EP1000152A2 - 5' ESTs FOR SECRETED PROTEINS EXPRESSED IN MUSCLE AND OTHER MESODERMAL TISSUES - Google Patents

5' ESTs FOR SECRETED PROTEINS EXPRESSED IN MUSCLE AND OTHER MESODERMAL TISSUES

Info

Publication number
EP1000152A2
EP1000152A2 EP98936596A EP98936596A EP1000152A2 EP 1000152 A2 EP1000152 A2 EP 1000152A2 EP 98936596 A EP98936596 A EP 98936596A EP 98936596 A EP98936596 A EP 98936596A EP 1000152 A2 EP1000152 A2 EP 1000152A2
Authority
EP
European Patent Office
Prior art keywords
sequences
cdna
seq
protein
nos
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP98936596A
Other languages
German (de)
French (fr)
Inventor
Jean-Baptiste Dumas Milne Edwards
Aymeric Duclert
Bruno Lacroix
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck Biodevelopment SAS
Original Assignee
Genset SA
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 Genset SA filed Critical Genset SA
Priority to EP03016881A priority Critical patent/EP1367124A1/en
Publication of EP1000152A2 publication Critical patent/EP1000152A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • C12N15/625DNA sequences coding for fusion proteins containing a sequence coding for a signal sequence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/036Fusion polypeptide containing a localisation/targetting motif targeting to the medium outside of the cell, e.g. type III secretion

Definitions

  • Cloning vectors such as yeast artificial chromosomes (YACs) and bacterial artificial chromosomes (BACs) are able to accept DNA inserts ranging from 300 to 1000 kilobases (kb) or 100-400 kb in length respectively, thereby facilitating the manipulation and ordering of DNA sequences distributed over great distances on the human chromosomes.
  • Automated DNA sequencing machines permit the rapid sequencing of human genes.
  • Bioinformatics software enables the comparison of nucleic acid and protein sequences, thereby assisting in the characterization of human gene products.
  • two different approaches are being pursued for identifying and characterizing the genes distributed along the human genome.
  • large fragments of genomic DNA are isolated, cloned, and sequenced. Potential open reading frames in these genomic sequences are identified using bioinformatics software.
  • bioinformatics software may mischaracterize the genomic sequences obtained.
  • the software may produce false positives in which non- coding DNA is mischaracterized as coding DNA or false negatives in which coding DNA is mislabeled as non-coding DNA.
  • An alternative approach takes a more direct route to identifying and characterizing human genes.
  • complementary DNAs cDNAs
  • mRNAs messenger RNAs
  • sequencing is only performed on DNA which is derived from protein coding portions of the genome. Often, only short stretches of the cDNAs are sequenced to obtain sequences called expressed sequence tags (ESTs). The ESTs may then be used to isolate or purify extended cDNAs which include sequences adjacent to the EST sequences.
  • the extended cDNAs may contain all of the sequence of the EST which was used to obtain them or only a portion of the sequence of the EST which was used to obtain them.
  • the extended cDNAs may contain the full coding sequence of the gene from which the EST was derived or, alternatively, the extended cDNAs may include portions of the coding sequence of the gene from which the EST was derived. It will be appreciated that there may be several extended cDNAs which include the EST sequence as a result of alternate splicing or the activity of alternative promoters.
  • these short EST sequences were often obtained from oligo-dT primed cDNA libraries. Accordingly, they mainly corresponded to the 3' untranslated region of the mRNA. In part, the prevalence of EST sequences derived from the 3' end of the mRNA is a result of the fact that typical techniques for obtaining cDNAs are not well suited for isolating cDNA sequences derived from the 5' ends of mRNAs. (Adams et al., Nature 377:3-174, 1996; Hfflier e/ ar/., Genome Res. 6:807-828, 1996).
  • the reported sequences typically correspond to coding sequences and do not include the full 5' untranslated region of the mRNA from which the cDNA is derived.
  • Such incomplete sequences may not include the first exon of the mRNA, particularly in situations where the first exon is short.
  • they may not include some exons, often short ones, which are located upstream of splicing sites.
  • secretory proteins including tissue plasminogen activator, G-CSF, GM-CSF, erythropoietin, human growth hormone, insulin, interferon- ⁇ , interferon- ⁇ , interferon- ⁇ , and interleukin-2.
  • tissue plasminogen activator G-CSF
  • GM-CSF GM-CSF
  • erythropoietin human growth hormone
  • insulin interferon- ⁇
  • interferon- ⁇ interferon- ⁇
  • interferon- ⁇ interleukin-2
  • interleukin-2 interleukin-2
  • secretory proteins include short peptides, called signal peptides, at their amino termini which direct their secretion.
  • signal peptides are encoded by the signal sequences located at the 5' ends of the coding sequences of genes encoding secreted proteins. Because these signal peptides will direct the extracellular secretion of any protein to which they are operably linked, the signal sequences may be exploited to direct the efficient secretion of any protein by operably linking the signal sequences to a gene encoding the protein for which secretion is desired.
  • portions of signal sequences may also be used to direct the intracellular import of a peptide or protein of interest.
  • signal sequences encoding signal peptides also find application in simplifying protein purification techniques. In such applications, the extracellular secretion of the desired protein greatly facilitates purification by reducing the number of undesired proteins from which the desired protein must be selected. Thus, there exists a need to identify and characterize the 5' portions of the genes for secretory proteins which encode signal peptides.
  • the present 5' ESTs may be used to efficiently identify and isolate upstream regulatory regions which control the location, developmental stage, rate, and quantity of protein synthesis, as well as the stability of the mRNA. (Theil, BioFactors 4:87-93, 1993). Once identified and characterized, these regulatory regions may be utilized in gene therapy or protein purification schemes to obtain the desired amount and locations of protein synthesis or to inhibit, reduce, or prevent the synthesis of undesirable gene products.
  • ESTs containing the 5' ends of secretory protein genes may include sequences useful as probes for chromosome mapping and the identification of individuals. Thus, there is a need to identify and characterize the sequences upstream of the 5' coding sequences of genes encoding secretory proteins.
  • the present invention relates to purified, isolated, or recombinant ESTs which include sequences derived from the authentic 5' ends of their corresponding mRNAs.
  • corresponding mRNA refers to the mRNA which was the template for the cDNA synthesis which produced the 5' EST.
  • sequences will be referred to hereinafter as "5' ESTs.”
  • purified does not require absolute purity; rather, it is intended as a relative definition.
  • Individual 5' EST clones isolated from a cDNA library have been conventionally purified to electrophoretic homogeneity. The sequences obtained from these clones could not be obtained directly either from the library or from total human DNA.
  • the cDNA clones are not naturally occurring as such, but rather are obtained via manipulation of a partially purified naturally occurring substance (messenger RNA).
  • the conversion of mRNA into a cDNA library involves the creation of a synthetic substance (cDNA) and pure individual cDNA clones can be isolated from the synthetic library by clonal selection.
  • cDNA synthetic substance
  • cDNA pure individual cDNA clones can be isolated from the synthetic library by clonal selection.
  • creating a cDNA library from messenger RNA and subsequently isolating individual clones from that library results in an approximately 10 4 - 10 6 fold purification of the native message.
  • Purification of starting material or natural material to at least one order of magnitude, preferably two or three orders, and more preferably four or five orders of magnitude is expressly contemplated.
  • isolated requires that the material be removed from its original environment (e.g., the natural environment if it is naturally occurring).
  • a naturally-occurring polynucleotide present in a living animal is not isolated, but the same polynucleotide, separated from some or all of the coexisting materials in the natural system, is isolated.
  • the term “recombinant” means that the 5' EST is adjacent to "backbone" nucleic acid to which it is not adjacent in its natural environment. Additionally, to be “enriched” the 5' ESTs will represent 5% or more of the number of nucleic acid inserts in a population of nucleic acid backbone molecules.
  • Backbone molecules according to the present invention include nucleic acids such as expression vectors, self-replicating nucleic acids, viruses, integrating nucleic acids, and other vectors or nucleic acids used to maintain or manipulate a nucleic acid insert of interest.
  • the enriched 5' ESTs represent 15% or more of the number of nucleic acid inserts in the population of recombinant backbone molecules.
  • the enriched 5' ESTs represent 50% or more of the number of nucleic acid inserts in the population of recombinant backbone molecules. In a highly preferred embodiment, the enriched 5' ESTs represent 90% or more of the number of nucleic acid inserts in the population of recombinant backbone molecules.
  • 5' ESTs in cDNA libraries in which one or more 5' ESTs make up 5% or more of the number of nucleic acid inserts in the backbone molecules are "enriched recombinant 5' ESTs" as defined herein.
  • 5' ESTs in a population of plasmids in which one or more 5' EST of the present invention have been inserted such that they represent 5% or more of the number of inserts in the plasmid backbone are " enriched recombinant 5' ESTs" as defined herein.
  • 5' ESTs in cDNA libraries in which 5' ESTs constitute less than 5% of the number of nucleic acid inserts in the population of backbone molecules, such as libraries in which backbone molecules having a 5' EST insert are extremely rare, are not "enriched recombinant 5' ESTs.”
  • the present invention relates to 5' ESTs which are derived from genes encoding secreted proteins.
  • a "secreted” protein is one which, when expressed in a suitable host cell, is transported across or through a membrane, including transport as a result of signal peptides in its amino acid sequence.
  • "Secreted” proteins include without limitation proteins secreted wholly (e.g. soluble proteins), or partially (e.g. receptors) from the cell in which they are expressed.
  • “Secreted” proteins also include without limitation proteins which are transported across the membrane of the endoplasmic reticulum.
  • Such 5' ESTs include nucleic acid sequences, called signal sequences, which encode signal peptides which direct the extracellular secretion of the proteins encoded by the genes from which the 5' ESTs are derived. Generally, the signal peptides are located at the amino termini of secreted proteins.
  • Secreted proteins are translated by ribosomes associated with the "rough" endoplasmic reticulum. Generally, secreted proteins are co-translationally transferred to the membrane of the endoplasmic reticulum. Association of the ribosome with the endoplasmic reticulum during translation of secreted proteins is mediated by the signal peptide. The signal peptide is typically cleaved following its co-translational entry into the endoplasmic reticulum.
  • secreted proteins may proceed through the Golgi apparatus.
  • the proteins may undergo post-translational modification before entering secretory vesicles which transport them across the cell membrane.
  • the 5' ESTs of the present invention have several important applications. For example, they may be used to obtain and express cDNA clones which include the full protein coding sequences of the corresponding gene products, including the authentic translation start sites derived from the 5' ends of the coding sequences of the mRNAs from which the 5' ESTs are derived. These cDNAs will be referred to hereinafter as "full length cDNAs.” These cDNAs may also include DNA derived from mRNA sequences upstream of the translation start site. The full length cDNA sequences may be used to express the proteins corresponding to the 5' ESTs. As discussed above, secreted proteins are therapeutically important.
  • the proteins expressed from the cDNAs may be useful in treating or controlling a variety of human conditions.
  • the 5' ESTs may also be used to obtain the corresponding genomic DNA.
  • the term "corresponding genomic DNA” refers to the genomic DNA which encodes the mRNA from which the 5' EST was derived.
  • the 5' ESTs may be used to obtain and express extended cDNAs encoding portions of the secreted protein.
  • the portions may comprise the signal peptides of the secreted proteins or the mature proteins generated when the signal peptide is cleaved off.
  • the portions may also comprise polypeptides having at least 10 consecutive amino acids encoded by the extended cDNAs or full length cDNAs.
  • the portions may comprise at least 15 consecutive amino acids encoded by the extended cDNAs or full length cDNAs.
  • the portions may comprise at least 25 consecutive amino acids encoded by the extended cDNAs or full length cDNAs.
  • the portions may comprise at least 40 amino acids encoded by the extended cDNAs or full length cDNAs.
  • Antibodies which specifically recognize the entire secreted proteins encoded by the extended cDNAs, full length cDNAs, or fragments thereof having at least 10 consecutive amino acids, at least 15 consecutive amino acids, at least 25 consecutive amino acids, or at least 40 consecutive amino acids may also be obtained as described below.
  • Antibodies which specifically recognize the mature protein generated when the signal peptide is cleaved may also be obtained as described below.
  • antibodies which specifically recognize the signal peptides encoded by the extended cDNAs or full length cDNAs may also be obtained.
  • the extended cDNAs obtained using the 5' ESTs include the signal sequence. In other embodiments, the extended cDNAs obtained using the 5' ESTs may include the full coding sequence for the mature protein (i.e. the protein generated when the signal polypeptide is cleaved off). In addition, the extended cDNAs obtained using the 5' ESTs may include regulatory regions upstream of the translation start site or downstream of the stop codon which control the amount, location, or developmental stage of gene expression.
  • the proteins expressed from the extended cDNAs or full length cDNAs obtained using the 5' ESTs may be useful in treating or controlling a variety of human conditions.
  • the 5' ESTs (or cDNAs or genomic DNAs obtained therefrom) may be used in forensic procedures to identify individuals or in diagnostic procedures to identify individuals having genetic diseases resulting from abnormal expression of the genes corresponding to the 5' ESTs.
  • the present invention is useful for constructing a high resolution map of the human chromosomes.
  • the present invention also relates to secretion vectors capable of directing the secretion of a protein of interest. Such vectors may be used in gene therapy strategies in which it is desired to produce a gene product in one cell which is to be delivered to another location in the body. Secretion vectors may also facilitate the purification of desired proteins.
  • the present invention also relates to expression vectors capable of directing the expression of an inserted gene in a desired spatial or temporal manner or at a desired level. Such vectors may include sequences upstream of the 5' ESTs, such as promoters or upstream regulatory sequences.
  • the present invention may also be used for gene therapy to control or treat genetic diseases.
  • Signal peptides may also be fused to heterologous proteins to direct their extracellular secretion.
  • Bacterial clones containing Bluescript plasmids having inserts containing the 5' ESTs of the present invention (SEQ ID NOs: 38-305 are presently stored at 80°C in 4% (v/v) glycerol in the inventor's laboratories under the designations listed next to the SEQ ID NOs in LI).
  • the inserts may be recovered from the deposited materials by growing the appropriate clones on a suitable medium.
  • the Bluescript DNA can then be isolated using plasmid isolation procedures familiar to those skilled in the art such as alkaline lysis minipreps or large scale alkaline lysis plasmid isolation procedures.
  • the plasmid DNA may be further enriched by centriftigation on a cesium chloride gradient, size exclusion chromatography, or anion exchange chromatography.
  • the plasmid DNA obtained using these procedures may then be manipulated using standard cloning techniques familiar to those skilled in the art.
  • a PCR can be done with primers designed at both ends of the EST insertion.
  • the PCR product which corresponds to the 5' EST can then be manipulated using standard cloning techniques familiar to those skilled in the art.
  • One aspect of the present invention is a purified or isolated nucleic acid having the sequence of one of SEQ ED NOs: 38-305 or having a sequence complementary thereto.
  • the nucleic acid is recombinant.
  • Another aspect of the present invention is a purified or isolated nucleic acid comprising at least 10 consecutive bases of the sequence of one of SEQ ID NOs: 38-305 or one of the sequences complementary thereto.
  • Yet another aspect of the present invention is a purified or isolated nucleic acid comprising at least 15 consecutive bases of one of the sequences of SEQ LD NOs: 38-305 or one of the sequences complementary thereto.
  • the nucleic acid is recombinant.
