
The determination of a therapeutically effective dose is well within the capability of those skilled in the art. A therapeutically effective dose refers to that amount of active ingredient which increases or decreases XPNPEP2 activity relative to XPNPEP2 activity which occurs in the absence of the therapeutically effective dose. For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rabbits, dogs, or pigs. The animal model also can 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.
Therapeutic efficacy and toxicity, e.g., ED50 (the dose therapeutically effective in 50% of the population) and LD50 (the dose lethal to 50% of the population), can be determined b y standard pharmaceutical procedures in cell cultures or experimental animals. The dose ratio of toxic to therapeutic effects is the therapeutic index, and it can be expressed as the ratio, LD5o/ED5o. 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, sensitivity of the patient, and the route of administration. The exact dosage will be determined by the practitioner, in light of factors related to the subject that requires treatment. Dosage and administration are adjusted to provide sufficient levels of the active ingredient or to maintain the desired effect. Factors which can be taken into account include the severity of the disease state, general health of the subject, 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 can 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.
Normal dosage amounts can vary from 0.1 micrograms to 100,000 micrograms, up to a total dose of about 1 g, 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. If the reagent is a single-chain antibody, polynucleotides encoding the antibody can be constructed and introduced into a cell either ex vivo or in vivo using well-established techniques including, but not limited to, transferrin-polycation- mediated DNA transfer, transfection with naked or encapsulated nucleic acids, liposome-mediated cellular fusion, intracellular transportation of DNA-coated latex beads, protoplast fusion, viral infection, electroporation, "gene gun", and DEAE- or calcium phosphate-mediated transfection.
If the expression product is mRNA, the reagent is preferably an antisense oligonucleotide or a ribozyme. Polynucleotides which express antisense oligonucleotides or ribozymes can be introduced into cells by a variety of methods, as described above. Preferably, a reagent reduces expression of XPNPEP2 gene or the activity of
XPNPEP2 by at least about 10, preferably about 50, more preferably about 75, 90, or 100% relative t o t he a bsence o f t he r eagent. T he e ffectiveness o f t he m echanism chosen to decrease the level of expression of XPNPEP2 gene or the activity of XPNPEP2 can be assessed using methods well known in the art, such as hybridization of nucleotide probes to XPNPEP2-specific mRNA, quantitative RT-
PCR, immunologic detection of XPNPEP2, or measurement of XPNPEP2 activity.
In any of the embodiments described above, any of the pharmaceutical compositions of the invention can be administered in combination with other appropriate therapeutic agents. Selection of the appropriate agents for use in combination therapy can be made by one of ordinary skill in the art, according to conventional pharmaceutical principles. The combination of therapeutic agents can 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. Any of the therapeutic methods described above can 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. Nucleic acid molecules of the invention are those nucleic acid molecules which are contained in a group of nucleic acid molecules consisting of (i) nucleic acid molecules encoding a polypeptide comprising the amino acid sequence of SEQ ID NO: 2, (ii) nucleic acid molecules comprising the sequence of SEQ ID NO: 1, (iii) nucleic acid molecules having the sequence of SEQ ID NO: 1, (iv)nucleic acid molecules the complementary strand of which hybridizes under stringent conditions to a nucleic acid molecule of (i), (ii), or (iii); and (v) nucleic acid molecules the sequence of which differs from the sequence of a nucleic acid molecule of (iii) due to the degeneracy of the genetic code, wherein the polypeptide encoded by said nucleic acid molecule has XPNPEP2 activity.
Polypeptides of the invention are those polypeptides which are contained in a group of polypeptides consisting of (i) polypeptides having the sequence of SEQ ID NO: 2, (ii) polypeptides comprising the sequence of SEQ ID NO: 2, (iii) polypeptides encoded by nucleic acid molecules of the invention and (iv) polypeptides which show at least 99%, 98%, 95%, 90%, or 80% homology with a polypeptide of (i), (ii), or (iii), wherein said purified polypeptide has XPNPEP2 activity.
An object of the invention is a method of screening for therapeutic agents useful in the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) contacting a test compound with a XPNPEP2 polypeptide, (ii) detect binding of said test compound to said XPNPEP2 polypeptide. E.g., compounds that bind to the XPNPEP2 polypeptide are identified potential therapeutic agents for such a disease.
Another object of the invention is a method of screening for therapeutic agents useful in the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) determining the activity of a XPNPEP2 polypeptide at a certain concentration of a test compound or in the absence of said test compound, (ii) determining the activity of said polypeptide at a different concentration of said test compound. E.g., compounds that lead to a difference in the activity of the XPNPEP2 polypeptide in (i) and (ii) are identified potential therapeutic agents for such a disease.
Another object of the invention is a method of screening for therapeutic agents useful in the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) determining the activity of a XPNPEP2 polypeptide at a certain concentration of a test compound, (ii) determining the activity of a XPNPEP2 polypeptide at the presence of a compound known to be a regulator of a XPNPEP2 p olypeptide. E .g., compounds that show similar effects on the activity of the XPNPEP2 polypeptide in (i) as compared to compounds used in (ii) are identified potential therapeutic agents for such a disease.
Other objects of the invention are methods of the above, wherein the step of contacting is in or at the surface of a cell.
Other objects of the invention are methods of the above, wherein the cell is in vitro.
Other objects of the invention are methods of the above, wherein the step of contacting is in a cell-free system.
Other objects of the invention are methods of the above, wherein the polypeptide is coupled to a detectable label.
Other objects of the invention are methods of the above, wherein the compound is coupled to a detectable label. Other objects of the invention are methods of the above, wherein the test compound displaces a ligand which is first bound to the polypeptide.
Other objects of the invention are methods of the above, wherein the polypeptide is attached to a solid support.
Other objects of the invention are methods of the above, wherein the compound is attached to a solid support.
Another object of the invention is a method of screening for therapeutic agents useful in the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) contacting a test compound with a XPNPEP2 polynucleotide, (ii) detect binding of said test compound to said XPNPEP2 polynucleotide. Compounds that, e.g., bind to the XPNPEP2 polynucleotide are potential therapeutic agents for the treatment of such diseases.
Another object of the invention is the method of the above, wherein the nucleic acid molecule is RNA.
Another object of the invention is a method of the above, wherein the contacting step is in or at the surface of a cell.
Another object of the invention is a method of the above, wherein the contacting step is in a cell-free system.