  • a further aspect of the present invention is a purified or isolated nucleic acid of at least 15 bases capable of hybridizing under stringent conditions to the sequence of one of SEQ ID NOs: 38-305 or one of the sequences complementary to the sequences of SEQ ID NOs: 38-305.
  • the nucleic acid is recombinant.
  • Another aspect of the present invention is a purified or isolated nucleic acid encoding a human gene product, said human gene product having a sequence partially encoded by one of the sequences of SEQ ID NO: 38-305.
  • Still another aspect of the present invention is a method of making a cDNA encoding a human secretory protein, said human secretory protein being partially encoded by one of SEQ ID NOs 38-305, comprising the steps of contacting a collection of mRNA molecules from human cells with a primer comprising at least 15 consecutive nucleotides of a sequence complementary to one of SEQ ID NOs: 38-305; hybridizing said primer to an mRNA in said collection that encodes said protein; reverse transcribing said hybridized primer to make a first cDNA strand from said mRNA; making a second cDNA strand complementary to said first cDNA strand; and isolating the resulting cDNA encoding said protein comprising said first cDNA strand and said second cDNA strand.
  • Another aspect of the invention is an isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein encoded by one of SEQ ID NOs 38-305 or a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method described in the preceding paragraph.
  • the cDNA comprises the full protein coding sequence of said protein which sequence is partially included in one of the sequences of SEQ ID NOs: 38-305.
  • Another aspect of the present invention is a method of making a cDNA encoding a human secretory protein that is partially encoded by one of SEQ ID NOs 38-305, comprising the steps of obtaining a cDNA comprising one of the sequences of SEQ ID NOs: 38-305; contacting said cDNA with a detectable probe comprising at least 15 consecutive nucleotides of said sequence of SEQ LD NO: 38-305 or a sequence complementary thereto under conditions which permit said probe to hybridize to said cDNA; identifying a cDNA which hybridizes to said detectable probe; and isolating said cDNA which hybridizes to said probe.
  • Another aspect of the present invention is an isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein encoded by one of SEQ ID NOs 38-305 or a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method described in the preceding paragraph.
  • the cDNA comprises the full protein coding sequence partially included in one of the sequences of SEQ ID NOs: 38-305.
  • Another aspect of the present invention is a method of making a cDNA comprising one of the sequence of SEQ ID NOs: 38-305, comprising the steps of contacting a collection of mRNA molecules from human cells with a first primer capable of hybridizing to the polyA tail of said mRNA; hybridizing said first primer to said polyA tail; reverse transcribing said mRNA to make a first cDNA strand; making a second cDNA strand complementary to said first cDNA strand using at least one primer comprising at least 15 nucleotides of one of the sequences of SEQ ED NOs 38-305; and isolating the resulting cDNA comprising said first cDNA strand and said second cDNA strand.
  • Another aspect of the present invention is an isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein encoded by one of SEQ ED NOs 38-305 or a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method described in the preceding paragraph.
  • the cDNA comprises the full protein coding sequence partially included in one of the sequences of SEQ ED NOs: 38-305.
  • the second cDNA strand is made by contacting said first cDNA strand with a first pair of primers, said first pair 'of primers comprising a second primer comprising at least 15 consecutive nucleotides of one of the sequences of SEQ ED NOs 38-305 and a third primer having a sequence therein which is included within the sequence of said first primer; performing a first polymerase chain reaction with said first pair of nested primers to generate a first PCR product; contacting said first PCR product with a second pair of primers, said second pair of primers comprising a fourth primer, said fourth primer comprising at least 15 consecutive nucleotides of said sequence of one of SEQ ID NOs: 38-305 , and a fifth primer, said fourth and fifth primers being capable of hybridizing to sequences within said first PCR product; and performing a second polymerase chain reaction, thereby generating a second PCR product.
  • One aspect of the present invention is an isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein encoded by one of SEQ ED NOs 38-305, or a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method of the preceding paragraph.
  • the cDNA comprises the full protein coding sequence partially included in one of the sequences of SEQ ED NOs: 38-305.
  • Another aspect of the present invention is the method described four paragraphs above in which the second cDNA strand is made by contacting said first cDNA strand with a second primer comprising at least 15 consecutive nucleotides of the sequences of SEQ ID NOs: 38-305; hybridizing said second primer to said first strand cDNA; and extending said hybridized second primer to generate said second cDNA strand.
  • Another aspect of the present invention is an isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein partially encoded by one of SEQ ED NOs 38-305 or comprising a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method described in the preceding paragraph.
  • the cDNA comprises the full protein coding sequence partially included in of one of the sequences of SEQ ED NOs: 38-305.
  • Another aspect of the present invention is a method of making a protein comprising one of the sequences of SEQ ED NOs: 306-573, comprising the steps of obtaining a cDNA encoding the full protein sequence partially included in one of the sequences of sequence of SEQ ED NOs: 38-305; inserting said cDNA in an expression vector such that said cDNA is operably linked to a promoter; introducing said expression vector into a host cell whereby said host cell produces the protein encoded by said cDNA; and isolating said protein.
  • Another aspect of the present invention is an isolated protein obtainable by the method described in the preceding paragraph.
  • Another aspect of the present invention is a method of obtaining a promoter DNA comprising the steps of obtaining DNAs located upstream of the nucleic acids of SEQ ID NOs: 38-305 or the sequences complementary thereto; screening said upstream DNAs to identify a promoter capable of directing transcription initiation; and isolating said DNA comprising said identified promoter.
  • the obtaining step comprises chromosome walking from said nucleic acids of SEQ ID NOs: 38-305 or sequences complementary thereto.
  • the screening step comprises inserting said upstream sequences into a promoter reporter vector.
  • the screening step comprises identifying motifs in said upstream DNAs which are transcription factor binding sites or transcription start sites.
  • Another aspect of the present invention is an isolated promoter obtainable by the method described above.
  • Another aspect of the present invention is an isolated or purified protein comprising one of the sequences of SEQ ID NOs: 306-573.
  • Another aspect of the present invention is the inclusion of at least one of the sequences of SEQ ED NOs: 38-305, or one of the sequences complementary to the sequences of SEQ ED NOs: 38-305, or a fragment thereof of at least 15 consecutive nucleotides in an array of discrete ESTs or fragments thereof of at least 15 nucleotides in length.
  • the array includes at least two of the sequences of SEQ ED NOs: 38-305, the sequences complementary to the sequences of SEQ ED NOs: 38-305, or fragments thereof of at least 15 consecutive nucleotides.
  • the array includes at least five of the sequences of SEQ ID NOs: 38-305, the sequences complementary to the sequences of SEQ ID NOs: 38-305, or fragments thereof of at least 15 consecutive nucleotides.
  • Another aspect of the present invention is a promoter having a sequence selected from the group consisting of SEQ ED NOs: 31, 34, and 37.
  • Figure 1 is a summary of a procedure for obtaining cDNAs which have been selected to include the 5' ends of the mRNAs from which they derived.
  • Figure 2 shows the distribution of Von Heijne scores for 5' ESTs in each of the categories described herein and the probability that these 5' ESTs encode a signal peptide.
  • Figure 3 summarizes a general method used to clone and sequence extended cDNAs containing sequences adjacent to 5' ESTs.
  • Figure 4 (description of promoters structure isolated from SignalTag 5' ESTs) provides a schematic description of promoters isolated and the way they are assembled with the corresponding 5' tags.
  • Table IV is an analysis of the 43 amino acids located at the N terminus of all human SwissProt proteins to determine the frequency of false positives and false negatives using the techniques for signal peptide identification described herein.
  • Table V shows the distribution of 5' ESTs in each category described herein and the number of 5' ESTs in each category having a given minimum Von Heijne's score.
  • Table VI shows the distribution of 5' ESTs in each category described herein with respect to the tissue from which the 5' ESTs of the corresponding mRNA were obtained.
  • Table VII describes the transcription factor binding sites present in each of these promoters.
  • mRNAs with intact 5' ends must be obtained.
  • approaches for obtaining such mRNAs with intact 5' ends as described below: either chemical (1) or enzymatic (2).
  • the 5' ends of eukaryotic mRNAs possess a structure referred to as a "cap" which comprises a guanosine methylated at the 7 position.
  • the cap is joined to the first transcribed base of the mRNA by a 5', 5 '-triphosphate bond.
  • the 5' guanosine is methylated in both the 2 and 7 positions.
  • the 5' guanosine is trimethylated at the 2, 7 and 7 positions.
  • the 5' cap is specifically derivatized and coupled to a reactive group on an immobilizing substrate.
  • This specific derivatization is based on the fact that only the ribose linked to the methylated guanosine at the 5' end of the mRNA and the ribose linked to the base at the 3' tei inus of the mRNA, possess 2', 3'-cis diols.
  • the 2', 3'-cis diol of the 3' terminal ribose may be chemically modified, substituted, converted, or eliminated, leaving only the ribose linked to the methylated guanosine at the 5' end of the mRNA with a 2', 3'-cis diol.
  • a variety of techniques are available for eliminating the 2', 3'-cis diol on the 3' terminal ribose. For example, controlled alkaline hydrolysis may be used to generate mRNA fragments in which the 3' terminal ribose is a 3 '-phosphate, 2'-phosphate or (2', 3')-cyclophosphate.
  • RNA ligase such as T4 RNA ligase
  • RNA was incubated in a final reaction medium of 10 ⁇ l in the presence of 5 U of T phage RNA ligase in the buffer provided by the manufacturer (Gibco - BRL), 40 U of the RNase inhibitor RNasin (Promega) and, 2 ⁇ l of 32 pCp (Amersham #PB 10208). The incubation was performed at 37°C for 2 hours or overnight at 7-8°C.
  • the 2', 3'-cis diol present at the 5' end of the mRNA may be oxidized using reagents such as NaBH,, NaBH 3 CN, or sodium periodate, thereby converting the 2', 3'-cis diol to a dialdehyde.
  • reagents such as NaBH,, NaBH 3 CN, or sodium periodate, thereby converting the 2', 3'-cis diol to a dialdehyde.
  • Example 2 describes the oxidation of the 2', 3'-cis diol at the 5' end of the mRNA with sodium periodate.
  • 0.1 OD unit of either a capped oligoribonucleotide of 47 nucleotides (including the cap) or an uncapped oligoribonucleotide of 46 nucleotides were treated as follows.
  • the oligoribonucleotides were produced by in vitro transcription using the transcription kit "AmpliScribe T7" (Epicentre Technologies). As indicated below, the DNA template for the RNA transcript contained a single cytosine. To synthesize the uncapped RNA, all four NTPs were included in the in vitro transcription reaction. To obtain the capped RNA, GTP was replaced by an analogue of the cap, m7G(5')ppp(5')G.
  • oligoribonucleotides were dissolved in 9 ⁇ l of acetate buffer (0.1 M sodium acetate, pH 5.2) and 3 ⁇ l of freshly prepared 0.1 M sodium periodate solution. The mixture was incubated for 1 hour in the dark at 4°C or room temperature. Thereafter, the reaction was stopped by adding 4 ⁇ l of 10% ethylene glycol. The product was ethanol precipitated, resuspended in at least 10 ⁇ l of water or appropriate buffer and dialyzed against water.
  • acetate buffer 0.1 M sodium acetate, pH 5.2
  • the resulting aldehyde groups may then be coupled to molecules having a reactive amine group, such as hydrazine, carbazide, thiocarbazide or semicarbazide groups, in order to facilitate enrichment of the 5' ends of the mRNAs.
  • a reactive amine group such as hydrazine, carbazide, thiocarbazide or semicarbazide groups
  • Molecules having reactive amine groups which are suitable for use in selecting mRNAs having intact 5' ends include avidin, proteins, antibodies, vitamins, ligands capable of specifically binding to receptor molecules, or oligonucleotides.
  • Example 3 below describes the coupling of the resulting dialdehyde to biotin.
  • Sample 1 The 46 nucleotide uncapped in vitro transcript prepared as in Example 2 and labeled with j2 pCp as described in Example 1.
  • Sample 2. The 46 nucleotide uncapped in vitro transcript prepared as in Example 2, labeled with o2 pCp as described in Example 1, treated with the oxidation reaction of Example 2, and subjected to the biotinylation conditions of Example 3.
  • Sample 3. The 47 nucleotide capped in vitro transcript prepared as in Example 2 and labeled with 32 pCp as described in Example 1.
  • Sample 4 The 47 nucleotide capped in vitro transcript prepared as in Example 2, labeled with 32 pCp as described in Example 1, treated with the oxidation reaction of Example 2, and subjected to the biotinylation conditions of Example 3.
  • Samples 1 and 2 had identical migration rates, demonstrating that the uncapped RNAs were not oxidized and biotinylated. Sample 3 migrated more slowly than Samples 1 and 2, while Sample 4 exhibited the slowest migration. The difference in migration of the RNAs in Samples 3 and 4 demonstrates that the capped RNAs were specifically biotinylated.
  • mRNAs having intact 5' ends may be enriched by binding the molecule containing a reactive amine group to a suitable solid phase substrate such as the inside of the vessel containing the mRNAs, magnetic beads, chromatography matrices, or nylon or nitrocellulose membranes.
  • a suitable solid phase substrate such as the inside of the vessel containing the mRNAs, magnetic beads, chromatography matrices, or nylon or nitrocellulose membranes.
  • the solid phase substrate may be coupled to avidin or streptavidin.
  • the molecule having the reactive amine group is an antibody or receptor ligand
  • the solid phase substrate may be coupled to the cognate antigen or receptor.
  • the molecule having a reactive amine group comprises an oligonucleotide
  • the solid phase substrate may comprise a complementary oligonucleotide.
  • the mRNAs having intact 5' ends may be released from the solid phase following the enrichment procedure.
  • the mRNAs may be released from the solid phase by simply heating to 95 degrees Celsius in 2% SDS.
  • the molecule having a reactive amine group may also be cleaved from the mRNAs having intact 5' ends following enrichment.
  • Example 5 describes the capture of biotinylated mRNAs with streptavidin coated beads and the release of the biotinylated mRNAs from the beads following enrichment.
  • the streptavidin coated magnetic beads were prepared according to the manufacturer's instructions (CPG Inc., USA).
  • the biotinylated mRNAs were added to a hybridization buffer (1.5 M NaCl, pH 5 - 6). After incubating for 30 minutes, the unbound and nonbiotinylated material was removed. The beads were then washed several times in water with 1% SDS. The beads thus obtained were incubated for 15 minutes at 95°C in water containing 2% SDS.
  • Example 6 demonstrates the efficiency with which biotinylated mRNAs were recovered from the streptavidin coated beads.
  • RNAs were labeled with 32 pCp, oxidized, biotinylated and bound to streptavidin coated beads as described above. Subsequently, the bound RNAs were incubated for 5, 15 or 30 minutes at 95°C in the presence of 2% SDS.
  • the products of the reaction were analyzed by electrophoresis on 12% polyacrylamide gels under denaturing conditions (7 M urea). The gels were subjected to autoradiography. During this manipulation, the hydrazone bonds were not reduced.
  • an oligonucleotide which has been derivatized to contain a reactive amine group is specifically coupled to mRNAs having an intact cap.
  • the 3' end of the mRNA is blocked prior to the step in which the aldehyde groups are joined to the derivatized oligonucleotide, as described above, so as to prevent the derivatized oligonucleotide from being joined to the 3' end of the mRNA.
  • pCp may be attached to the 3' end of the mRNA using T4 RNA ligase as described in example 1.
  • blocking the 3' end of the mRNA is an optional step.
  • Derivatized oligonucleotides may be prepared as described in Example 7. EXAMPLE 7
  • oligonucleotide phosphorylated at its 3' end was converted to a 3' hydrazide in 3' by treatment with an aqueous solution of hydrazine or of dihydrazide of the formula H 2 N(R1)NH 2 at about 1 to 3 M, and at pH 4 5 at a temperature of 8°C overnight
  • This incubation was performed in the presence of a carbodiimide type agent soluble in water such as l-ethyl-3-(3-dimethylaminopropyl)carbodii ide at a final concentration of 0 3 M
  • the derivatized oligonucleotide was then separated from the other agents and products using a standard technique for isolating oligonucleotides
  • the mRNAs to be enriched may be treated to eliminate the 3' OH groups which may be present thereon This may be accomplished by enzymatic ligation of sequences lacking a 3' OH, such as pCp, as desc ⁇ bed in Example 1 Alternatively, the 3' OH groups may be eliminated by alkaline hydrolysis as descnbed in Example 8 below
  • the oxidized mRNA was dissolved in an acidic medium such as 50 ⁇ l of sodium acetate pH 4-6. Fifty ⁇ l of a solution of the derivatized oligonucleotide were added in order to obtain an mRNA: derivatized oligonucleotide ratio of 1:20. The mixture was reduced with a borohydride and incubated for 2 h at 37°C or overnight (14 h) at 10°C. The mixture was then ethanol precipitated, resuspended in 10 ⁇ l or more of water or appropriate buffer and dialyzed against distilled water. If desired, the resulting product may be analyzed using acrylamide gel electrophoresis, HPLC analysis, or other conventional techniques.
  • a reverse transcription reaction may be performed as described in Example 11 below.
  • oligodeoxyribonucleotide was derivatized as follows. Three OD units of an oligodeoxyribonucleotide of sequence 5 ⁇ TCAAGAATTCGCACGAGACCATTA3' (SEQ ED NO:3) having 5'-OH and 3'-P ends were dissolved in 70 ⁇ l of a 1.5 M hydroxybenzotriazole solution, pH 5.3, prepared in dimethylformamide/water (75:25) containing 2 ⁇ g of l-ethyl-3-(3-dimethylan ⁇ inopropyl)carbodiimide.
  • the mixture was incubated for 2 h 30 min at 22°C and then precipitated twice in LiCl ⁇ 4/acetone.
  • the pellet was resuspended in 200 ⁇ l of 0.25 M hydrazine and incubated at 8°C from 3 to 14 h. Following the hydrazine reaction, the mixture was precipitated twice in LiClO acetone.
  • the messenger RNAs to be reverse transcribed were extracted from blocks of placenta having sides of 2 cm which had been stored at -80°C. The total RNA was extracted using conventional acidic phenol techniques. Oligo-dT chromatography was used to purify the mRNAs. The integrity of the mRNAs was checked by Northern-blotting. The diol groups on 7 ⁇ g of the placental mRNAs were oxidized as described above in Example 9. The derivatized oligonucleotide was joined to the mRNAs as described in Example 10 above except that the precipitation step was replaced by an exclusion chromatography step to remove derivatized oligodeoxyribonucleotides which were not joined to mRNAs. Exclusion chromatography was performed as follows:
  • the pipette was filled with the gel suspension until the height of the gel stabilized at 1 cm from the top of the pipette.
  • the column was then equilibrated with 20 ml of equilibration buffer (10 mM Tris HCl pH 7.4, 20 mM NaCl).
  • RNA of the cDNA:RNA heteroduplexes obtained from the reverse transcription reaction by an alkaline hydrolysis
  • a portion of the resulting single stranded cDNAs was spotted on a positively charged membrane and hybridized, using conventional methods, to a 32 P labeled probe having a sequence identical to that of the derivatized oligonucleotide.
  • Control spots containing, 1 pmol, 100 finol, 50 frnol, 10 frnol and 1 frnol of a control oligodeoxyribonucleotide of sequence identical to that of the derivatized oligonucleotide were included.
  • the signal observed in the spots containing the cDNA indicated that approximately 15 frnol of the derivatized oligonucleotide had been reverse transcribed.
  • the single stranded cDNAs obtained from the above first strand synthesis were used as template for PCR reactions. Two types of reactions were carried out. First, specific amplification of the mRNAs for alpha globin, dehydrogenase, ppl5 and elongation factor E4 were carried out using the following pairs of oligodeoxyribonucleotide primers.
  • GLO-S 5'CCG ACA AGA CCA ACG TCA AGG CCG C3' (SEQ ED NO:5)
  • GLO-As 5'TCA CCA GCA GGC AGT GGC TTA GGA G 3' (SEQ ED NO:6)
  • Elongation factor E4 EFA1-S 5'ATG GGA AAG GAA AAG ACT CAT ATC A3' (SEQ ID NO:l 1)
  • EFIA-As 5'AGC AGC AAC AAT CAG GAC AGC ACA G3' (SEQ ID NO.T2)
  • Sample 1 The products of a PCR reaction using the globin primers of SEQ ID NOs 5 and 6 in the presence of cDNA
  • Sample 2 The products of a PCR reaction using the globin primers of SEQ ED NOs
  • Sample 3 The products of a PCR reaction using the dehydrogenase primers of SEQ ED NOs 7 and 8 in the presence of cDNA.
  • Sample 4 The products of a PCR reaction using the dehydrogenase primers of SEQ ID NOs 7 and 8 in the absence of added cDNA.
  • Sample 5 The products of a PCR reaction using the ppl5 primers of SEQ ED NOs 9 and 10 in the presence of cDNA.
  • Sample 6 The products of a PCR reaction using the ppl5 primers of SEQ ID NOs 9 and 10 in the absence of added cDNA.
  • Sample 7 The products of a PCR reaction using the EEF4 primers of SEQ ID NOs
  • Sample 8 The products of a PCR reaction using the EEF4 primers of SEQ ID NOs 11 and 12 in the absence of added cDNA.
  • Single stranded RNAs are eliminated to obtain a population of cDNA/mRNA heteroduplexes in which the mRNA includes an intact 5' end.
  • the resulting heteroduplexes may be captured on a solid phase coated with a molecule capable of interacting with the molecule used to derivatize the 5' end of the mRNA.
  • the strands of the heteroduplexes are separated to recover single stranded first cDNA strands which include the 5' end of the mRNA.
  • Second strand cDNA synthesis may then proceed using conventional techniques. For example, the procedures disclosed in WO 96/34981 or in Caminci.
  • cDNAs which include the sequence derived from the 5' end of the coding sequence of the mRNA.
  • a reverse transcription reaction is conducted to extend a primer complementary to the mRNA to the 5' end of the mRNA.
  • second strand cDNA synthesis is conducted with a primer complementary to the oligonucleotide tag.
  • isolated mRNA is treated with alkaline phosphatase to remove the phosphate groups present on the 5' ends of uncapped incomplete mRNAs.
  • alkaline phosphatase to remove the phosphate groups present on the 5' ends of uncapped incomplete mRNAs.
  • the cap present on full length mRNAs is enzymatically removed with a decapping enzyme such as T4 polynucleotide kinase or tobacco acid pyrophosphatase.
  • oligonucleotide which may be either a DNA oligonucleotide or a DNA-RNA hybrid oligonucleotide having RNA at its 3' end, is then ligated to the phosphate present at the 5' end of the decapped mRNA using T4 RNA ligase.
  • the oligonucleotide may include a restriction site to facilitate cloning of the cDNAs following their synthesis.
  • Example 12 below describes one enzymatic method based on the doctoral thesis of Dumas.
  • Oligonucleotides having an unphosphorylated 5' end may be synthesized by adding a fluorochrome at the 5' end. The inclusion of a stretch of adenosine ribophosphates at the 3' end of the oligonucleotide increases ligation efficiency. It will be appreciated that the oligonucleotide may contain cloning sites other than EcoRl. Following ligation of the oligonucleotide to the phosphate present at the 5' end of the decapped mRNA, first and second strand cDNA synthesis is carried out using conventional methods or those specified in EPO 625,572 and Kato et al.
  • cDNA may then be ligated into vectors such as those disclosed in Kato et al., supra or other nucleic acid vectors known to those skilled in the art using techniques such as those described in Sambrook et al., Molecular Cloning: A Laboratory Manual 2d Ed., Cold Spring Harbor Laboratory Press, 1989, the disclosure of which is inco ⁇ orated herein by reference.
  • the 5' ESTs of the present invention were obtained using the aforementioned chemical and enzymatic approaches for enriching mRNAs for those having intact 5' ends as decribed below.
  • mRNAs were prepared as described in Example 13 below.
  • RNAs or polyA + RNAs derived from 29 different tissues were respectively purchased from LABIMO and CLONTECH and used to generate 44 cDNA libraries as follows. The purchased RNA had been isolated from cells or tissues using acid guanidium thiocyanate-phenol-chloroform extraction (Chomczyniski and Sacchi, Analytical Biochemistry 162:156-159, 1987). PolyA + RNA was isolated from total RNA (LABIMO) by two passes of oligo dT chromatography, as described by Aviv and Leder, Proc. Natl.
  • RNAs The quality and the integrity of the polyA+ RNAs were checked.
  • Northern blots hybridized with a globin probe were used to confirm that the mRNAs were not degraded.
  • Contamination of the polyA " mRNAs by ribosomal sequences was checked using Northern blots and a probe derived from the sequence of the 28 S rRNA.
  • Preparations of mRNAs with less than 5% of rRNAs were used in library construction. To avoid constructing libraries with RNAs contaminated by exogenous sequences (prokaryotic or fungal), the presence of bacterial 16S ribosomal sequences or of two highly expressed fungal mRNAs was examined using PCR.
  • oligonucleotide tag was attached to the 5' ends of the mRNAs.
  • the oligonucleotide tag had an EcoRl site therein to facilitate later cloning procedures.
  • the same nucleotidic sequence was used to design the ligated oligonucleotide in both chemical and enzymatic approaches.
  • the tag used was an oligodeoxyribonucleotide which was linked to the cap of the mRNA whereas in the enzymatic ligation, the tag was a chimeric hemi 5'DNA/RNA3' oligonucleotide which was ligated to the 5' end of decapped mRNA as described in example 12.
  • the integrity of the mRNA was examined by performing a Northern blot with 200 to 500 ng of mRNA using a probe complementary to the oligonucleotide tag before performing the first strand synthesis as described in example 14.
  • EXAMPLE 14 cDNA Synthesis Using mRNA Templates Having Intact 5' Ends
  • first strand cDNA synthesis was performed using the Superscript II (Gibco BRL) or the Rnase H Minus M-MLV (Promega) reverse transcriptase with random nonamers as primers.
  • Superscript II Gibco BRL
  • Rnase H Minus M-MLV Promega reverse transcriptase with random nonamers
  • methylated dCTP was used for first strand synthesis.
  • the first strand of cDNA was precipitated using isopropanol in order to eliminate residual primers.
  • the second strand of the cDNA was synthesized with a Klenow fragment using a primer corresponding to the 5' end of the ligated oligonucleotide described in Example 12.
  • the primer is 20-25 bases in length.
  • Methylated dCTP was also used for second strand synthesis in order to protect internal EcoRI sites in the cDNA from digestion during the cloning process.
  • the ends of the cDNA were blunted with T4 DNA polymerase (Biolabs) and the cDNA was digested with EcoRI. Since methylated dCTP was used during cDNA synthesis, the EcoRI site present in the tag was the only hemi-methylated site, hence the only site susceptible to EcoRI digestion.
  • the cDNA was then size fractionated using exclusion chromatography (Ac A, Biosepra) and fractions corresponding to cDNAs of more than 150 bp were pooled and ethanol precipitated.
  • the cDNA was directionally cloned into the Smal and EcoRI ends of the phagemid pBlueScript vector (Stratagene). The ligation mixture was electroporated into bacteria and propagated under appropriate antibiotic selection.
  • Clones containing the oligonucleotide tag attached were then selected as described in Example 16 below.
  • EXAMPLE 16 Selection of Clones Having the Oligonucleotide Tag Attached Thereto
  • the plasmid DNAs containing 5' EST libraries made as described above were purified (Qiagen).
  • a positive selection of the tagged clones was performed as follows. Briefly, in this selection procedure, the plasmid DNA was converted to single stranded DNA using gene II endonuclease of the phage FI in combination with an exonuclease (Chang et al, Gene 127:95-8, 1993) such as exonuclease HI or T7 gene 6 exonuclease.
  • the resulting single stranded DNA was then purified using paramagnetic beads as described by Fry et al, Biotechniques, 13: 124-131, 1992.
  • the single stranded DNA was hybridized with a biotinylated oligonucleotide having a sequence corresponding to the 3' end of the oligonucleotide described in Example 13.
  • the primer has a length of 20-25 bases. Clones including a sequence complementary to the biotinylated oligonucleotide were captured by incubation with streptavidin coated magnetic beads followed by magnetic selection.
  • the plasmid DNA was released from the magnetic beads and converted into double stranded DNA using a DNA polymerase such as the ThermoSequenase obtained from Amersham Pharmacia Biotech. Alternatively, protocoles such as the one described in the Gene Trapper kit available from Gibco BRL may be used.
  • the double stranded DNA was then electroporated into bacteria. The percentage of positive clones having the 5' tag oligonucleotide was estimated to typically rank between 90 and 98%o using dot blot analysis. Following electroporation, the libraries were ordered in 384-microtiter plates (MTP).
  • Plasmid inserts were first amplified by PCR on PE 9600 thermocyclers (Perkin-Elmer, Applied Biosystems Division, Foster City, CA), using standard SETA-A and SETA-B primers (Genset SA), AmpliTaqGold (Perkin-Elmer), dNTPs (Boehringer), buffer and cycling conditions as recommended by the Perkin-Elmer Co ⁇ oration. PCR products were then sequenced using automatic ABI Prism 377 sequencers
  • Sequencing reactions were performed using PE 9600 thermocyclers with standard dye-primer chemistry and ThermoSequenase (Amersham Pharmacia Biotech).
  • the primers used were either T7 or 21M13 (available from Genset SA) as appropriate.
  • the primers were labeled with the JOE, FAM, ROX and TAMRA dyes.
  • the dNTPs and ddNTPs used in the sequencing reactions were purchased from Boehringer. Sequencing buffer, reagent concentrations and cycling conditions were as recommended by Amersham.
  • sequence data from the 44 cDNA libraries made as described above were transferred to a proprietary database, where quality control and validation steps were performed.
  • a proprietary base-caller working using a Unix system, automatically flagged suspect peaks, taking into account the shape of the peaks, the inter-peak resolution, and the noise level.
  • the proprietary base-caller also performed an automatic trimming. Any stretch of 25 or fewer bases having more than 4 suspect peaks was considered unreliable and was discarded.
  • Sequences corresponding to cloning vector or ligation oligonucleotides were automatically removed from the EST sequences. However, the resulting EST sequences may contain 1 to 5 bases belonging to the above mentioned sequences at their 5' end. If needed, these can easily be removed on a case to case basis.
  • Computer readable media include magnetically, optically, or electronically readable media.
  • the computer readable media may be a hard disc, a floppy disc, a magnetic tape, CD-ROM RAM, or ROM as well as other types of other media known to those skilled in the art.
  • sequence data may be stored and manipulated in a variety of data processor programs in a diversity of formats.