Another object of the invention is a method of the above, wherein the polynucleotide is coupled to a detectable label. Another object of the invention is a method of the above, wherein the test compound is coupled to a detectable label.
Another object of the invention is a method of diagnosing a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) determining the amount of a XPNPEP2 polynucleotide in a sample taken from said mammal, (ii) determining the amount of XPNPEP2 polynucleotide in healthy and or diseased mammal. A disease is diagnosed, e.g., if there is a substantial similarity in the amount of XPNPEP2 polynucleotide in said test mammal as compared to a diseased mammal.
Another object of the invention is a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a therapeutic agent which binds to a XPNPEP2 polypeptide.
Another object of the invention is a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a therapeutic agent which regulates the activity of a
XPNPEP2 polypeptide.
Another object of the invention is a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a therapeutic agent which regulates the activity of a XPNPEP2 polypeptide, wherein said therapeutic agent is (i) a small molecule, (ii) an RNA molecule, (iii) an antisense oligonucleotide, (iv) a polypeptide, (v) an antibody, or (vi) a ribozyme.
Another object of the invention is a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a XPNPEP2 polynucleotide.
Another object of the invention is a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a XPNPEP2 polypeptide.
Another object of the invention is the use of regulators of a XPNPEP2 for the preparation of a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal.
Another object of the invention is a method for the preparation of a pharmaceutical composition useful for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of (i) identifying a regulator of XPNPEP2, (ii) determining whether said regulator ameliorates the symptoms of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal; and (iii) combining of said regulator with an acceptable pharmaceutical carrier.
Another object of the invention is the use of a regulator of XPNPEP2 for the regulation of XPNPEP2 activity in a mammal having a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders.
The examples below are provided to illustrate the subject invention. These examples are provided by way of illustration and are not included for the puφose of limiting the invention.
Examples
Example 1: Search for homologous sequences in public sequence data bases
The degree of homology can readily be calculated by known methods. Preferred methods to determine homology are designed to give the largest match between the sequences tested. Methods to determine homology are codified in publicly available computer programs such as BestFit, BLASTP, BLASTN, and FASTA. The BLAST programs are publicly available from NCBI and other sources in the internet.
For XPNPEP2 the following hits to known sequences were identified by using the BLAST algorithm [Altschul SF, Madden TL, Schaffer AA, Zhang J, Zhang Z, Miller W, Lipman DJ; Nucleic Acids Res 1997 Sep 1; 25(17): 3389-402] and the following set of parameters: matrix = BLOSUM62 and low complexity filter. The following databases were searched: NCBI (non-redundant database) and DERWENT patent database (Geneseq).
The following hits were found:
>ref]NP_003390.2| X-prolyl aminopeptidase 2, membrane-bound; X-prolyl aminopeptidase 2 (aminopeptidase P); aminoacylproline aminopeptidase; aminopeptidase P [Homo sapiens] gb|AAB96394.2| aminopeptidase P [Homo sapiens] Length = 674 Score = 1380 bits (3572), Expect = 0.0 Identities = 674/674 (100%), Positives = 674/674 (100%)
>sp|O43895|XPP2_HUMAN Xaa-Pro aminopeptidase 2 precursor (X-Pro aminopeptidase 2) (Membrane-bound aminopeptidase P) (Membrane-bound APP) (Membrane-bound AmP) (rnAmP) (Aminoacylproline aminopeptidase) gb|AAG28480.1j membrane-bound aminopeptidase P [Homo sapiens] Length = 674 Score = 1378 bits (3566), Expect = 0.0 Identities = 672/674 (99%), Positives =
674/674 (99%) >gb|AAM60421.1| Sequence 2 from patent US 6399349 Length = 673 Score = 1362 bits (3526), Expect = 0.01 dentities = 669/674 (99%), Positives = 669/674 (99%), Gaps = 1/674 (0%)
>sp|Q95333|XPP2_PIG Xaa-Pro aminopeptidase 2 precursor (X-Pro aminopeptidase 2) (Membrane-bound aminopeptidase P) (Membrane-bound APP) (Membrane-bound A P) (mAmP) (Aminoacylproline aminopeptidase) pir||S72440 X-Pro aminopeptidase (EC 3.4.11.9) precursor - pig gb|AAC48664.1| aminopeptidase P Length = 673 Score = 1177 bits (3044), Expect = 0.0 Identities = 570/674 (84%),
Positives = 615/674 (90%), Gaps = 1/674 (0%)
>refjNP_476496.1| X-prolyl aminopeptidase (aminopeptidase P) 2, membrane-bound [Rattus norvegicus] gb|AAK30297.1|AF359355_l membrane-bound aminopeptidase P [Rattus norvegicus] Length = 674 Score = 1147 bits (2966), Expect = 0.0 Identities
= 548/673 (81%), Positives = 604/673 (89%)
>gb|AAK52065.1|AF367247_l membrane-bound aminopeptidase P [Mus musculus] Length = 674 Score = 1144 bits (2959), Expect = 0.0 Identities = 546/673 (81%), Positives = 602/673 (89%)
>ref]NP_573476.1| X-prolyl aminopeptidase (aminopeptidase P) 2, membrane- bound; membrane-bound aminopeptidase P [Mus musculus] gb|AAL26562.1|AF428102_l membrane bound aminopeptidase P [Mus musculus] Length = 674 Score = 1144 bits (2959), Expect = 0.0 Identities = 547/673 (81%),
Positives = 602/673 (89%)
>ref]NP_835175.1| X-prolyl aminopeptidase (aminopeptidase P) 2, membrane- bound; membrane-bound aminopeptidase P [Mus musculus] dbj|BAC37415.1| unnamed protein product [Mus musculus] Length = 673 Score = 1131 bits (2925),
Expect = 0.0 Identities = 540/663 (81%), Positives = 595/663 (89%) >gb|AAB34314.1| aminopeptidase P, AP-P [swine, kidney cortex, Peptide, 624 aa] Length = 624 Score = 1102 bits (2851), Expect = 0.0 Identities = 534/620 (86%), Positives = 574/620 (92%)
>dbj|BAC39947.1| unnamed protein product [Mus musculus] Length = 582 Score = 989 bits (2558), Expect = 0.0 Identities = 471/580 (81%), Positives = 522/580 (89%)
>ref NP_065116.2] X-prolyl aminopeptidase (aminopeptidase P) 1, soluble; X-prolyl aminopeptidase (aminopeptidase P) 1; X-prolyl aminopeptidase (aminopeptidase P)- like [Homo sapiens] gb|AAF75795.1|AF272981_l cytosolic aminopeptidase P [Homo sapiens] gb|AAH05126.1|AAH05126 X-prolyl aminopeptidase (aminopeptidase P) 1, soluble [Homo sapiens] gb|AAH07579.1|AAH07579 X-prolyl aminopeptidase (aminopeptidase P) 1, soluble [Homo sapiens] Length = 623 Score = 491 bits (1263), Expect = e-137 Identities = 261/620 (42%), Positives = 384/620
(61%), Gaps = 25/620 (4%)
Example 2: Expression profiling
Total cellular RNA was isolated from cells by one of two standard methods: 1) guanidine isothiocyanate/Cesium chloride density gradient centrifugation [Kellogg, (1990)]; or with the Tri-Reagent protocol according to the manufacturer's specifications (Molecular Research Center, Inc., Cincinatti, Ohio). Total RNA prepared by the Tri-reagent protocol was treated with DNAse I to remove genomic DNA contamination.
For relative quantitation o f the mRNA distribution o f XPNPEP2, total RNA from each cell or tissue source was first reverse transcribed. 85 μg of total RNA was reverse transcribed using 1 μmole random hexamer primers, 0.5 mM each of dATP, dCTP, dGTP and dTTP (Qiagen, Hilden, Germany), 3000 U RnaseQut (Invitrogen,
Groningen, Netherlands) in a final volume of 680 μl. The first strand synthesis buffer and Omniscript reverse transcriptase (2 u/μl) were from (Qiagen, Hilden, Germany). The reaction was incubated at 37°C for 90 minutes and cooled on ice. The v olume w as a djusted t o 6800 μ 1 w ith w ater, yielding a final c oncenfration of 12.5 ng μl of starting RNA.