  • sequence data may be stored as text in a word processing file, such as Microsoft WORD or WORDPERFECT or as an ASCII file in a variety of database programs familiar to those of skill in the art, such as DB2, SYBASE, or ORACLE.
  • the computer readable media on which the sequence information is stored may be in a personal computer, a network, a server or other computer systems known to those skilled in the art.
  • the computer or other system preferably includes the storage media described above, and a processor for accessing and manipulating the sequence data.
  • the sequence data Once the sequence data has been stored, it may be manipulated and searched to locate those stored sequences which contain a desired nucleic acid sequence or which encode a protein having a particular functional domain.
  • the stored sequence information may be compared to other known sequences to identify homologies, motifs implicated in biological function, or structural motifs.
  • Programs which may be used to search or compare the stored sequences include the
  • EMBL MacPattern
  • BLAST BLAST
  • NCBI basic local alignment search tool programs for nucleotide
  • BLASTX peptide
  • BLAST programs then extend the alignments on the basis of defined match and mismatch criteria.
  • Motifs which may be detected using the above programs and those described in Example 28 include sequences encoding leucine zippers, helix-turn-helix motifs, glycosylation sites, ubiquitination sites, alpha helices, and beta sheets, signal sequences encoding signal peptides which direct the secretion of the encoded proteins, sequences implicated in transcription regulation such as homeoboxes, acidic stretches, enzymatic active sites, substrate binding sites, and enzymatic cleavage sites.
  • cDNAs derived from mRNAs which were not of interest were identified and eliminated from further consideration as described in Example 18 below.
  • 5' ESTs in the NetGeneTM database which were derived from undesired sequences such as transfer RNAs, ribosomal RNAs, mitochondrial RNAs, prokaryotic RNAs, fungal RNAs, Alu sequences, Ll sequences, .or repeat sequences were identified using the FASTA and BLASTN programs with the parameters listed in Table I.
  • the 5' EST sequences were compared to the sequences of 1190 known tRNAs obtained from EMBL release 38, of which 100 were human. The comparison was performed using FASTA on both strands of the 5' ESTs.
  • the 5' ESTs were compared to 6093 Alu sequences and 1115 Ll sequences to mask 5' ESTs containing such repeat sequences.
  • 5' ESTs including THE and MER repeats, SSTR sequences or satellite, micro-satellite, or telomeric repeats were also eliminated from further consideration.
  • 11.5% of the sequences in the libraries contained repeat sequences. Of this 11.5%, 7% contained Alu repeats, 3.3% contained Ll repeats and the remaining 1.2% were derived from the other screened types of repetitive sequences.
  • These percentages are consistent with those found in cDNA libraries prepared by other groups.
  • the cDNA libraries of Adams et al. contained between 0% and 7.4% Alu repeats depending on the source of the RNA which was used to prepare the cDNA library (Adams et al, Nature 377:174, 1996).
  • sequences of those 5' ESTs remaining after the elimination of undesirable sequences were compared with the sequences of known human mRNAs to determine the accuracy of the sequencing procedures described above.
  • sequences of 5' ESTs derived from known sequences were identified and compared to the original known sequences.
  • a FASTA analysis with overhangs shorter than 5 bp on both ends was conducted on the 5' ESTs to identify those matching an entry in the public human mRNA database.
  • the 6655 5' ESTs which matched a known human mRNA were then realigned with their cognate mRNA and dynamic programming was used to include substitutions, insertions, and deletions in the list of "errors" which would be recognized. Errors occurring in the last 10 bases of the 5' EST sequences were ignored to avoid the inclusion of spurious cloning sites in the analysis of sequencing accuracy.
  • the EST libraries made above included multiple 5' ESTs derived from the same mRNA. The sequences of such 5' ESTs were compared to one another and the longest 5'
  • ESTs for each mRNA were identified. Overlapping cDNAs were assembled into continuous sequences (contigs). The resulting continuous sequences were then compared to public databases to gauge their similarity to known sequences, as described in Example 21 below.
  • NR novelty rate
  • Example 23 To confirm the accuracy of the above method for identifying signal sequences, the analysis of Example 23 was performed. EXAMPLE 23
  • 5' region of the mRNA is in fact a genuine signal peptide based on its Von Heijne's score was calculated based on either the assumption that 10% of human proteins are secreted or the assumption that 20% of human proteins are secreted.
  • the results of this analysis are shown in Figure 2 and table IV.
  • 5' ESTs of the following polypeptides known to be secreted were obtained: human glucagon, gamma interferon induced monokine precursor, secreted cyclophilin-like protein, human pleiotropin, and human biotinidase precursor.
  • the above method successfully identified those 5' ESTs which encode a signal peptide.
  • the signal sequences from the 5' ESTs may be cloned into a vector designed for the identification of signal peptides.
  • a vector designed for the identification of signal peptides are designed to confer the ability to grow in selective medium only to host cells containing a vector with an operably linked signal sequence.
  • the signal sequence of the 5' EST may be inserted upstream and in frame with a non-secreted form of the yeast invertase gene in signal peptide selection vectors such as those described in U.S. Patent No. 5,536,637, the disclosure of which is inco ⁇ orated herein by reference.
  • the medium from these cells is compared to the medium from control cells containing vectors lacking the signal sequence or extended cDNA insert to identify vectors which encode a functional signal peptide or an authentic secreted protein. Those 5' ESTs which encoded a signal peptide, as determined by the method of
  • Example 22 above were further grouped into four categories based on their homology to known sequences as described in Example 24 below.
  • Each of the 5' ESTs was also categorized based on the tissue from which its corresponding mRNA was obtained, as described below in Example 25.
  • Table VI shows the distribution of 5' ESTs in each of the above defined category with respect to the tissue from which the 5'ESTs of the corresponding mRNA were obtained.
  • Table II provides the sequence identification numbers of 5' EST sequences derived from muscle and other mesodermal tissues, the categories in which these sequences fall, and the von Heijne's score of the signal peptides which they encode.
  • the 5' EST sequences and the amino acid sequences they encode are provided in the appended sequence listings.
  • Table III provides the sequence ED numbers of the 5' ESTs and the sequences of the signal peptides which they encode.
  • the sequences of the 5' ESTs and the polypeptides they encode are provided in the sequence listing appended hereto.
  • sequences of DNA SEQ ID NOs: 38-305 can readily be screened for any errors therein and any sequence ambiguities can be resolved by resequencing a fragment containing such errors or amibiguities on both strands.
  • Such fragments may be obtained from the plasmids stored in the inventors' laboratory or can be isolated using the techniques described herein. Resolution of any such ambiguities or errors may be facilitated by using primers which hybridize to sequences located close to the ambiguous or erroneous sequences. For example, the primers may hybridize to sequences within 50-75 bases of the amibiguity or error.
  • the corresponding corrections can be made in the protein sequences encoded by the DNA containing the error or amibiguity.
  • the spatial and temporal expression patterns of the mRNAs corresponding to the 5' ESTs, as well as their expression levels may be determined as described in Example 26 below. Characterization of the spatial and temporal expression patterns and expression levels of these mRNAs is useful for constructing expression vectors capable of producing a desired level of gene product in a desired spatial or temporal manner, as will be discussed in more detail below.
  • 5' ESTs whose corresponding mRNAs are associated with disease states may also be identified. For example, a particular disease may result from the lack of expression, over expression, or under expression of an mRNA corresponding to a 5' EST. By comparing mRNA expression patterns and quantities in samples taken from healthy individuals with those from individuals suffering from a particular disease, 5' ESTs responsible for the disease may be identified.
  • ESTs also apply to extended cDNAs (obtainable as described below) which contain sequences adjacent to the 5' ESTs. It will also be appreciated that if desired, characterization may be delayed until extended cDNAs have been obtained rather than characterizing the
  • mRNAs corresponding to 5' ESTs or extended cDNAs may be analyzed by solution hybridization with long probes as described in International Patent Application No. WO 97/05277, the entire contents of which are hereby inco ⁇ orated by reference. Briefly, a 5' EST, extended cDNA, or fragment thereof corresponding to the gene encoding the mRNA to be characterized is inserted at a cloning site immediately downstream of a bacteriophage (T3, T7 or SP6) RNA polymerase promoter to produce antisense RNA. Preferably, the 5' EST or extended cDNA has 100 or more nucleotides.
  • the plasmid is linearized and transcribed in the presence of ribonucleotides comprising modified ribonucleotides (i.e. biotin-UTP and DIG- UTP).
  • ribonucleotides comprising modified ribonucleotides (i.e. biotin-UTP and DIG- UTP).
  • An excess of this doubly labeled RNA is hybridized in solution with mRNA isolated from cells or tissues of interest.
  • the hybridizations are performed under standard stringent conditions (40-50°C for 16 hours in an 80% formamide, 0.4 M NaCl buffer, pH 7-8).
  • the unhybridized probe is removed by digestion with ribonucleases specific for single-stranded RNA (i.e. RNases CL3, Tl, Phy M U2 or A).
  • the presence of the biotin-UTP modification enables capture of the hybrid on a microtitration plate coated with streptavidin.
  • the presence of the DIG modification enables the hybrid to be detected and quantified by ELISA using an anti-DIG antibody coupled to alkaline phosphatase.
  • the 5' ESTs, extended cDNAs, or fragments thereof may also be tagged with nucleotide sequences for the serial analysis of gene expression (SAGE) as disclosed in UK Patent Application No. 2 305 241 A, the entire contents of which are inco ⁇ orated by reference.
  • SAGE serial analysis of gene expression
  • cDNAs are prepared from a cell, tissue, organism or other source of nucleic acid for which gene expression patterns must be determined.
  • the resulting cDNAs are separated into two pools.
  • the cDNAs in each pool are cleaved with a first restriction endonuclease, called an anchoring enzyme, having a recognition site which is likely to be present at least once in most cDNAs.
  • the fragments which contain the 5' or 3' most region of the cleaved cDNA are isolated by binding to a capture medium such as streptavidin coated beads.
  • a first oligonucleotide linker having a first sequence for hybridization of an amplification primer and an internal restriction site for a so-called tagging endonuclease is ligated to the digested cDNAs in the first pool. Digestion with the second endonuclease produces short tag fragments from the cDNAs.
  • a second oligonucleotide having a second sequence for hybridization of an amplification primer and an internal restriction site is ligated to the digested cDNAs in the second pool.
  • the cDNA fragments in the second pool are also digested with the tagging endonuclease to generate short tag fragments derived from the cDNAs in the second pool.
  • the tags resulting from digestion of the first and second pools with the anchoring enzyme and the tagging endonuclease are ligated to one another to produce so-called ditags.
  • the ditags are concatamerized to produce ligation products containing from 2 to 200 ditags.
  • the tag sequences are then determined and compared to the sequences of the 5' ESTs or extended cDNAs to determine which 5' ESTs or extended cDNAs are expressed in the cell, tissue, organism, or other source of nucleic acids from which the tags were derived. In this way, the expression pattern of the 5' ESTs or extended cDNAs in the cell, tissue, organism, or other source of nucleic acids is obtained.
  • arrays means a one dimensional, two dimensional, or multidimensional arrangement of full length cDNAs (i.e. extended cDNAs which include the coding sequence for the signal peptide, the coding sequence for the mature protein, and a stop codon), extended cDNAs, 5' ESTs or fragments thereof of sufficient length to permit specific detection of gene expression.
  • the fragments are at least 15 nucleotides in length. More preferably, the fragments are at least 100 nucleotide long. More preferably, the fragments are more than 100 nucleotides in length. In some embodiments, the fragments may be more than 500 nucleotide long.
  • cDNAs as defined below, extended cDNAs, 5' ESTs, or fragments thereof in a complementary DNA microarray as described by Schena et al. (Science 270:467-470, 1995; Proc. Natl. Acad Sci. U.S.A. 93:10614-10619, 1996).
  • Full length cDNAs, extended cDNAs, 5' ESTs or fragments thereof are amplified by PCR and arrayed from 96-well microtiter plates onto sOylated microscope slides using high-speed robotics.
  • Printed arrays are incubated in a humid chamber to allow rehydration of the array elements and rinsed, once in 0.2% SDS for 1 min, twice in water for 1 min and once for 5 min in sodium borohydride solution. The arrays are submerged in water for 2 min at 95°C, transferred into 0.2% SDS for 1 min, rinsed twice with water, air dried and stored in the dark at 25°C.
  • Cell or tissue mRNA is isolated or commercially obtained and probes are prepared by a single round of reverse transcription. Probes are hybridized to 1 cm 2 icroarrays under a 14 x 14 mm glass coverslip for 6-12 hours at 60°C.
  • Arrays are washed for 5 min at 25°C in low stringency wash buffer (1 x SSC/0.2%> SDS), then for 10 min at room temperature in high stringency wash buffer (0.1 x SSC/0.2% SDS). Arrays are scanned in 0.1 x SSC using a fluorescence laser scarining device fitted with a custom filter set. Accurate differential expression measurements are obtained by taking the average of the ratios of two independent hybridizations.
  • Quantitative analysis of the expression of genes may also be performed with full length cDNAs, extended cDNAs, 5' ESTs, or fragments thereof in complementary DNA arrays as described by Pietu et al. (Genome Research 6:492-503, 1996).
  • the full length cDNAs, extended cDNAs, 5' ESTs or fragments thereof are PCR amplified and spotted on membranes.
  • mRNAs originating from various tissues or cells are labeled with radioactive nucleotides. After hybridization and washing in controlled conditions, the hybridized mRNAs are detected by phospho-imaging or autoradiography. Duplicate experiments are performed and a quantitative analysis of differentially expressed mRNAs is then performed.
  • expression analysis of the 5' ESTs or extended cDNAs can be done through high density nucleotide arrays as described by Lockhart et al. (Nature Biotechnology 14: 1675-1680, 1996) and Sosnowsky et al (Proc. Natl. Acad Sci. 94:1119-1123, 1997).
  • Oligonucleotides of 15-50 nucleotides corresponding to sequences of the 5' ESTs or extended cDNAs are synthesized directly on the chip (Lockhart et al, supra) or synthesized and then addressed to the chip (Sosnowsky et al, supra).
  • the oligonucleotides are about 20 nucleotides in length.
  • cDNA probes labeled with an appropriate compound such as biotin, digoxigenin or fluorescent dye, are synthesized from the appropriate mRNA population and then randomly fragmented to an average size of 50 to 100 nucleotides. The said probes are then hybridized to the chip. After washing as described in Lockhart et al, supra and application of different electric fields (Sonowsky et al, supra , the dyes or labeling compounds are detected and quantified. Duplicate hybridizations are performed.
  • the extended cDNAs may include the entire coding sequence of the protein encoded by the corresponding mRNA, including the authentic translation start site, the signal sequence, and the sequence encoding the mature protein remaining after cleavage of the signal peptide. Such extended cDNAs are referred to herein as "full length cDNAs.” Alternatively, the extended cDNAs may include only the sequence encoding the mature protein remaining after cleavage of the signal peptide, or only the sequence encoding the signal peptide.
  • Example 27 describes a general method for obtaining extended cDNAs using 5' ESTs.
  • Example 28 below provides experimental results, using the method explained in example 27, describing several extended cDNAs including the entire coding sequence and authentic 5' end of the corresponding mRNA for several secreted proteins.
  • Examples 27, 28, and 29 can also be used to obtain extended cDNAs which encode less than the entire coding sequence of the secreted proteins encoded by the genes corresponding to the 5' ESTs.
  • the extended cDNAs isolated using these methods encode at least 10 amino acids of one of the proteins encoded by the sequences of SEQ ID NOs: 38-305.
  • the extended cDNAs encode at least 20 amino acids of the proteins encoded by the sequences of SEQ ED NOs: 38-305.
  • the extended cDNAs encode at least 30 amino amino acids of the sequences of SEQ ID NOs: 38-305.
  • the extended cDNAs encode a full length protein sequence, which includes the protein coding sequences of SEQ ED NOs: 38-305.
  • the method takes advantage of the known 5' sequence of the mRNA.
  • a reverse transcription reaction is conducted on purified mRNA with a poly 14dT primer containing a 49 nucleotide sequence at its 5' end allowing the addition of a known sequence at the end of the cDNA which corresponds to the 3' end of the mRNA.
  • the primer may have the following sequence: 5'-ATC GTT GAG ACT CGT ACC AGC AGA GTC ACG AGA GAG ACT ACA CGG TAC TGG TTT TTT TTT TTT TTVN -3' (SEQ ID NO: 14).
  • sequences may also be added to the poly dT sequence and used to prime the first strand synthesis.
  • RNAs Using this primer and a reverse transcriptase such as the Superscript II (Gibco BRL) or Rnase H Minus M-MLV (Promega) enzyme, a reverse transcript anchored at the 3' polyA site of the RNAs is generated.
  • a reverse transcriptase such as the Superscript II (Gibco BRL) or Rnase H Minus M-MLV (Promega) enzyme
  • a pair of nested primers on each end is designed based on the known 5' sequence from the 5' EST and the known 3' end added by the poly dT primer used in the first strand synthesis.
  • Softwares used to design primers are either based on GC content and melting temperatures of oligonucleotides, such as OSP (Illier and Green, PCR Melh. Appl. 1:124- 128, 1991), or based on the octamer frequency disparity method (Griffais et al., Nucleic Acids Res. 19: 3887-3891, 1991) such as PC-Rare
  • the nested primers at the 5' end are separated from one another by four to nine bases.
  • the 5' primer sequences may be selected to have melting temperatures and specificities suitable for use in PCR.
  • the nested primers at the 3' end are separated from one another by four to nine bases.
  • the nested 3' primers may have the following sequences: (5'- CCA GCA GAG TCA CGA GAG AGA CTA CAC GG -3 '(SEQ ID NO: 15), and 5'- CAC GAG AGA GAC TAC ACG GTA CTG G -3' (SEQ ID NO: 16). These primers were selected because they have melting temperatures and specificities compatible with their use in PCR. However, those skilled in the art will appreciate that other sequences may also be used as primers.
  • the first PCR run of 25 cycles is performed using the Advantage Tth Polymerase Mix (Clontech) and the outer primer from each of the nested pairs.
  • a second 20 cycle PCR using the same enzyme and the inner primer from each of the nested pairs is then performed on 1/2500 of the first PCR product. Thereafter, the primers and nucleotides are removed.
  • the second 5' primer is located upstream of the translation initiation codon thus yielding a nested PCR product containing the whole coding sequence.
  • Such a full length extended cDNA undergoes a direct cloning procedure as described in section a.
  • the second 5' primer is located downstream of the translation initiation codon, thereby yielding a PCR product containing only part of the ORF.
  • Such incomplete PCR products are submitted to a modified procedure described in section b. a) Nested PCR products containing complete ORFs
  • the resulting nested PCR product contains the complete coding sequence, as predicted from the 5'EST sequence, it is cloned in an appropriate vector such as pED6dpc2, as described in section 3.
  • pED6dpc2 an appropriate vector
  • Nested PCR products containing incomplete ORFs When the amplicon does not contain the complete coding sequence, intermediate steps are necessary to obtain both the complete coding sequence and a PCR product containing the full coding sequence.
  • the complete coding sequence can be assembled from several partial sequences determined directly from different PCR products as described in the following section.
  • Sequencing of extended cDNAs is performed using a Die Terminator approach with the AmpliTaq DNA polymerase FS kit available from Perkin Elmer.
  • primer walking is performed using software such as OSP to choose primers and automated computer software such as ASMG (Sutton et al., Genome Science Technol. 1: 9-19, 1995) to construct contigs of walking sequences including the initial 5' tag using minimum overlaps of 32 nucleotides.
  • ASMG Sutton et al., Genome Science Technol. 1: 9-19, 1995
  • primer walking is performed until the sequences of full length cDNAs are obtained.
  • the length of the sequence determined from the computer analysis is not at least 70% of the length of the nested PCR product, these PCR products are cloned and the sequence of the insertion is determined.
  • the size of the mRNA detected for a given PCR product is used to finally assess that the sequence is complete. Sequences which do not fulfill the above criteria are discarded and will undergo a new isolation procedure.
  • Sequence data of all extended cDNAs are then transferred to a proprietary database, where quality controls and validation steps are carried out as described in example 15.
  • the PCR product containing the full coding sequence is then cloned in an appropriate vector.
  • the extended cDNAs can be cloned into the expression vector pED6dpc2 (DiscoverEase, Genetics Institute, Cambridge, MA) as follows.
  • pED6dpc2 vector DNA is prepared with blunt ends by performing an EcoRI digestion followed by a fill in reaction.
  • the blunt ended vector is dephosphorylated.
  • the PCR product containing the full coding sequence or the extended cDNA obtained as described above is phosphorylated with a kinase subsequently removed by phenol-Sevag extraction and precipitation.
  • the double stranded extended cDNA is then ligated to the vector and the resulting expression plasmid introduced into appropriate host cells.
  • the orientation of several clones for each PCR product is determined. Then, 4 to 10 clones are ordered in microtiter plates and subjected to a PCR reaction using a first primer located in the vector close to the cloning site and a second primer located in the portion of the extended cDNA corresponding to the 3' end of the mRNA.
  • This second primer may be the antisense primer used in anchored PCR in the case of direct cloning (case a) or the antisense primer located inside the 3 'UTR in the case of indirect cloning (case b).
  • Clones in which the start codon of the extended cDNA is operably linked to the promoter in the vector so as to permit expression of the protein encoded by the extended cDNA are conserved and sequenced.
  • approximately 50 bp of vector DNA on each side of the cDNA insert are also sequenced.
  • the cloned PCR products are then entirely sequenced according to the aforementioned procedure.
  • contigation of long fragments is then performed on walking sequences that have already contigated for uncloned PCR products during primer walking.
  • Sequencing of cloned amplicons is complete when the resulting contigs include the whole coding region as well as overlapping sequences with vector DNA on both ends.
  • Structural features e.g. polyA tail and polyadenylation signal, of the sequences of full length extended cDNAs are subsequently determined as follows.
  • a polyA tail is defined as a homopolymeric stretch of at least 11 A with at most one alternative base within it.
  • the polyA tail search is restricted to the last 100 nt of the sequence and limited to stretches of 11 consecutive As because sequencing reactions are often not readable after such a polyA stretch. Stretches having more than 90% homology over 8 nucleotides are identified as polyA tails using BLAST2N.
  • the polyA tail is clipped from the full- length sequence.
  • the 50 bp preceding the polyA tail are first searched for the canonic polyadenylation AAUAAA signal and, if the canonic signal is not detected, for the alternative AUUAAA signal (Sheets et al, Nuc. Acids Res. 18: 5799-5805, 1990). If neither of these consensus polyadenylation signals is found, the canonic motif is searched again allowing one mismatch to account for possible sequencing errors. More than 85 %> of identified polyadenylation signals of either type actually ends 10 to 30 bp from the polyA tail.
  • Alternative AUUAAA signals represents approximately 15 % of the total number of identified polyadenylation signals.
  • ORFs and signal sequences of the sequences of full length extended cDNAs were subsequently determined as follows.
  • the 3 upper strand frames of extended cDNAs are searched for ORFs defined as the maximum length fragments beginning with a translation intiation codon and ending with a stop codon.
  • ORFs encoding at least 20 amino acids are preferred.
  • Each found ORF is then scanned for the presence of a signal peptide in the first 50 amino-acids or, where appropriate, within shorter regions down to 20 amino acids or less in the ORF, using the matrix method of von Heijne (Niic. Acids Res. 14: 4683-4690, 1986), the disclosure of which is inco ⁇ orated herein by reference as described in Example 22.
  • von Heijne Neic. Acids Res. 14: 4683-4690, 1986
  • Categorization of full-length sequences may be achieved using procedures essentially similar to those described for 5 'ESTs in Example 24.
  • Extended cDNAs prepared as described above may be subsequently engineered to obtain nucleic acids which include desired portions of the extended cDNA using conventional techniques such as subcloning, PCR, or in vitro oligonucleotide synthesis.
  • nucleic acids which include only the full coding sequences i.e. the sequences encoding the signal peptide and the mature protein remaining after the signal peptide is cleaved oft
  • nucleic acids which include only the full coding sequences i.e. the sequences encoding the signal peptide and the mature protein remaining after the signal peptide is cleaved oft
  • conventional techniques may be applied to obtain nucleic acids which contain only the coding sequences for the mature protein remaining after the signal peptide is cleaved off or nucleic acids which contain only the coding sequences for the signal peptides.
  • nucleic acids containing any other desired portion of the coding sequences for the secreted protein may be obtained.
  • the nucleic acid may contain at least 10 consecutive bases of an extended cDNA such as one of the extended cDNAs described below.
  • the nucleic acid may contain at least 15 consecutive bases of an extended cDNA such as one of the extended cDNAs described below.
  • the nucleic acid may contain at least 20 consecutive bases of an extended cDNA such as one of the extended cDNAs described below.
  • nucleic acid may contain at least 25 consecutive bases of an extended cDNA such as one of the extended cDNAs described below.
  • the nucleic acid may contain at least 40 consecutive bases of an extended cDNA such as one of the extended cDNAs described below.
  • an extended cDNA Once an extended cDNA has been obtained, it can be sequenced to determine the amino acid sequence it encodes. Once the encoded amino acid sequence has been determined, one can create and identify any of the many conceivable cDNAs that will encode that protein by simply using the degeneracy of the genetic code. For example, allelic variants or other homologous nucleic acids can be identified as described below. Alternatively, nucleic acids encoding the desired amino acid sequence can be synthesized in vitro.
  • the coding sequence may be selected using the known codon or codon pair preferences for the host organism in which the cDNA is to be expressed.
  • the full length cDNA of SEQ ID NO: 19 (internal identification number 58-34-2-E7- FL2) was also obtained using this procedure.
  • This cDNA falls into the "EST-ext" category described above and encodes the signal peptide MWWFQQGLSFLPSALVIWTSA (SEQ ID NO:20) having a von Heijne score of 5.5.
  • Another f ll length cDNA obtained using the procedure described above has the sequence of SEQ ED NO:21 (internal identification number 51-27-1-E8-FL1).
  • This cDNA falls into the "EST-ext” category described above and encodes the signal peptide MVLTTLPSANSANSPVNMPTTGPNSLSYASSALSPCLT (SEQ ED NO:22) having a von Heijne score of 5.9.
  • the above procedure was also used to obtain a full length cDNA having the sequence of SEQ ED NO:23 (internal identification number 76-4-1-G5-FL1).
  • This cDNA falls into the "EST-ext” category described above and encodes the signal peptide ILSTVTALTFAXA (SEQ ID NO:24) having a von Heijne score of 5.5.
  • the full length cDNA of SEQ ID NO:25 (internal identification number 51-3-3-B10- FL3) was also obtained using this procedure.
  • This cDNA falls into the "new" category described above and encodes a signal peptide LVLTLCTLPLAVA (SEQ ED NO: 26) having a von Heijne score of 10.1.
  • the full length cDNA of SEQ ED NO:27 (internal identification number 58-35-2-F10- FL2) was also obtained using this procedure.
  • This cDNA falls into the "new" category described above and encodes a signal peptide LWLLFFLVTALHA (SEQ ED NO:28) having a von Heijne score of 10.7.
  • Bacterial clones containing plasmids containing the full length cDNAs described above are presently stored in the inventor's laboratories under the internal identification numbers provided above.
  • the inserts may be recovered from the stored materials by growing an aliquot of the appropriate bacterial clone in the appropriate medium.
  • the plasmid DNA can then be isolated using plasmid isolation procedures familiar to those skilled in the art such as alkaline lysis minipreps or large scale alkaline lysis plasmid isolation procedures. If desired the plasmid DNA may be further enriched by centrifugation on a cesium chloride gradient, size exclusion chromatography, or anion exchange chromatography.
  • the plasmid DNA obtained using these procedures may then be manipulated using standard cloning techniques familiar to those skilled in the art.
  • a PCR can be done with primers designed at both ends of the cDNA insertion.
  • the PCR product which corresponds to the cDNA can then be manipulated using standard cloning techniques familiar to those skilled in the art.
  • polypeptides encoded by the extended cDNAs may be screened for the presence of known structural or functional motifs or for the presence of signatures, small amino acid sequences which are well conserved amongst the members of a protein family.
  • the conserved regions have been used to derive consensus patterns or matrices included in the PROSITE data bank, in particular in the file prosite.dat (Release 13.0 of November 1995, located at http://expasy.hcuge.ch/sprot/prosite.html.
  • Prosite_convert and prosite_scan programs http://ulrec3.unil.ch/ftpserveur/prosite_scan) may be used to find signatures on the extended cDNAs.
  • the accuracy of the detection on a new protein sequence may be assessed by evaluating the frequency of irrelevant hits on the population of human secreted proteins included in the data bank SWISSPROT.
  • the ratio between the number of hits on shuffled proteins (with a window size of 20 amino acids) and the number of hits on native (unshuffled) proteins may be used as an index. Every pattern for which the ratio is greater than 20% (one hit on shuffled proteins for 5 hits on native proteins) may be skipped during the search with prosite_scan.
  • db_shuffled The program used to shuffle protein sequences (db_shuffled) and the program used to determine the statistics for each pattern in the protein data banks (prosite_statistics) are available on the ftp site http://ulrec3.unil.ch/ftpserveur/prosite_scan.
  • PCR based methods for obtaining extended cDNAs traditional hybridization based methods may also be employed. These methods may also be used to obtain the genomic DNAs which encode the mRNAs from which the 5' ESTs were derived, mRNAs corresponding to the extended cDNAs, or nucleic acids which are homologous to extended cDNAs or 5' ESTs.
  • Example 29 below provides examples of such methods.
  • a full length cDNA library can be made using the strategies described in Examples 13, 14, 15, and 16 above by replacing the random nonamer used in Example 14 with an oligo- dT primer.
  • the oligonucleotide of SEQ ED NO: 14 may be used.
  • a cDNA library or genomic DNA library may be obtained from a commercial source or made using techniques familiar to those skilled in the art. Such cDNA or genomic DNA librairies may be used to isolate extended cDNAs obtained from 5' EST or nucleic acids homologous to extended cDNAs or 5' EST as follows.
  • the cDNA library or genomic DNA library is hybridized to a detectable probe comprising at least 10 consecutive nucleotides from the 5' EST or extended cDNA using conventional techniques.
  • the probe comprises at least 12, 15, or 17 consecutive nucleotides from the 5' EST or extended cDNA. More preferably, the probe comprises at least 20 to 30 consecutive nucleotides from the 5' EST or extended cDNA. In some embodiments, the probe comprises more than 30 nucleotides from the 5 ' EST or extended cDNA.
  • cDNA or genomic DNA clones which hybridize to the detectable probe are identified and isolated for further manipulation as follows.