For relative quantitation of the distribution of XPNPEP2 mRNA in cells and tissues the Perkin Elmer ABI Prism RTM. 7700 Sequence Detection system or Biorad iCycler was used according to the manufacturer's specifications and protocols. PCR reactions were set up to quantitate XPNPEP2 and the housekeeping genes HPRT (hypoxanthine phosphoribosyltransferase), GAPDH (glyceraldehyde-3 -phosphate dehydrogenase), β-actin, and others. Forward and reverse primers and probes for XPNPEP2 were designed using the Perkin Elmer ABI Primer Express™ software and were synthesized by TibMolBiol (Berlin, Germany). The XPNPEP2 forward primer sequence was: Primerl (SEQ ID NO: 3). The XPNPEP2 reverse primer sequence was Primer2 (SEQ ID NO: 4). Probel (SEQ ID NO: 5), labelled with FAM
(carboxyfluorescein succinimidyl ester) as the reporter dye and TAMRA (carboxytetramethylrhodamine) as the quencher, is used as a probe for XPNPEP2. The following reagents were prepared in a total of 25 μl : lx TaqMan buffer A, 5.5 mM MgCl2, 200 nM of dATP, dCTP, dGTP, and dUTP, 0.025 U/μl AmpliTaq Gold™, 0.01 U/ μl A pErase and Probel (SEQ ID NO: 4), XPNPEP2 forward and reverse primers each at 200 nM, 200 nM XPNPEP2 FAM/TAMRA-labelled probe, and 5 μl of template cDNA. Thermal cycling parameters were 2 min at 50°C, followed by 10 min at 95°C, followed by 40 cycles of melting at 95°C for 15 sec and annealing extending at 60°C for 1 min.
Calculation of corrected CT values
The CT (threshold cycle) value is calculated as described in the "Quantitative determination of nucleic acids" section. The CF-value (factor for threshold cycle correction) is calculated as follows : 1. PCR reactions were set up to quantitate the housekeeping genes (HKG) for each cDNA sample.
2. CTHκG-values (threshold cycle for housekeeping gene) were calculated as described in the "Quantitative determination of nucleic acids" section.
3. CTnKG-mean values (CT mean value of all HKG tested on one cDNAs) of all HKG for each cDNA are calculated (n = number of HKG):
CTuKG-n-mean value = (CTHKGI -value + CTHKG2-value +... + CTHKG-I.- value) / n
4. CTpannei mean value (CT mean value of all HKG in all tested cDNAs) =
(CTHKGI -mean value + CTHKG2-mean value +...+ CTuKG-y-mean value) / y
(y = number of cDNAs)
5. CFcDNA-n (correction factor for cDNA n) = CTpanneι-mean value - CTHKG-Π- mean value
6. CTcDNA-n (CT value of the tested gene for the cDNA n) + CFcDNA-n (correction factor for cDNA n) = CTCOr-cDNA-n (corrected CT value for a gene on cDNA n)
Calculation of relative expression
Definition : highest CTcor-cDNA-n ≠ 40 is defined as CTCOr-cDNA [high]
Relative Expression = 2<CTcor-cDNA^ " cxcor-cDNA-n)
Tissues
The expression of XPNPEP2 was investigated in the tissues in table 1. Expression profile
The results of the the mRNA-quantification (expression profiling) is shown in Table 1.
Table 1: Relative expression ofXPNPEP2 in various human tissues.
Tissue Relative
Expression fetal heart 72 heart 209 pericardium 116 heart atrium (right) 286 heart atrium (left) 2557 heart ventricle (left) 161 interventricular septum 46 fetal aorta 50 aorta 22 artery 52 coronary artery 48 vein 7 coronary artery smooth muscle primary cells 34
HUVEC cells 719
skin 1992
adrenal gland 102 thyroid 77 thyroid tumor 2 pancreas 58 pancreas liver cirrhosis 27 esophagus 251 esophagus tumor 54 stomach 1323 stomach tumor 734 colon 212 colon tumor 57 small intestine 41765 ileum 143431 ileum tumor 35 ileum chronic inflammation 0 rectum 474 salivary gland 33 fetal liver 8079 liver 160 liver liver cirrhosis 523 liver tumor 324
HEP G2 cells 676
leukocytes (peripheral blood) 8 Jurkat (T-cells) 6 bone marrow 1 erythrocytes 2 lymphnode 6 thymus 171 thrombocytes 1 bone marrow stromal cells 71 bone marrow CD71+ cells 0 bone marrow CD33+ cells 0 bone marrow CD34+ cells 15 bone marrow CD15+ cells 0 cord blood CD71+ cells 0 spleen 12 spleen liver cirrhosis 302 skeletal muscle 168 adipose 553
fetal brain 20 brain 75
Alzheimer brain 61 cerebellum 3 cerebellum (right) 6 cerebellum (left) 28 cerebral cortex 12
Alzheimer cerebral cortex 0 frontal lobe 9
Alzheimer brain frontal lobe 17 occipital lobe 11 parietal lobe 29 temporal lobe 1193 precentral gyrus 8 postcentral gyrus 3 tonsilla cerebelli 27 vermis cerebelli 12 pons 51 substantia nigra 0 cerebral meninges 4 cerebral peduncles 59 corpus callosum 67 hippocampus 17 thalamus 4 dorsal root ganglia 38 spinal cord 69 neuroblastoma SK-N-MC cells 541 neuroblastoma SH-SY5Y cells 815 neuroblastoma IMR32 cells 9 glial tumor H4 cells 0 glial tumor H4 cells + APP 0 HEK CNS 0
HEK CNS + APP 0 retina 0
fetal lung 613 fetal lung fibroblast IMR-90 cells 12 lung 11 lung right upper lobe 541 lung right mid lobe 0 lung right lower lobe 95 lung tumor 51 lung COPD 37 trachea 1152
cervix 46 testis 32
HeLa cells (cervix tumor) 5 placenta 226 uterus 87 uterus tumor 20 ovary 1105 ovary tumor 10 breast 2180 breast tumor 172 MDA MB 231 cells (breast tumor) 7 mammary gland 781
prostate 63 prostate BPH 167 bladder 247 ureter 42 penis 80 corpus cavernosum 61 fetal kidney 6123 kidney 65536 kidney tumor 49
HEK 293 cells 2
Example 3; Antisense Analysis
Knowledge of the correct, complete cDNA sequence coding for XPNPEP2 enables its use as a tool for antisense technology in the investigation of gene function. Oligonucleotides, cDNA or genomic fragments comprising the antisense strand of a polynucleotide coding for XPNPEP2 are used either in vitro or in vivo to inhibit translation of the mRNA. Such technology is now well known in the art, and antisense molecules can be designed at various locations along the nucleotide sequences. By treatment of cells or whole test animals with such antisense sequences, the gene of interest is effectively turned off. Frequently, the function of the gene is ascertained by observing behavior at the intracellular, cellular, tissue or organismal level (e.g., lethality, loss of differentiated function, changes in morphology, etc.).
In addition to using sequences constructed to interrupt transcription of a particular open reading frame, modifications of gene expression is obtained by designing antisense sequences to intron regions, promoter/enhancer elements, or even to transacting regulatory genes.
Example 4: Expression of XPNPEP2
Expression of XPNPEP2 is accomplished by subcloning the cDNAs into appropriate expression vectors and transfecting the vectors into expression hosts such as, e.g., E. coli. In a particular case, the vector is engineered such that it contains a promoter for β-galactosidase, upstream of the cloning site, followed by sequence containing the amino-terminal Methionine and the subsequent seven residues of β-galactosidase.
Immediately following these eight residues is an engineered bacteriophage promoter useful for artificial priming and transcription and for providing a number of unique endonuclease restriction sites for cloning.
Induction of the isolated, transfected bacterial strain with Isopropyl- β-D-thio- galactopyranoside (IPTG) using standard methods produces a fusion protein corresponding to the first seven residues of β-galactosidase, about 15 residues of "linker", and the peptide encoded within the cDNA. Since cDNA clone inserts are generated by an essentially random process, there is probability of 33% that the included cDNA will lie in the correct reading frame for proper translation. If the cDNA is not in the proper reading frame, it is obtained by deletion or insertion of the appropriate number of bases using well known methods including in vitro mutagenesis, digestion with exonuclease m or mung bean nuclease, or the inclusion of an oligonucleotide linker of appropriate length.
The XPNPEP2 cDNA is shuttled into other vectors known to be useful for expression of proteins in specific hosts. Oligonucleotide primers containing cloning sites as well as a segment of DNA (about 25 bases) sufficient to hybridize to stretches at both ends of the target cDNA is synthesized chemically by standard methods. These primers are then used to amplify the desired gene segment by PCR. The r esulting g ene segment is digested with appropriate restriction enzymes under standard conditions and isolated by gel electrophoresis. Alternately, similar gene segments are produced by digestion of the cDNA with appropriate restriction enzymes. Using appropriate primers, segments of coding sequence from more than one gene are ligated together and cloned in appropriate vectors. It is possible to optimize expression by construction of such chimeric sequences.
Suitable expression hosts for such chimeric molecules include, but are not limited to, mammalian cells such as Chinese Hamster Ovary (CHO) and human 293 cells., insect cells such as Sf9 cells, yeast cells such as Saccharomyces cerevisiae and bacterial cells such as E. coli. For each of these cell systems, a useful expression vector also includes an origin of replication to allow propagation in bacteria, and a selectable marker such as the β-lactamase antibiotic resistance gene to allow plasmid selection in bacteria. In addition, the vector may include a second selectable marker such as the neomycin phosphotransferase gene to allow selection in transfected eukaryotic host cells. Vectors for use in eukaryotic expression hosts require RNA processing elements such as 3' polyadenylation sequences if such are not part of the cDNA of interest.
Additionally, the vector contains promoters or enhancers which increase gene expression. Such promoters are host specific and include MMTV, SN40, and metallothionine promoters for CHO cells; tip, lac, tac and T7 promoters for bacterial hosts; and alpha factor, alcohol oxidase and PGH promoters for yeast. Transcription enhancers, such as the rous sarcoma virus enhancer, are used in mammalian host cells. Once homogeneous cultures of recombinant cells are obtained through standard culture methods, large quantities of recombinantly produced XPΝPEP2 are recovered from the conditioned medium and analyzed using chromatographic methods known in the art. For example, XPNPEP2 can be cloned into the expression vector pcDNA3, as exemplified herein. This product can be used to transform, for example, HEK293 or COS by methodology standard in the art. Specifically, for example, using Lipofectamine (Gibco BRL c atolog no. 1 8324-020) mediated gene transfer.
Example 5; Isolation of Recombinant XPNPEP2
XPNPEP2 is expressed as a chimeric protein with one or more additional polypeptide domains added to facilitate protein purification. Such purification facilitating domains include, but are not limited to, metal chelating peptides such as histidine- tryptophan modules that allow purification on immobilized metals [Appa Rao, 1997] and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp., Seattle, Washington). The inclusion of a cleavable linker sequence such as Factor Xa or enterokinase (Invitrogen, Groningen, The Netherlands) between the purification domain and the XPNPEP2 sequence is useful to facilitate expression ofXPNPEP2.
The following example provides a method for purifying XPNPEP2.
XPNPEP2 is generated using the baculovirus expression system BAC-TO-BAC (GIBCO BRL) based on Autographa calif ornica nuclear polyhedrosis virus (AcNPV) infection of Spodoptera frugiperda insect cells (Sf9 cells).
cDNA encoding proteases cloned into either the donor plasmid pFASTBACl or pFASTBAC-HT which contain a mini-Tn7 transposition element. The recombinant plasmid is transformed into DH10BAC competent cells which contain the parent bacmid bMON14272 (AcNPV infectious DNA) and a helper plasmid. The mini-Tn7 element on the pFASTBAC donor can transpose to the attTn7 attachment site on the bacmid thus introducing the protease gene into the viral genome. Colonies containing recombinant bacmids are identified by disruption of the lacZ gene. The protease/bacmid construct can then be isolated and infected into insect cells (Sf9 cells) resulting in the production of infectious recombinant baculovirus particles and expression of either unfused recombinant enzyme (pFastbacl) or XPNPEP2-His fusion protein (pFastbacHT).
Cells are harvested and extracts prepared 24, 48 and 72 hours after transfection. Expression of XPNPEP2 is confirmed by coomassie staining after sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting onto a PNDF membrane of an unstained SDS-PAGE. The protease-His fusion protein is detected due to the interaction between the Νi-ΝTA HRP conjugate and the His-tag which is fused to XPΝPEP2. Example 6: Production of XPNPEP2 Specific Antibodies
Two approaches are utilized to raise antibodies to XPNPEP2, and each approach is useful for generating either polyclonal or monoclonal antibodies, hi one approach, denatured protein from reverse phase HPLC separation is obtained in quantities up to
75 mg. This denatured protein is used to immunize mice or rabbits using standard protocols; about 100 μg are adequate for immunization of a mouse, while up to 1 mg might be used to immunize a rabbit. For identifying mouse hybridomas, the denatured protein is radioiodinated and used to screen potential murine B-cell hybridomas for those which produce antibody. This procedure requires only small quantities of protein, such that 20 mg is sufficient for labeling and screening of several thousand clones.
In the second approach, the amino acid sequence of an appropriate XPNPEP2 domain, as deduced from translation of the cDNA, is analyzed to determine regions of high antigenicity. Ohgopeptides comprising appropriate hydrophilic regions are synthesized and used in suitable immunization protocols to raise antibodies. The optimal amino acid sequences for immunization are usually at the C-terminus, the N- terminus and those intervening, hydrophilic regions of the polypeptide which are likely to be exposed to the external environment when the protein is in its natural conformation.
Typically, selected peptides, about 15 residues in length, are synthesized using an Applied Biosystems Peptide Synthesizer Model 431 A using frnoc-chemistry and coupled to keyhole limpet hemocyanin (EXH; Sigma, St. Louis, MO) by reaction with M-maleimidobenzoyl-N-hydroxysuccinimide ester, MBS. If necessary, a cysteine is introduced at the N-terminus of the peptide to permit coupling to KLH. Rabbits are immunized with the peptide-KLH complex in complete Freund's adjuvant. The resulting antisera are tested for antipeptide activity by binding the peptide to plastic, blocking with 1% bovine serum albumin, reacting with antisera, - Ill -
washing and reacting with labeled (radioactive or fluorescent), affinity purified, specific goat anti-rabbit IgG.
Hybridomas are prepared and screened using standard techniques. Hybridomas of interest are detected by screening with labeled XPNPEP2 to identify those fusions producing the monoclonal antibody with the desired specificity. In a typical protocol, wells of plates (FAST; Becton-Dickinson, Palo Alto, CA) are coated during incubation with affinity purified, specific rabbit anti-mouse (or suitable antispecies 1 g) antibodies at 10 mg/ml. The coated wells are blocked with 1% bovine serum albumin, (BSA), washed and incubated with supernatants from hybridomas. After washing the wells are incubated with labeled XPNPEP2 at 1 mg/ml. Supernatants with specific antibodies bind more labeled XPNPEP2 than is detectable in the background. Then clones producing specific antibodies are expanded and subjected to two cycles of cloning at limiting dilution. Cloned hybridomas are injected into pristane-treated mice to produce ascites, and monoclonal antibody is purified from mouse ascitic fluid by affinity chromatography on Protein A. Monoclonal antibodies with affinities of at least
108 M"1, preferably 109 to 1010 M"1 or stronger, are typically made by standard procedures.
Example 7: Diagnostic Test Using XPNPEP2 Specific Antibodies
Particular XPNPEP2 antibodies are useful for investigating signal transduction and the diagnosis of infectious or hereditary conditions which are characterized by differences in the amount or distribution of XPNPEP2 or downstream products of an active signaling cascade.
Diagnostic tests for XPNPEP2 include methods utilizing antibody and a label to detect XPNPEP2 in human body fluids, membranes, cells, tissues or extracts of such.