  • a probe comprising at least 10 consecutive nucleotides from the 5' EST or extended cDNA is labeled with a detectable label such as a radioisotope or a fluorescent molecule.
  • the probe comprises at least 12, 15, or 17 consecutive nucleotides from the 5' EST or extended cDNA. More preferably, the probe comprises 20 to 30 consecutive nucleotides from the 5' EST or extended cDNA. In some embodiments, the probe comprises more than 30 nucleotides from the 5' EST or extended cDNA.
  • cDNAs or genomic DNAs in the library are transferred to a nitrocellulose or nylon filter and denatured. After blocking of non specific sites, the filter is incubated with the labeled probe for an amount of time sufficient to allow binding of the probe to cDNAs or genomic DNAs containing a sequence capable of hybridizing thereto.
  • extended cDNAS having different levels of homology to the probe can be identified and isolated as described below.
  • Prehybridization may be carried out in 6X SSC, 5X Denhardt's reagent, 0.5% SDS, 100 ⁇ g denatured fragmented salmon sperm DNA or 6X SSC, 5X Denhardt's reagent, 0.5% SDS, 100 ⁇ g denatured fragmented salmon sperm DNA, 50%> formamide.
  • 6X SSC 6X SSC
  • 5X Denhardt's reagent 0.5% SDS
  • 100 ⁇ g denatured fragmented salmon sperm DNA 50%> formamide.
  • the formulas for SSC and Denhardt's solutions are listed in Sambrook et al. , supra.
  • Hybridization is conducted by adding the detectable probe to the prehybridization solutions listed above. Where the probe comprises double stranded DNA, it is denatured before addition to the hybridization solution. The filter is contacted with the hybridization solution for a sufficient period of time to allow the probe to hybridize to extended cDNAs or genomic DNAs containing sequences complementary thereto or homologous thereto. For probes over 200 nucleotides in length, the hybridization may be carried out at 15-25°C below the Tm. For shorter probes, such as oligonucleotide probes, the hybridization may be conducted at 15-25°C below the Tm. Preferably, for hybridizations in 6X SSC, the hybridization is conducted at approximately 68°C. Preferably, for hybridizations in 50% formamide containing solutions, the hybridization is conducted at approximately 42°C.
  • the filter is washed in 2X SSC, 0.1% SDS at room temperature for 15 minutes.
  • the filter is then washed with 0.1X SSC, 0.5% SDS at room temperature for 30 minutes to 1 hour. Thereafter, the solution is washed at the hybridization temperature in 0.1X SSC, 0.5% SDS.
  • a final wash is conducted in 0.1X SSC at room temperature.
  • Extended cDNAs nucleic acids homologous to extended cDNAs or 5' ESTs, or genomic DNAs which have hybridized to the probe are identified by autoradiography or other conventional techniques. 2. Obtention of Extended cDNA or Genomic cDNA Sequences Having Lower Degrees of Homology to the Labeled Probe
  • the above procedure may be modified to identify extended cDNAs, nucleic acids homologous to extended cDNAs, or genomic DNAs having decreasing levels of homology to the probe sequence.
  • less stringent conditions may be used.
  • the hybridization temperature may be decreased in increments of 5°C from 68°C to 42°C in a hybridization buffer having a sodium concentration of approximately IM.
  • the filter may be washed with 2X SSC, 0.5% SDS at the temperature of hybridization. These conditions are considered to be "moderate” conditions above 50°C and "low” conditions below 50°C.
  • the hybridization may be carried out in buffers, such as 6X SSC, containing formamide at a temperature of 42°C.
  • concentration of formamide in the hybridization buffer may be reduced in 5% increments from 50% to 0%o to identify clones having decreasing levels of homology to the probe.
  • the filter may be washed with 6X SSC, 0.5% SDS at 50°C. These conditions are considered to be “moderate” conditions above 25% formamide and "low” conditions below 25%> formamide.
  • Extended cDNAs, nucleic acids homologous to extended cDNAs, or genomic DNAs which have hybridized to the probe are identified by autoradiography.
  • the level of homology between the hybridized nucleic acid and the extended cDNA or 5' EST used as the probe may be further determined using BLAST2N; parameters may be adapted depending on the sequence length and degree of homology studied.
  • the nucleotide sequences of the hybridized nucleic acid and the extended cDNA or 5'EST from which the probe was derived are compared.
  • nucleic acids having at least 95%> nucleic acid homology to the extended cDNA or 5'EST from which the probe was derived may be obtained and identified.
  • nucleic acids having at least 90%, at least 85%, at least 80%) or at least 75% homology to the extended cDNA or 5'EST from which the probe was derived may be obtained and identified.
  • the amino acid sequence encoded by the extended cDNA or 5' EST is compared to the amino acid sequence encoded by the hybridizing nucleic acid. Homology is determined to exist when an amino acid sequence in the extended cDNA or 5' EST is closely related to an amino acid sequence in the hybridizing nucleic acid. A sequence is closely related when it is identical to that of the extended cDNA or 5' EST or when it contains one or more amino acid substitutions therein in which amino acids having similar characteristics have been substituted for one another.
  • Extended cDNAs may be prepared by obtaining mRNA from the tissue, cell, or organism of interest using mRNA preparation procedures utilizing polyA selection procedures or other techniques known to those skilled in the art.
  • a first primer capable of hybridizing to the polyA tail of the mRNA is hybridized to the mRNA and a reverse transcription reaction is performed to generate a first cDNA strand.
  • the first cDNA strand is hybridized to a second primer containing at least 10 consecutive nucleotides of the sequences of SEQ ED NOs 38-305.
  • the primer comprises at least 12, 15, or 17 consecutive nucleotides from the sequences of SEQ ED NOs 38-305. More preferably, the primer comprises 20 to 30 consecutive nucleotides from the sequences of SEQ ED NOs 38-305. In some embodiments, the primer comprises more than 30 nucleotides from the sequences of SEQ ED NOs 38-305.
  • the second primer used contains sequences located upstream of the translation initiation site. The second primer is extended to generate a second cDNA strand complementary to the first cDNA strand.
  • RT-PCR may be performed as described above using primers from both ends of the cDNA to be obtained.
  • Extended cDNAs containing 5' fragments of the mRNA may be prepared by hybridizing an mRNA comprising the sequence of the 5'EST for which an extended cDNA is desired with a primer comprising at least 10 consecutive nucleotides of the sequences complementary to the 5'EST and reverse transcribing the hybridized primer to make a first cDNA strand from the mRNAs.
  • the primer comprises at least 12, 15, or 17 consecutive nucleotides from the 5'EST. More preferably, the primer comprises 20 to 30 consecutive nucleotides from the 5'EST.
  • a second cDNA strand complementary to the first cDNA strand is synthesized.
  • the second cDNA strand may be made by hybridizing a primer complementary to sequences in the first cDNA strand to the first cDNA strand and extending the primer to generate the second cDNA strand.
  • the double stranded extended cDNAs made using the methods described above are isolated and cloned.
  • the extended cDNAs may be cloned into vectors such as plasmids or viral vectors capable of replicating in an appropriate host cell.
  • the host cell may be a bacterial, mammalian, avian, or insect cell.
  • full length or extended cDNAs are prepared from mRNA and cloned into double stranded phagemids as follows.
  • the cDNA library in the double stranded phagemids is then rendered single stranded by treatment* with an endonuclease, such as the Gene El product of the phage FI, and an exonuclease (Chang et al, Gene 127:95-8, 1993).
  • a biotinylated oligonucleotide comprising the sequence of a 5' EST, or a fragment containing at least 10 nucleotides thereof, is hybridized to the single stranded phagemids.
  • the fragment comprises at least 12, 15, or 17 consecutive nucleotides from the 5' EST. More preferably, the fragment comprises 20-30 consecutive nucleotides from the 5' EST. In some procedures, the fragment may comprise more than 30 consecutive nucleotides from the 5' EST.
  • Hybrids between the biotinylated oligonucleotide and phagemids having inserts containing the 5' EST sequence are isolated by incubating the hybrids with streptavidin coated paramagnetic beads and retrieving the beads with a magnet (Fry et al, Biotechniques, 13: 124-131, 1992). Therafter, the resulting phagemids containing the 5' EST sequence are released from the beads and converted into double stranded DNA using a primer specific for the 5' EST sequence. Alternatively, protocoles such as the Gene Trapper kit (Gibco BRL) may be used. The resulting double stranded DNA is transformed into bacteria. Extended cDNAs containing the 5' EST sequence are identified by colony PCR or colony hybridization.
  • a plurality of extended cDNAs containing full length protein coding sequences or sequences encoding only the mature protein remaining after the signal peptide is cleaved off may be provided as cDNA libraries for subsequent evaluation of the encoded proteins or use in diagnostic assays as described below.
  • Extended cDNAs containing the full protein coding sequences of their corresponding mRNAs or portions thereof may be used to express the encoded secreted proteins or portions thereof as described in Example 30 below.
  • the extended cDNAs may contain the sequences encoding the signal peptide to facilitate secretion of the expressed protein. It will be appreciated that a plurality of extended cDNAs containing the full protein coding sequences or portions thereof may be simultaneously cloned into expression vectors to create an expression library for analysis of the encoded proteins as described below.
  • nucleic acids may contain the sequences encoding the signal peptide to facilitate secretion of the expressed protein.
  • the nucleic acids inserted into the expression vectors may also contain sequences upstream of the sequences encoding the signal peptide, such as sequences which regulate expression levels or sequences which confer tissue specific expression.
  • the nucleic acid encoding the protein or polypeptide to be expressed is operably linked to a promoter in an expression vector using conventional cloning technology.
  • the expression vector may be any of the mammalian, yeast, insect or bacterial expression systems known in the art. Commercially available vectors and expression systems are available from a variety of suppliers including Genetics Institute (Cambridge, MA), Stratagene (La Jolla, California), Promega (Madison, Wisconsin), and Invitrogen (San Diego, California). If desired, to enhance expression and facilitate proper protein folding, the codon context and codon pairing of the sequence may be optimized for the particular expression organism in which the expression vector is introduced, as explained by Hatfield, et al, U.S. Patent No. 5,082,767, inco ⁇ orated herein by this reference.
  • the cDNA cloned into the expression vector may encode the entire protein (i.e. the signal peptide and the mature protein), the mature protein (i.e. the protein created by cleaving the signal peptide off), only the signal peptide or any other portion thereof.
  • the following is provided as one exemplary method to express the proteins encoded by the extended cDNAs corresponding to the 5' ESTs or the nucleic acids described above. First, the methionine initiation codon for the gene and the polyA signal of the gene are identified. If the nucleic acid encoding the polypeptide to be expressed lacks a methionine to serve as the initiation site, an initiating methionine can be introduced next to the first codon of the nucleic acid using conventional techniques.
  • this sequence can be added to the construct by, for example, splicing out the polyA signal from pSG5 (Stratagene) using Bglll and Sail restriction endonuclease enzymes and inco ⁇ orating it into the mammalian expression vector pXTl (Stratagene).
  • pXTl contains the LTRs and a portion of the gag gene from Moloney Murine Leukemia Virus. The position of the LTRs in the construct allow efficient stable transfection.
  • the vector includes the He ⁇ es Simplex thymidine kinase promoter and the selectable neomycin gene.
  • the extended cDNA or portion thereof encoding the polypeptide to be expressed is obtained by PCR from the bacterial vector using oligonucleotide primers complementary to the extended cDNA or portion thereof and containing restriction endonuclease sequences for Pst I inco ⁇ orated into the 5'primer and Bglll at the 5' end of the corresponding cDNA 3' primer, taking care to ensure that the extended cDNA is positioned with the poly A signal.
  • the purified fragment obtained from the resulting PCR reaction is digested with Pstl, blunt ended with an exonuclease, digested with Bgl II, purified and ligated to pXTl containing a poly A signal and prepared for this ligation (blunt/Bglll).
  • the ligated product is transfected into mouse NLH 3T3 cells using Lipofectin (Life Technologies, Inc., Grand Island, New York) under conditions outlined in the product specification. Positive transfectants are selected after growing the transfected cells in 600 ⁇ g/ml G418 (Sigma, St. Louis, Missouri). Preferably the expressed protein is released into the culture medium, thereby facilitating purification.
  • the extended cDNAs may be cloned into pED6dpc2 as described above.
  • the resulting pED6dpc2 constructs may be transfected into a suitable host cell, such as COS 1 cells. Methotrexate resistant cells are selected and expanded.
  • the protein expressed from the extended cDNA is released into the culture medium thereby facilitating purification.
  • Proteins in the culture medium are separated by gel electrophoresis. If desired, the proteins may be ammonium sulfate precipitated or separated based on size or charge prior to electrophoresis.
  • the expression vector lacking a cDNA insert is introduced into host cells or organisms and the proteins in the medium are harvested.
  • the secreted proteins present in the medium are detected using techniques familiar to those skilled in the art such as Coomassie blue or silver staining or using antibodies against the protein encoded by the extended cDNA.
  • Antibodies capable of specifically recognizing the protein of interest may be generated using synthetic 15-mer peptides having a sequence encoded by the appropriate 5' EST, extended cDNA, or portion thereof.
  • the synthetic peptides are injected into mice to generate antibody to the polypeptide encoded by the 5' EST, extended cDNA, or portion thereof.
  • Secreted proteins from the host cells or organisms containing an expression vector which contains the extended cDNA derived from a 5' EST or a portion thereof are compared to those from the control cells or organism.
  • the presence of a band in the medium from the cells containing the expression vector which is absent in the medium from the control cells indicates that the extended cDNA encodes a secreted protein.
  • the band corresponding to the protein encoded by the extended cDNA will have a mobility near that expected based on the number of amino acids in the open reading frame of the extended cDNA.
  • the band may have a mobility different than that expected as a result of modifications such as glycosylation, ubiquitination, or enzymatic cleavage.
  • the proteins expressed from host cells containing an expression vector with an insert encoding a secreted protein or portion thereof can be compared to the proteins expressed in control host cells containing the expression vector without an insert.
  • the presence of a band in samples from cells containing the expression vector with an insert which is absent in samples from cells containing the expression vector without an insert indicates that the desired protein or portion thereof is being expressed.
  • the band will have the mobility expected for the secreted protein or portion thereof.
  • the band may have a mobility different than that expected as a result of modifications such as glycosylation, ubiquitination, or enzymatic cleavage.
  • the protein encoded by the extended cDNA may be purified using standard immunochromatography techniques.
  • a solution containing the secreted protein such as the culture medium or a cell extract, is applied to a column having antibodies against the secreted protein attached to the chromatography matrix.
  • the secreted protein is allowed to bind the immunochromatography column. Thereafter, the column is washed to remove non-specifically bound proteins.
  • the specifically bound secreted protein is then released from the column and recovered using standard techniques. If antibody production is not possible, the extended cDNA sequence or portion thereof may be inco ⁇ orated into expression vectors designed for use in purification schemes employing chimeric polypeptides.
  • the coding sequence of the extended cDNA or portion thereof is inserted in frame with the gene encoding the other half of the chimera.
  • the other half of the chimera may be ⁇ -globin or a nickel binding polypeptide.
  • a chromatography matrix having antibody to ⁇ -globin or nickel attached thereto is then used to purify the chimeric protein.
  • Protease cleavage sites may be engineered between the ⁇ -globin gene or the nickel binding polypeptide and the extended cDNA or portion thereof
  • the two polypeptides of the chimera may be separated from one another by protease digestion.