The polypeptides and antibodies of the present invention are used with or without modification. Frequently, the polypeptides and antibodies are labeled by joining them, either covalently or noncovalently, with a substance which provides for a detectable signal. A wide variety of labels and conjugation techniques are known and have been reported extensively in both the scientific and patent literature. Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent agents, chemiluminescent agents, chromogenic agents, magnetic particles and the like.
A variety of protocols for measuring soluble or membrane-bound XPNPEP2, using either polyclonal or monoclonal antibodies specific for the protein, are known in the art. Examples include enzyme-linked immunosorbent assay (ELISA), radioimmuno- assay (RIA) and fluorescent activated cell sorting (FACS). A two-site monoclonal- based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on XPNPEP2 is preferred, but a competitive binding assay may be employed.
Example 8: Purification of Native XPNPEP2 Using Specific Antibodies
Native or recombinant XPNPEP2 is purified by immunoaffinity chromatography using antibodies s pecific for X PNPEP2. I n g eneral, an i mmunoaffinity c olumn i s constructed by covalently coupling the anti-TRH antibody to an activated chromatographic resin.
Polyclonal immunoglobulms are prepared from immune sera either by precipitation with ammonium sulfate or by purification on immobilized Protein A (Pharmacia
LKB Biotechnology, Piscataway NJ.). Likewise, monoclonal antibodies are prepared from mouse ascites fluid by ammonium sulfate precipitation or chromatography on immobilized Protein A. Partially purified immunoglobulin is covalently attached to a chromatographic resin such as CnBr-activated Sepharose (Pharmacia LKB Biotechnology). The antibody is coupled to the resin, the resin is blocked, and the derivative resin is washed according to the manufacturer's instructions.
Such immunoaffinity columns are utilized in the purification of XPNPEP2 by pre- paring a fraction from cells containing XPNPEP2 in a soluble form. This preparation is derived by solubilization of whole cells or of a subcellular fraction obtained via differential centrifugation (with or without addition of detergent) or by other methods well known in the art. Alternatively, soluble XPNPEP2 containing a signal sequence is secreted in useful quantity into the medium in which the cells are grown.
A soluble XPNPEP2-containing preparation is passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of XPNPEP2 (e.g., high ionic strength buffers in the presence of detergent). Then, the column is eluted under conditions that disrupt antibody/protein binding (e.g., a buffer of pH 2-3 or a high concentration of a chaofrope such as urea or thiocyanate ion), and XPNPEP2 is collected.
Example 9: Drug Screening
This invention is particularly useful for screening therapeutic compounds by using
XPNPEP2 or fragments thereof in any of a variety of drug screening techniques.
The following example provides a system for drug screening measuring the protease activity.
The recombinant protease-His fusion protein can be purified from the crude lysate by metal-affinity chromatography using Ni-NTA agarose. This allows the specific retention of the recombinant material (since this is fused to the His-tag) whilst the endogenous insect proteins are washed off. The recombinant material is then eluted by competition with imidazol. The activity of XPNPEP2 molecules of the present invention can be measured using a variety of assays that measure XPNPEP2 activity. For example, XPNPEP2 enzyme activity can be assessed by a standard in vitro serine/metallo/... protease assay (see, for example, [U.S. 5 ,057,414]). Those of skill in the art are aware of a variety of substrates suitable for in vitro assays, such as SucAla-Ala-Pro-Phe-pNA, fluorescein mono-p-guanidinobenzoate hydrochloride, benzyloxycarbonyl-L-Arginyl-S-benzyl- ester, Nalpha-Benzoyl-L-arginine ethyl ester hydrochloride, and the like, i addition, protease assay kits available from commercial sources, such as Calbiochem™ (San Diego, Calif). For general references, see Barrett (Ed.), Methods in Enzymology, Proteolytic Enzymes: Serine and Cysteine Peptidases (Academic Press Inc. 1994), and Barrett et al., (Eds.), Handbook of Proteolytic Enzymes (Academic Press Inc. 1998).
Example 10: Rational Drug Design
The goal of rational drug design is to produce structural analogs of biologically active polypeptides of interest or of small molecules with which they interact, agonists, antagonists, or inhibitors. Any of these examples are used to fashion drugs which are more active or stable forms of the polypeptide or which enhance or interfere with the function of a polypeptide in vivo.
In one approach, the three-dimensional structure of a protein of interest, or of a protein-inhibitor complex, is determined by x-ray crystallography, by computer modeling or, most typically, by a combination of the two approaches. Both the shape and charges of the polypeptide must be ascertained to elucidate the structure and to determine active site(s) of the molecule. Less often, useful information regarding the structure of a polypeptide is gained by modeling based on the structure of homologous proteins. In both cases, relevant structural information is used to design efficient inhibitors. Useful examples of rational drug design include molecules which have improved activity or stability or which act as inhibitors, agonists, or antagonists of native peptides. It is also possible to isolate a target-specific antibody, selected by functional assay, as described above, and then to solve its crystal structure. This approach, in principle, yields a pharmacore upon which subsequent drug design is based. It is possible to bypass protein crystallography altogether by generating anti-idiotypic antibodies
(anti-ids) to a functional, pharmacologically active antibody. As a mirror image of a mirror image, the binding site of the anti-ids is expected to be an analog of the original receptor. The anti-id is then used to identify and isolate peptides from banks of chemically or biologically produced peptides. The isolated peptides then act as the pharmacore.
By virtue of the present invention, sufficient amount of polypeptide are made available to p erform s uch analytical s tudies a s X-ray crystallography. In addition, knowledge of the XPNPEP2 amino acid sequence provided herein provides guidance to those employing computer modeling techniques in place of or in addition to x-ray crystallography.
Example 11: Identification of Other Members of the Signal Transduction Complex
Labeled XPNPEP2 is useful as a reagent for the purification of molecules with which it interacts. In one embodiment of affinity purification, XPNPEP2 is covalently coupled to a chromatography column. Cell-free extract derived from synovial cells or putative target cells i s p assed over the column, and molecules with appropriate affinity bind to XPNPEP2. XPNPEP2-complex is recovered from the column, and the XPNPEP2-binding ligand disassociated and subjected to N-terminal protein sequencing. The amino acid sequence information is then used to identify the captured molecule or to design degenerate oligonucleotide probes for cloning the relevant gene from an appropriate cDNA library. In an alternate method, antibodies are raised against XPNPEP2, specifically monoclonal antibodies. The monoclonal antibodies are screened to identify those which inhibit the binding of labeled XPNPEP2. These monoclonal antibodies are then used therapeutically.
Example 12: Use and Administration of Antibodies, Inhibitors, or Antagonists
Antibodies, inhibitors, or antagonists of XPNPEP2 or other treatments and compunds that are limiters of signal transduction (LSTs), provide different effects when administered therapeutically. LSTs are formulated in a nontoxic, inert, pharmaceutically acceptable aqueous carrier medium preferably at a pH of about 5 to 8, more preferably 6 to 8, although pH may vary according to the characteristics of the antibody, inhibitor, or antagonist being formulated and the condition to be treated. Characteristics of LSTs include solubility of the molecule, its half-life and antigenicity/immunogenicity. These and other characteristics aid in defining an effective carrier. Native human proteins are preferred as LSTs, but organic or synthetic molecules resulting from drug screens are equally effective in particular situations.
LSTs are delivered by known routes of administration including but not limited to topical creams and gels; transmucosal spray and aerosol; transdermal patch and bandage; injectable, intravenous and lavage formulations; and orally administered liquids and p ills p articularly formulated to r esist s tomach a cid a nd e nzymes. T he particular formulation, exact dosage, and route of administration is determined by the attending physician and varies according to each specific situation.
Such determinations are made by considering multiple variables such as the condition to be treated, the LST to be administered, and the pharmacokinetic profile of a particular LST. Additional factors which are taken into account include severity of the disease state, patient's age, weight, gender and diet, time and frequency of LST administration, possible combination with other drugs, reaction sensitivities, and tolerance/response to therapy. Long acting LST formulations might be administered every 3 to 4 days, every week, or once every two weeks depending on half-life and clearance rate of the particular LST.
Normal dosage amounts vary from 0.1 to 105 μg, up to a total dose of about 1 g, depending upon the route of administration. Guidance as to particular dosages and methods o f d elivery i s p rovided i n t he 1 iterature; s ee U .S. P at. Nos. 4,657,760; 5,206,344; or 5,225,212. Those skilled in the art employ different formulations for different LSTs. Administration to cells such as nerve cells necessitates delivery in a manner different from that to other cells such as vascular endothelial cells.
It is contemplated that abnormal signal transduction, trauma, or diseases which trigger XPNPEP2 activity are treatable with LSTs. These conditions or diseases are specifically diagnosed by the tests discussed above, and such testing should be performed in suspected cases of viral, bacterial or fungal infections, allergic responses, mechanical injury associated with trauma, hereditary diseases, lymphoma or carcinoma, or other conditions which activate the genes of lymphoid or neuronal tissues.
Example 13: Production of Non-human Transgenic Animals
Animal model systems which elucidate the physiological and behavioral roles of the XPNPEP2 are produced by creating nonhuman transgenic animals in which the activity of the XPNPEP2 is either increased or decreased, or the amino acid sequence of the expressed XPNPEP2 is altered, by a variety of techniques. Examples of these techniques include, but are not limited to: 1) Insertion of normal or mutant versions of DNA encoding a XPNPEP2, by microinj ection, electroporation, retro viral transfection or other means well known to those skilled in the art, into appropriately fertilized embryos in order to produce a transgenic animal or 2) homologous recombination of mutant or normal, human or animal versions of these genes with the native gene locus in transgenic animals to alter the regulation of expression or the structure of these XPNPEP2 sequences. The technique of homologous recombination is well known in the art. It replaces the native gene with the inserted gene and hence is useful for producing an animal that cannot express native XPNPEP2s but does express, for example, an inserted mutant XPNPEP2, which has replaced the native XPNPEP2 in the animal's genome by recombination, resulting in underexpression o f the transporter. M icroinj ection adds genes to the genome, but does not remove them, and the technique is useful for producing an animal which expresses its own and added XPNPEP2, resulting in overexpression of the XPNPEP2.
One means available for producing a transgenic animal, with a mouse as an example, is as follows: Female mice are mated, and the resulting fertilized eggs are dissected out of their oviducts. The eggs are stored in an appropriate medium such as cesiumchloride M2 medium. DNA or cDNA encoding XPNPEP2 is purified from a vector by methods well known to the one skilled in the art. Inducible promoters may be fused with the coding region of the DNA to provide an experimental means to regulate expression of the transgene. Alternatively or in addition, tissue specific regulatory elements may be fused with the coding region to permit tissue-specific expression of the transgene. The DNA, in an appropriately buffered solution, is put into a microinj ection needle (which may be made from capillary tubing using a piper puller) and the egg to be injected is put in a depression slide. The needle is inserted into the pronucleus of the egg, and the DNA solution is injected. The injected egg is then transferred into the oviduct of a pseudopregnant mouse which is a mouse stimulated by the appropriate hormones in order to maintain false pregnancy, where it proceeds to the uterus, implants, and develops to term. As noted above, microinj ection is not the only method for inserting DNA into the egg but is used here only for exemplary purposes. References
U.S. 4,522,811
U.S. 5,057,414 U.S. 5,283,317
U.S. 5,565,332
U.S. 5,723,323.
U.S. 5,747,334
U.S. 5,783,384 U.S. 5,885,814
U.S. 5,985,629
U.S. 6,399,349
WO 84/03564
WO 93/03151 WO 94/13804
WO 02/61131
Altschul SF, Madden TL, Schaffer AA, Zhang J, Zhang Z, Miller W, Lipman DJ;
Nucleic Acids Res 1997 Sep 1; 25(17): 3389-402
Appa Rao et al., 1997, Protein Expr Purif Nov, 11(2): 201-8 Avalle et al., Ann. N Y Acad.Sci. 864:118 (1998)).
Barnes, 2000, Chest, 117:10S14S
Barrett et al., (Eds.), Handbook of Proteolytic Enzymes (Academic Press Inc. 1998).
Barrett (Ed.), Methods in Enzymology, Proteolytic Enzymes: Serine and Cysteine
Peptidases (Academic Press Inc. 1994) Botstein et al., 1980, Am J Hum Genet. 32: 314-31
Colbere-Garapin et al, 1981, J. Mol. Biol. 150, 1-14
Cunningham and Wells, j. Mol. Biol. 234:554 (1993).
Engelhard et al, 1994, Proc. Nat. Acad. Sci. 91, 3224-3227
Friboulet et al., Appl. Biochem. Biotechnol. 47:229 (1994) Gergen and Weiss , 1992, Am Rev Respir Dis 146:823-824
Gibson et al., 1996, Genome Research 6: 995-1001 Haseloff et al, 1988 , Nature 334, 585-591
Heid et al, 1996, Genome Research 6: 986-994
Holland et al., 1991, PNAS 88: 7276-7280
Jeffreys et al., 1985, Nature 316: 76-9 Johnson et al, 1989, Endoc. Rev. 10, 311-331
Joron et al., Ann. N Y Acad. Sci. 672:216 (1992)
Karlsson, Immunol. Methods 145:229 (1991)
Kellogg et al., 1990, Anal. Biochem. 189:202-208
Lam , 1997, Anticancer Drug Res. 12(3):145-67 Livak et al., 1995 , PCR Methods and Applications 357-362
Logan, Shenk, 1984, Proc. Natl. Acad. Sci. 81, 3655-3659
Lowy et al, 1980, Cell 22, 817-23
Maddox et al, 1983, J. Exp. Med. 158, 1211-1216
Monfardim et al., Proc. Assoc. Am. Physicians 108:420 (1996) McConnell et al, 1992 , Science 257, 1906-1912
Nicholls et al, 1993, J. Immunol. Meth. 7 5, 81-91
Piatak et al., 1993, BioTechniques 14:70-81
Piatak et al., 1993, Science 259:1749-1754
Porath et al, 1992, Prot. Exp. Purif. 3, 263-281 Prueitt et al., Cytogenet. Cell Genet. 89: 44-50, 2000
Roberge et al, 1995, Science 269, 202-204
Scott and Smith (1990) Science 249:386-390
Sjolander, Urbaniczky, 1991, Anal. Chem. (53, 2338-2345
Szabo et α/., 1995, Curr. Opin. Struct. Biol. 5, 699-705 Thomas, 1980, Proc. Nat. Acad. Sci., 77:5201-5205
Uhlmann et al, 1987, Tetrahedron. Lett. 215, 3539-3542
Venema et al., Biochim. Biophys. Acta 1354: 45-48, 1997
Weber et al., 1990, Genomics 7: 524-30
Wigler et al, 1977, Cell 11, 223-32 Wigler et al, 1980, Proc. Natl. Acad. Sci. 77, 3567-70