  • One useful expression vector for generating ⁇ -globin chimerics is pSG5 (Stratagene), which encodes rabbit ⁇ -globin.
  • Intron II of the rabbit ⁇ -globin gene facilitates splicing of the expressed transcript, and the polyadenylation signal inco ⁇ orated into the construct increases the level of expression.
  • Standard methods are published in methods texts such as Davis et al.., (Basic Methods in Molecular Biology, Davis, Dibner, and Battey, ed., Elsevier Press, NY, 1986) and many of the methods are available from Stratagene, Life Technologies, Inc., or Promega.
  • Polypeptide may additionally be produced from the construct using in vitro translation systems such as the In vitro ExpressTM Translation Kit (Stratagene).
  • the purified proteins may be tested for the ability to bind to the surface of various cell types as described in Example 31 below. It will be appreciated that a plurality of proteins expressed from these cDNAs may be included in a panel of proteins to be simultaneously evaluated for the activities specifically described below, as well as other biological roles for which assays for determining activity are available.
  • the proteins encoded by the 5' ESTs, extended cDNAs, or fragments thereof are cloned into expression vectors such as those described in Example 30.
  • the proteins are purified by size, charge, immunochromatography or other techniques familiar to those skilled in the art.
  • the proteins are labeled using techniques known to those skilled in the art.
  • the labeled proteins are incubated with cells or cell lines derived from a variety of organs or tissues to allow the proteins to bind to any receptor present on the cell surface. Following the incubation, the cells are washed to remove non-specifically bound protein.
  • the labeled proteins are detected by autoradiography. Alternatively, unlabeled proteins may be incubated with the cells and detected with antibodies having a detectable label, such as a fluorescent molecule, attached thereto.
  • Specificity of cell surface binding may be analyzed by conducting a competition analysis in which various amounts of unlabeled protein are incubated along with the labeled protein.
  • the amount of labeled protein bound to the cell surface decreases as the amount of competitive unlabeled protein increases.
  • various amounts of an unlabeled protein unrelated to the labeled protein is included in some binding reactions.
  • the amount of labeled protein bound to the cell surface does not decrease in binding reactions containing increasing amounts of unrelated unlabeled protein, indicating that the protein encoded by the cDNA binds specifically to the cell surface.
  • secreted proteins have been shown to have a number of important physiological effects and, consequently, represent a valuable therapeutic resource.
  • the secreted proteins encoded by the extended cDNAs or portions thereof made according to Examples 27-29 may be evaluated to determine their physiological activities as described below.
  • secreted proteins may act as cytokines or may affect cellular proliferation or differentiation.
  • Many protein factors discovered to date including all known cytokines, have exhibited activity in one or more factor dependent cell proliferation assays, and hence the assays serve as a convenient confirmation of cytokine activity.
  • the activity of a protein encoded by the extended cDNAs is evidenced by any one of a number of routine factor dependent cell proliferation assays for cell lines including, without limitation, 32D, DA2, DA1G, T10, B9, B9/11, BaF3, MC9/G, M * (preB M * ), 2E8, RB5, DAI, 123, T1165, HT2, CTLL2, TF-1, Mo7c and CMK.
  • the proteins encoded by the above extended cDNAs or portions thereof may be evaluated for their ability to regulate T cell or thymocyte proliferation in assays such as those described above or in the following references, which are inco ⁇ orated herein by reference: Current Protocols in Immunology, Ed. by Coligan et al.., Greene Publishing Associates and Wiley-Interscience; Takai et al. J. Immunol. 137:3494- 3500, 1986., Bertagnolli et al, J. Immunol. 145:1706-1712, 1990., Bertagnolli et al, Cell. Immunol. 133:327-341, 1991; Bertagnolli, et al, J.
  • the proteins encoded by the cDNAs may also be assayed for the ability to regulate the proliferation and differentiation of hematopoietic or lymphopoietic cells.
  • Many assays for such activity are familiar to those skilled in the art, including the assays in the following references, which are inco ⁇ orated herein by reference: Bottomly et al, In Current Protocols in Immunology., supra. 1 : 6.3.1-6.3.12,; deVries et al, J. Exp. Med. 173:1205-1211, 1991; Moreau et al, Nature 36:690-692, 1988; Greenberger et al, Proc. Natl. Acad Sci. U.S.A.
  • the proteins encoded by the cDNAs may also be assayed for their ability to regulate T-cell responses to antigens.
  • Many assays for such activity are familiar to those skilled in the art, including the assays described in the following references, which are inco ⁇ orated herein by reference: Chapter 3 (In Vitro Assays for Mouse Lymphocyte Function), Chapter 6 (Cytokines and Their Cellular Receptors) and Chapter 7, (Immunologic Studies in Humans) in Current Protocols in Immunology supra; Weinberger et al, Proc. Natl. Acad. Sci. USA 77:6091-6095, 1980; Weinberger et al, Eur. J. Immun. 11:405-411, 1981; Takai et al, J.
  • the proteins encoded by the cDNAs may also be evaluated for their effects as immune regulators.
  • the proteins may be evaluated for their activity to influence thymocyte or splenocyte cytotoxicity.
  • Numerous assays for such activity are familiar to those skilled in the art including the assays described in the following references, which are inco ⁇ orated herein by reference: Chapter 3 (In Vitro Assays for Mouse Lymphocyte
  • the proteins encoded by the cDNAs may also be evaluated for their effects on T-cell dependent immunoglobulin responses and isotype switching. Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are inco ⁇ orated herein by reference: Maliszewski, J. Immunol. 144:3028-
  • the proteins encoded by the cDNAs may also be evaluated for their effect on immune effector cells, including their effect on Thl cells and cytotoxic lymphocytes.
  • Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are inco ⁇ orated herein by reference: Chapter 3 (In Vitro Assays for Mouse 1 Lymphocyte Function 3.1-3.19) and Chapter 7 (Immunologic Studies in Humans) in Current Protocols in Immunology, supra; Takai et al, J. Immunol. 137:3494-3500, 1986; Takai et ⁇ /., J. Immunol 140:508-512, 1988; Bertagnolli et l., J. Immunol.
  • the proteins encoded by the cDNAs may also be evaluated for their effect on dendritic cell mediated activation of naive T-cells.
  • Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are inco ⁇ orated herein by reference: Guery et al, J. Immunol 134:536-544, 1995; Inaba et tf/., J. Exp. Med 173:549-559, 1991; Macatonia et ⁇ /., J. Immunol 154:5071-5079, 1995; Porgador et al.J. Exp.
  • the proteins encoded by the cDNAs may also be evaluated for their influence on the lifetime of lymphocytes.
  • Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are inco ⁇ orated herein by reference: Darzynkiewicz et al, Cytometry 13:795-808, 1992; Gorczyca et al, Leukemia 7:659-670, 1993; Gorczyca etal, Cancer Res. 53:1945-1951, 1993; Ito etal, Cell 66:233- 243, 1991; Zacharchuk, J. Immunol. 145:4037-4045, 1990; Zamai et al, Cytometry 14:891- 897, 1993; Gorczyca et al, Int. J. Oncol. 1:639-648, 1992.
  • the proteins encoded by the cDNAs may also be evaluated for their influence on early steps of T-cell commitment and development.
  • Numerous assays for such activity are familiar to those skilled in the art, including without limitation the assays disclosed in the following references, which are inco ⁇ orated herein by references: Antica et al, Blood 84:111-117, 1994; Fine et al, Cell. Immunol. 155:1 11-122, 1994; Galy et al, Blood 85:2770-2778, 1995; Toki etal, Proc. Nat. Acad Sci. USA 88:7548-7551, 1991.
  • proteins which exhibit activity as immune system regulators activity may then be formulated as pharmaceuticals and used to treat clinical conditions in which regulation of immune activity is beneficial.
  • the protein may be useful in the treatment of various immune deficiencies and disorders (including severe combined immunodeficiency), e.g., in regulating (up or down) growth and proliferation of T and/or B lymphocytes, as well as effecting the cytolytic activity of NK cells and other cell populations.
  • immune deficiencies may be genetic or be caused by viral (e.g., HIV) as well as bacterial or fungal infections, or may result from autoimmune disorders.
  • infectious diseases caused by viral, bacterial, fungal or other infection may be treatable using a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention, including infections by HIV, hepatitis viruses, he ⁇ esviruses, mycobacteria, Leishmania spp., plamodium and various fungal infections such as candidiasis.
  • a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also be useful where a boost to the immune system generally may be desirable, i.e., in the treatment of cancer.
  • proteins encoded by extended cDNAs derived from the 5' ESTs of the present invention may be used in treatment of autoimmune disorders including, for example, connective tissue disease, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, autoimmune pulmonary inflammation, Guillain-Barre syndrome, autoimmune thyroiditis, insulin dependent diabetes mellitis, myasthenia gravis, graft-versus-host disease and autoimmune inflammatory eye disease.
  • autoimmune disorders including, for example, connective tissue disease, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, autoimmune pulmonary inflammation, Guillain-Barre syndrome, autoimmune thyroiditis, insulin dependent diabetes mellitis, myasthenia gravis, graft-versus-host disease and autoimmune inflammatory eye disease.
  • Such a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also to be useful in the treatment of
  • proteins of the invention it may also be possible to regulate immune responses either up or down.
  • Down regulation may involve inhibiting or blocking an immune response already in progress or may involve preventing the induction of an immune response.
  • the functions of activated T-cells may be inhibited by suppressing T cell responses or by inducing specific tolerance in T cells, or both.
  • Immunosuppression of T cell responses is generally an active non-antigen-specific process which requires continuous exposure of the T cells to the suppressive agent.
  • Tolerance which involves inducing non-responsiveness or anergy in T cells, is distinguishable from immunosuppression in that it is generally antigen-specific and persists after the end of exposure to the tolerizing agent. Operationally, tolerance can be demonstrated by the lack of a T cell response upon reexposure to specific antigen in the absence of the tolerizing agent.
  • Down regulating or preventing one or more antigen functions (including without limitation B lymphocyte antigen functions, such as, for example, B7 costimulation), e.g., preventing high level lymphokine synthesis by activated T cells, will be useful in situations of tissue, skin and organ transplantation and in graft-versus-host disease (GVHD).
  • B lymphocyte antigen functions such as, for example, B7 costimulation
  • GVHD graft-versus-host disease
  • blockage of T cell function should result in reduced tissue destruction in tissue transplantation.
  • rejection of the transplant is initiated through its recognition as foreign by T cells, followed by an immune reaction that destroys the transplant.
  • a molecule which inhibits or blocks interaction of a B7 lymphocyte antigen with its natural ligand(s) on immune cells can lead to the binding of the molecule to the natural ligand(s) on the immune cells without transmitting the corresponding costimulatory signal.
  • a B7 lymphocyte antigen e.g., B7-1, B7-3 or blocking antibody
  • Blocking B lymphocyte antigen function in this matter prevents cytokine synthesis by immune cells, such as T cells, and thus acts as an immunosuppressant.
  • the lack of costimulation may also be sufficient to anergize the T cells, thereby inducing tolerance in a subject.
  • Induction of long-term tolerance by B lymphocyte antigen-blocking reagents may avoid the necessity of repeated administration of these blocking reagents.
  • the efficacy of particular blocking reagents in preventing organ transplant rejection or GVHD can be assessed using animal models that are predictive of efficacy in humans.
  • appropriate systems which can be used include allogeneic cardiac grafts in rats and xenogeneic pancreatic islet cell grafts in mice, both of which have been used to examine the immunosuppressive effects of CTLA4Ig fusion proteins in vivo as described in Lenschow et al, Science 257:789-792, 1992 and Turka et al, Proc. Natl Acad. Sci USA, 89:11102- 11105, 1992.
  • murine models of GVHD see Paul ed., Fundamental Immunology, Raven Press, New York, 1989, pp.
  • Blocking antigen function may also be therapeutically useful for treating autoimmune diseases. Many autoimmune disorders are the result of inappropriate activation of T cells that are reactive against self tissue and which promote the production of cytokines and autoantibodies involved in the pathology of the diseases. Preventing the activation of autoreactive T cells may reduce or eliminate disease symptoms. Administration of reagents which block costimulation of T cells by disrupting receptor/ligand interactions of B lymphocyte antigens can be used to inhibit T cell activation and prevent production of autoantibodies or T cell-derived cytokines which potentially involved in the disease process.
  • blocking reagents may induce antigen-specific tolerance of autoreactive T cells which could lead to long-term relief from the disease.
  • the efficacy of blocking reagents in preventing or alleviating autoimmune disorders can be determined using a number of well- characterized animal models of human autoimmune diseases. Examples include murine experimental autoimmune encephalitis, systemic lupus erythmatosis in MRL/pr/pr mice or NZB hybrid mice, murine autoimmuno collagen arthritis, diabetes mellitus in OD mice and BB rats, and murine experimental myasthenia gravis (see Paul ed., supra, pp. 840-856).
  • Upregulation of an antigen function (preferably a B lymphocyte antigen function), as a means of up regulating immune responses, may also be useful in therapy. Upregulation of immune responses may involve either enhancing an existing immune response or eliciting an initial immune response as shown by the following examples. For instance, enhancing an immune response through stimulating B lymphocyte antigen function may be useful in cases of viral infection. In addition, systemic viral diseases such as influenza, the common cold, and encephalitis might be alleviated by the administration of stimulatory form of B lymphocyte antigens systemically.
  • antiviral immune responses may be enhanced in an infected patient by removing T cells from the patient, costimulating the T cells in vitro with viral antigen-pulsed APCs either expressing a peptide encoded by extended cDNAs derived from the 5' ESTs of the present invention or together with a stimulatory form of a soluble peptide encoded by extended cDNAs derived from the 5' ESTs of the present invention and reintroducing the in vitro primed T cells into the patient.
  • the infected cells would now be capable of delivering a costimulatory signal to T cells in vivo, thereby activating the T cells.
  • upregulation or enhancement of antigen function may be useful in the induction of tumor immunity.
  • Tumor cells e.g., sarcoma, melanoma, lymphoma, leukemia, neuroblastoma, carcinoma
  • a nucleic acid encoding at least one peptide encoded by extended cDNAs derived from the 5' ESTs of the present invention can be administered to a subject to overcome tumor-specific tolerance in the subject.
  • the tumor cell can be transfected to express a combination of peptides.
  • tumor cells obtained from a patient can be transfected ex vivo with an expression vector directing the expression of a peptide having B7-2-like activity alone, or in conjunction with a peptide having B7-l-like activity and or B7-3-like activity.
  • the transfected tumor cells are returned to the patient to result in expression of the peptides on the surface of the transfected cell.
  • gene therapy techniques can be used to target a tumor cell for transfection in vivo.
  • the presence of the peptide encoded by extended cDNAs derived from the 5' ESTs of the present invention having the activity of a B lymphocyte antigen(s) on the surface of the tumor cell provides the necessary costimulation signal to T cells to induce a T cell mediated immune response against the transfected tumor cells.
  • tumor cells which lack or which fail to reexpress sufficient amounts of MHC class I or MHC class II molecules can be transfected with nucleic acids encoding all or a portion of (e.g., a cytoplasmic-domain truncated portion) of an MHC class I ⁇ chain and ⁇ microglobulin or an MHC class II ⁇ chain and an MHC class El ⁇ chain to thereby express MHC class I or MHC class II proteins on the cell surface, respectively.
  • Expression of the appropriate MHC class I or class II molecules in conjunction with a peptide having the activity of a B lymphocyte antigen (e.g., B7-1, B7-2, B7-3) induces a T cell mediated immune response against the transfected tumor cell.