Claims

Claims
1. A method of screening for therapeutic agents useful in the treatment of a disease comprised in a group of diseases consisting of cardiovascular
> disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of
i) contacting a test compound with a XPNPEP2 polypeptide,
ii) detect binding of said test compound to said XPNPEP2 polypeptide.
2. A method of screening for therapeutic agents useful in the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of
i) determining the activity of a XPNPEP2 polypeptide at a certain concentration of a test compound or in the absence of said test compound,
ii) determining the activity of said polypeptide at a different concentration of said test compound.
3. A method of screening for therapeutic agents useful in the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of i) determining the activity of a XPNPEP2 polypeptide at a certain concentration of a test compound,
ii) determining the activity of a XPNPEP2 polypeptide at the presence of a compound known to be a regulator of a XPNPEP2 polypeptide.
4. The method of any of claims 1 to 3, wherein the step of contacting is in or at the surface of a cell.
5. The method of any of claims 1 to 3, wherein the cell is in vitro.
6. The method of any of claims 1 to 3, wherein the step of contacting is in a cell- free system.
7. The method of any of claims 1 to 3, wherein the polypeptide is coupled to a detectable label.
8. The method of any of claims 1 to 3, wherein the compound is coupled to a detectable label.
9. The method of any of claims 1 to 3, wherein the test compound displaces a ligand which is first bound to the polypeptide.
10. The method of any of claims 1 to 3, wherein the polypeptide is attached to a solid support.
11. The method of any of claims 1 to 3, wherein the compound is attached to a solid support.
12. A method of screening for therapeutic agents useful in the freatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of
i) contacting a test compound with a XPNPEP2 polynucleotide,
ii) detect binding of said test compound to said XPNPEP2 polynucleotide.
13. The method of claim 12 wherein the nucleic acid molecule is RNA.
14. The method of claim 12 wherein the contacting step is in or at the surface of a cell.
15. The method of claim 12 wherein the contacting step is in a cell-free system.
16. The method of claim 12 wherein polynucleotide is coupled to a detectable label.
17. The method of claim 12 wherein the test compound is coupled to a detectable label.
18. A method of diagnosing a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of
i) determining the amount of a XPNPEP2 polynucleotide in a sample taken from said mammal, ii) determining the amount of XPNPEP2 polynucleotide in healthy and/or diseased mammals.
19. A pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a therapeutic agent which binds to a XPNPEP2 polypeptide.
20. A pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a therapeutic agent which regulates the activity of a
XPNPEP2 polypeptide.
21. A pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a therapeutic agent which regulates the activity of a XPNPEP2 polypeptide, wherein said therapeutic agent is
0 a small molecule, ϋ) an RNA molecule, iϋ) an antisense oligonucleotide, iv) a polypeptide, v) an antibody, or vi) a ribozyme.
22. A pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a XPNPEP2 polynucleotide.
23. A pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising a XPNPEP2 polypeptide.
24. Use of regulators of a XPNPEP2 for the preparation of a pharmaceutical composition for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases , and urological disorders in a mammal.
25. Method for the preparation of a pharmaceutical composition useful for the treatment of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal comprising the steps of
i) identifying a regulator of XPNPEP2,
ii) determining whether said regulator ameliorates the symptoms of a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders in a mammal; and
iii) combining of said regulator with an acceptable pharmaceutical carrier.
26. Use of a regulator of XPNPEP2 for the regulation of XPNPEP2 activity in a mammal having a disease comprised in a group of diseases consisting of cardiovascular disorders, dermatological disorders, gastrointestinal and liver diseases, metabolic diseases, cancer disorders, neurological disorders, respiratory diseases and urological disorders.
PCT/EP2004/005021 2003-05-23 2004-05-11 Diagnostics and therapeutics for diseases associated with x-prolyl aminopeptidase 2 (xpnpep2) WO2004104575A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP03011704 2003-05-23
EP03011704.8 2003-05-23