  • a B lymphocyte antigen e.g., B7-1, B7-2, B7-3
  • a gene encoding an antisense construct which blocks expression of an MHC class II associated protein, such as the invariant chain can also be cotransfected with a DNA encoding a peptide having the activity of a B lymphocyte antigen to promote presentation of tumor associated antigens and induce tumor specific immunity.
  • a T cell mediated immune response in a human subject may be sufficient to overcome tumor- specific tolerance in the subject.
  • genes encoding these immune system regulator proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
  • EXAMPLE 34 Assaying the Proteins Expressed from Extended cDNAs or Portions Thereof for Hematopoiesis Regulating Activity
  • the proteins encoded by the extended cDNAs or portions thereof may also be evaluated for their hematopoiesis regulating activity. For example, the effect of the proteins on embryonic stem cell differentiation may be evaluated.
  • Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are inco ⁇ orated herein by reference: Johansson et al Cell Biol 15: 141- 151, 1995; Keller et al, Mol Cell. Biol 13:473-486, 1993; McClanahan et al, Blood 81:2903-2915, 1993.
  • the proteins encoded by the extended cDNAs or portions thereof may also be evaluated for their influence on the lifetime of stem cells and stem cell differentiation.
  • Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are inco ⁇ orated herein by reference: Freshney, Methylcellulose Colony Forming Assays, in Culture of Hematopoietic Cells., Freshney, et al.. Eds. pp. 265-268, Wiley-Liss, Inc., New York, NY. 1994; Hirayama et al, Proc. Natl. Acad Sci.
  • proteins which exhibit hematopoiesis regulatory activity may then be formulated as pharmaceuticals and used to treat clinical conditions in which regulation of hematopoeisis is beneficial, such as in the treatment of myeloid or lymphoid cell deficiencies. Involvement in regulating hematopoiesis is indicated even by marginal biological activity in support of colony forming cells or of factor-dependent cell lines.
  • proteins supporting the growth and proliferation of erythroid progenitor cells alone or in combination with other cytokines indicates utility, for example, in treating various anemias or for use in conjunction with irradiation/chemotherapy to stimulate the production of erythroid precursors and or erythroid cells.
  • Proteins supporting the growth and proliferation of myeloid cells such as granulocytes and monocytes/macrophages (i.e., traditional CSF activity) may be useful, for example, in conjunction with chemotherapy to prevent or treat consequent myelo- suppression.
  • Proteins supporting the growth and proliferation of megakaryocytes and consequently of platelets allows prevention or treatment of various platelet disorders such as thrombocytopenia, and generally may be used in place of or complementary to platelet transfusions.
  • Proteins supporting the growth and proliferation of hematopoietic stem cells which are capable of maturing to any and all of the above-mentioned hematopoietic cells may therefore find therapeutic utility in various stem cell disorders (such as those usually treated with transplantion, including, without limitation, aplastic anemia and paroxysmal nocturnal hemoglobinuria), as well as in repopulating the stem cell compartment post irradiation/chemotherapy, either in vivo or ex vivo (i.e., in conjunction with bone marrow transplantation or with peripheral progenitor cell transplantation (homologous or heterologous)) as normal cells or genetically manipulated for gene therapy.
  • genes encoding hematopoiesis regulating activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
  • the proteins encoded by the extended cDNAs or portions thereof may also be evaluated for their effect on tissue growth.
  • Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in International Patent Publication No. WO95/16035, International Patent Publication No. WO95/05846 and International Patent
  • Assays for wound healing activity include, without limitation, those described in: Winter, Epidermal Wound Healing, pps. 71-112, Maibach and Rovee, eds., Year Book Medical Publishers, Inc., Chicago, as modified by Eaglstein and Mertz, J. Invest. Dermatol 71:382-84, 1978, which are inco ⁇ orated herein by reference. Those proteins which are involved in the regulation of tissue growth may then be formulated as pharmaceuticals and used to treat clinical conditions in which regulation of tissue growth is beneficial.
  • a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention also may have utility in compositions used for bone, cartilage, tendon, ligament and or nerve tissue growth or regeneration, as well as for wound healing and tissue repair and replacement, and in the treatment of burns, incisions and ulcers.
  • a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention which induces cartilage and/or bone growth in circumstances where bone is not normally formed, has application in the healing of bone fractures and cartilage damage or defects in humans and other animals.
  • Such a preparation employing a protein of the invention may have prophylactic use in closed as well as open fracture reduction and also in the improved fixation of artificial joints.
  • De novo bone synthesis induced by an osteogenic agent contributes to the repair of congenital, trauma induced, or oncologic resection induced craniofacial defects, and also is useful in cosmetic plastic surgery.
  • a protein of this invention may also be used in the treatment of periodontal disease, and in other tooth repair processes.
  • Such agents may provide an environment to attract bone- forming cells, stimulate growth of bone-forming cells or induce differentiation of bone- forming cell progenitors.
  • a protein of the invention may also be useful in the treatment of osteoporosis or osteoarthritis, such as through stimulation of bone and/or cartilage repair or by blocking inflammation or processes of tissue destruction (coUagenase activity, osteoclast activity, etc.) mediated by inflammatory processes.
  • tissue regeneration activity that may be attributable to the protein encoded by extended cDNAs derived from the 5' ESTs of the present invention is tendon/ligament formation.
  • a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention which induces tendon/ligament-like tissue or other tissue formation in circumstances where such tissue is not normally formed, has application in the healing of tendon or ligament tears, deformities and other tendon or ligament defects in humans and other animals.
  • Such a preparation employing a tendon/ligament-like tissue inducing protein may have prophylactic use in preventing damage to tendon or ligament tissue, as well as use in the improved fixation of tendon or ligament to bone or other tissues, and in repairing defects to tendon or ligament tissue.
  • compositions encoded by extended cDNAs derived from the 5' ESTs of the present invention contributes to the repair of tendon or ligaments defects of congenital, traumatic or other origin and is also useful in cosmetic plastic surgery for attachment or repair of tendons or ligaments.
  • the compositions encoded by extended cDNAs derived from the 5' ESTs of the present invention may provide an environment to attract tendon- or ligament- forming cells, stimulate growth of tendon- or ligament-forming cells, induce differentiation of progenitors of tendon- or ligament-forming cells, or induce growth of tendon/ligament cells or progenitors ex vivo for return in vivo to effect tissue repair.
  • the compositions of the invention may also be useful in the treatment of tendinitis, ca ⁇ al tunnel syndrome and other tendon or ligament defects.
  • the compositions may also include an appropriate matrix and/or sequestering agent as a carrier as is well known in the art.
  • the protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also be useful for proliferation of neural cells and for regeneration of nerve and brain tissue, i.e., for the treatment of central and peripheral nervous system diseases and neuropathies, as well as mechanical and traumatic disorders, which involve degeneration, death or trauma to neural cells or nerve tissue. More specifically, a protein may be used in the treatment of diseases of the peripheral nervous system, such as peripheral nerve injuries, peripheral neuropathy and localized neuropathies, and central nervous system diseases, such as Alzheimer's, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome.
  • diseases of the peripheral nervous system such as peripheral nerve injuries, peripheral neuropathy and localized neuropathies
  • central nervous system diseases such as Alzheimer's, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome.
  • Further conditions which may be treated in accordance with the present invention include mechanical and traumatic disorders, such as spinal cord disorders, head trauma and cerebrovascular diseases such as stroke.
  • Peripheral neuropathies resulting from chemotherapy or other medical therapies may also be treatable using a protein of the invention. Proteins of the invention may also be useful to promote better or faster closure of non-healing wounds, including without limitation pressure ulcers, ulcers associated with vascular insufficiency, surgical and traumatic wounds, and the like.
  • a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also exhibit activity for generation or regeneration of other tissues, such as organs (including, for example, pancreas, liver, intestine, kidney, skin, endothelium) muscle (smooth, skeletal or cardiac) and vascular (including vascular endothelium) tissue, or for promoting the growth of cells comprising such tissues.
  • organs including, for example, pancreas, liver, intestine, kidney, skin, endothelium
  • vascular including vascular endothelium tissue
  • a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also be useful for gut protection or regeneration and treatment of lung or liver fibrosis, reperfusion injury in various tissues, and conditions resulting from systemic cytokine damage.
  • a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also be useful for promoting or inhibiting differentiation of tissues described above from precursor tissues or cells; or for inhibiting the growth of tissues described above.
  • genes encoding tissue growth regulating activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
  • EXAMPLE 36 Assaying the Proteins Expressed from Extended cDNAs or Portions Thereof for Regulation of Reproductive Hormones
  • the proteins encoded by the extended cDNAs or portions thereof may also be evaluated for their ability to regulate reproductive hormones, such as follicle stimulating hormone.
  • reproductive hormones such as follicle stimulating hormone.
  • Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are inco ⁇ orated herein by reference: Vale et al, Endocrinol.
  • Those proteins which exhibit activity as reproductive hormones or regulators of cell movement may then be formulated as pharmaceuticals and used to treat clinical conditions in which regulation of reproductive hormones are beneficial.
  • a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also exhibit activin- or inhibin-related activities. Inhibins are characterized by their ability to inhibit the release of follicle stimulating hormone (FSH), while activins are characterized by their ability to stimulate the release of FSH.
  • FSH follicle stimulating hormone
  • a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may be useful as a contraceptive based on the ability of inhibins to decrease fertility in female mammals and decrease spermatogenesis in male mammals. Administration of sufficient amounts of other inhibins can induce infertility in these mammals.
  • the protein of the invention as a homodimer or as a heterodimer with other protein subunits of the inhibin-B group, may be useful as a fertility inducing therapeutic, based upon the ability of activin molecules in stimulating FSH release from cells of the anterior pituitary.
  • a protein of the invention may also be useful for advancement of the onset of fertility in sexually immature mammals, so as to increase the lifetime reproductive performance of domestic animals such as cows, sheep and pigs.
  • genes encoding reproductive hormone regulating activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
  • a protein encoded by extended cDNAs or portions thereof may also be evaluated for chemotactic/chemokinetic activity.
  • a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may have chemotactic or chemokinetic activity (e.g., act as a chemokine) for mammalian cells, including, for example, monocytes, fibroblasts, neutrophils, T-cells, mast cells, eosinophils, epithelial and/or endothelial cells.
  • Chemotactic and chemokinetic proteins can be used to mobilize or attract a desired cell population to a desired site of action.
  • Chemotactic or chemokinetic proteins provide particular advantages in treatment of wounds and other trauma to tissues, as well as in treatment of localized infections. For example, attraction of lymphocytes, monocytes or neutrophils to tumors or sites of infection may result in improved immune responses against the tumor or infecting agent.
  • a protein or peptide has chemotactic activity for a particular cell population if it can stimulate, directly or indirectly, the directed orientation or movement of such cell population.
  • the protein or peptide has the ability to directly stimulate directed movement of cells. Whether a particular protein has chemotactic activity for a population of cells can be readily determined by employing such protein or peptide in any known assay for cell chemotaxis.
  • the activity of a protein of the invention may, among other means, be measured by the following methods:
  • Assays for chemotactic activity consist of assays that measure the ability of a protein to induce the migration of cells across a membrane as well as the ability of a protein to induce the adhesion of one cell population to another cell population.
  • Suitable assays for movement and adhesion include, without limitation, those described in: Current Protocols in Immunology, Ed by Coligan, Kruisbeek, Margulies, Shevach and Strober, Pub. Greene Publishing Associates and Wiley- Interscience, Chapter 6.12: 6.12.1-6.12.28; Taub et ⁇ /., J. Clin. Invest.
  • the proteins encoded by the extended cDNAs or portions thereof may also be evaluated for their effects on blood clotting.
  • Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are inco ⁇ orated herein by reference: Linet et al, J. Clin. Pharmacol. 26: 131-140, 1986; Burdick et al, Thrombosis Res. 45:413-419, 1987; Humphrey et al, Fibrinolysis 5:71-79, 1991; Schaub, Prostagl ndins 35:467-474, 1988.
  • a protein of the invention may also exhibit hemostatic or thrombolytic activity.
  • a protein is expected to be useful in treatment of various coagulations disorders (including hereditary disorders, such as hemophilias) or to enhance coagulation and other hemostatic events in treating wounds resulting from trauma, surgery or other causes.
  • a protein of the invention may also be useful for dissolving or inhibiting formation of thromboses and for treatment and prevention of conditions resulting therefrom (such as infarction of cardiac and central nervous system vessels (e.g., stroke)).
  • genes encoding blood clotting activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
  • proteins encoded by the extended cDNAs or a portion thereof may also be evaluated for their involvement in receptor/ligand interactions. Numerous assays for such involvement are familiar to those skilled in the art, including the assays disclosed in the following references, which are inco ⁇ orated herein by reference: Chapter 7. 7.28.1-7.28.22 in Current Protocols in Immunology, Coligan et al. Eds. Greene Publishing Associates and Wiley-Interscience; Takai et al, Proc. Natl Acad Sci. USA 84:6864-6868, 1987; Bierer et al, J. Exp. Med.
  • the proteins encoded by extended cDNAs derived from the 5' ESTs of the present invention may also demonstrate activity as receptors, receptor ligands or inhibitors or agonists of receptor/ligand interactions.
  • receptors and ligands include, without limitation, cytokine receptors and their ligands, receptor kinases and their ligands, receptor phosphatases and their ligands, receptors involved in cell-cell interactions and their ligands (including without limitation, cellular adhesion molecules (such as selectins, integrins and their ligands) and receptor/ligand pairs involved in antigen presentation, antigen recognition and development of cellular and humoral immune responses).
  • Receptors and ligands are also useful for screening of potential peptide or small molecule inhibitors of the relevant receptor/ligand interaction.
  • a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may themselves be useful as inhibitors of receptor/ligand interactions.
  • genes encoding proteins involved in receptor/ligand interactions or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
  • EXAMPLE 40 Assaying the Proteins Expressed from Extended cDNAs or Portions Thereof for Anti-Inflammatory Activity
  • the proteins encoded by the extended cDNAs or a portion thereof may also be evaluated for anti-inflammatory activity.
  • the anti-inflammatory activity may be achieved by providing a stimulus to cells involved in the inflammatory response, by inhibiting or promoting cell-cell interactions (such as, for example, cell adhesion), by inhibiting or promoting chemotaxis of cells involved in the inflammatory process, inhibiting or promoting cell extravasation, or by stimulating or suppressing production of other factors which more directly inhibit or promote an inflammatory response.
  • Proteins exhibiting such activities can be used to treat flammatory conditions including chronic or acute conditions, including without limitation inflammation associated with infection (such as septic shock, sepsis or systemic inflammatory response syndrome), ischemia-reperfusioninury, endotoxin lethality, arthritis, complement-mediated hyperacute rejection, nephritis, cytokine- or chemokine- induced lung injury, inflammatory bowel disease, Crohn's disease or resulting from over production of cytokines such as TNF or IL-1. Proteins of the invention may also be useful to treat anaphylaxis and hypersensitivity to an antigenic substance or material. Alternatively, as described in more detail below, genes encoding anti-inflammatory activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
  • a protein of the invention may exhibit other anti-tumor activities.
  • a protein may inhibit tumor growth directly or indirectly (such as, for example, via ADCC).
  • a protein may exhibit its tumor inhibitory activity by acting on tumor tissue or tumor precursor tissue, by inhibiting formation of tissues necessary to support tumor growth (such as, for example, by