Publications (2)

Publication Number Publication Date
WO2004104575A2 true WO2004104575A2 (en) 2004-12-02
WO2004104575A3 WO2004104575A3 (en) 2005-01-27

Family

ID=33462090

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2004/005021 WO2004104575A2 (en) 2003-05-23 2004-05-11 Diagnostics and therapeutics for diseases associated with x-prolyl aminopeptidase 2 (xpnpep2)

Country Status (1)

Country Link
WO (1) WO2004104575A2 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5656603A (en) * 1995-05-31 1997-08-12 Loyola University Of Chicago Aminopeptidase P inhibitors and uses thereof
WO2001000811A2 (en) * 1999-06-30 2001-01-04 Millennium Pharmaceuticals, Inc. 17867, a novel human aminopeptidase
WO2001089569A1 (en) * 2000-05-23 2001-11-29 Institut Für Medizintechnologie Magdeburg Gmbh Imtm Combinations of enzyme inhibitor-containing preparations and the use thereof
WO2002020736A2 (en) * 2000-09-08 2002-03-14 Incyte Genomics Inc Proteases
US6399349B1 (en) * 1997-09-02 2002-06-04 James W. Ryan Human aminopeptidase P gene
WO2002053170A2 (en) * 2001-01-02 2002-07-11 Institut Für Medizintechnologie Magdeburg Gmbh (Imtm) Combined use of enzyme inhibitors and pharmaceutical preparations thereof for the treatment and prophylaxis of arteriosclerosis, for the treatment and prevention of allergic reactions of type i according to the gell and coombs classification, and for the treatment and prevention of dermatological diseases associated with fo
WO2002099379A2 (en) * 2001-06-01 2002-12-12 The Burnham Institute Breast homing peptides and methods of identifying same using aminopeptidase p

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5656603A (en) * 1995-05-31 1997-08-12 Loyola University Of Chicago Aminopeptidase P inhibitors and uses thereof
US6399349B1 (en) * 1997-09-02 2002-06-04 James W. Ryan Human aminopeptidase P gene
WO2001000811A2 (en) * 1999-06-30 2001-01-04 Millennium Pharmaceuticals, Inc. 17867, a novel human aminopeptidase
WO2001089569A1 (en) * 2000-05-23 2001-11-29 Institut Für Medizintechnologie Magdeburg Gmbh Imtm Combinations of enzyme inhibitor-containing preparations and the use thereof
WO2002020736A2 (en) * 2000-09-08 2002-03-14 Incyte Genomics Inc Proteases
WO2002053170A2 (en) * 2001-01-02 2002-07-11 Institut Für Medizintechnologie Magdeburg Gmbh (Imtm) Combined use of enzyme inhibitors and pharmaceutical preparations thereof for the treatment and prophylaxis of arteriosclerosis, for the treatment and prevention of allergic reactions of type i according to the gell and coombs classification, and for the treatment and prevention of dermatological diseases associated with fo
WO2002099379A2 (en) * 2001-06-01 2002-12-12 The Burnham Institute Breast homing peptides and methods of identifying same using aminopeptidase p

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MARSHALL PETER ET AL: "Study of bradykinin metabolism in human and rat plasma by liquid chromatography with inductively coupled plasma mass spectrometry and orthogonal acceleration time-of-flight mass spectrometry." RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM. 2002, vol. 16, no. 3, 2002, pages 220-228, XP002296819 ISSN: 0951-4198 *

Also Published As

Publication number Publication date
WO2004104575A3 (en) 2005-01-27

Similar Documents

Publication Publication Date Title
EP1631680A2 (en) Diagnostics and therapeutics for diseases associated with dipeptidylpeptidase iv (dpp4)
WO2006010517A2 (en) Diagnostics and therapeutics for diseases associated with fibroblast activation protein (fap)
WO2005114207A2 (en) Diagnostics and therapeutics for diseases associated with aminopeptidase n (anpep)
US20070191296A1 (en) Diagnostics and therapeutics for diseases associated with kallikrein 13 (klk13)
EP1718975A2 (en) Diagnostics and therapeutics for diseases associated with kallikrein 6 (klk6)
US20070196372A1 (en) Diagnostics and therapeutics for diseases associated with kallikrein 8 (klk8)
WO2006013013A2 (en) Diagnostics and therapeutics for diseases associated with protective protein for beta-galactosidase (ppgb)
EP1745293A1 (en) Diagnostics and therapeutics for diseases associated with dipeptidyl-peptidase 9 (dpp9)
US20070099255A1 (en) Diagnostics and therapeutics for diseases associated with n-acetylated alpha-linked acidic dipeptidase 2(naaladase 2)
EP1629282A2 (en) Diagnostics and therapeutics for diseases associated with x-pro dipeptidase (pepd)
WO2004097034A2 (en) Diagnostics and therapeutics for diseases associated with human transmembrane serine protease 4 (tmprss4)
US20070254287A1 (en) Diagnostics and Therapeutics for Diseases Associated with Dipeptidyl-Peptidase 10 (Dpp10)
WO2005106021A1 (en) Diagnostics and therapeutics for diseases associated with dipeptidyl-peptidase 8 (dpp8)
WO2004106536A2 (en) Diagnostics and therapeutics for diseases associated with serine protease 8 (prss8)
WO2006010516A1 (en) Diagnostics and therapeutics for diseases associated with methionine aminopeptidase 2 like protein (metap2-like)
US20070042423A1 (en) Diagnostics and therapeutics for diseases associated with arginyl aminopeptidase rnpep-like (rnpep-like)
US20070166715A1 (en) Diagnostics and therapeutics for diseases associated with aminopeptidase-like 1 (npepl1)
US20070081997A1 (en) Diagnostics and therapeutics for diseases associated with kallikrein 9 (klk9)
WO2004104575A2 (en) Diagnostics and therapeutics for diseases associated with x-prolyl aminopeptidase 2 (xpnpep2)
US20070258974A1 (en) Diagnostics and Therapeutics for Diseases Associated with Puromycin Sensitive Aminopeptidase Npepps (Npepps)
US20070202567A1 (en) Diagnostics And Therapeutics For Diseases Associated With Arginyl Aminopeptidase Rnpep (Rnpep)
WO2004098487A2 (en) Diagnostics and therapeutics for diseases associated with human transmembrane serine protease 5 (tmprss5)
WO2004101814A1 (en) Diagnostics and therapeutics for diseases associated with x-pro dipeptidase like peptidase (pepdlike)
EP1714156A1 (en) Diagnostics and therapeutics for diseases associated with phosphate regulating endopeptidase homolog (phex)
WO2004104218A1 (en) Diagnostics and therapeutics for diseases associated with x-prolyl aminopeptidase (aminopeptidase p) 1 (xpnpep1)

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase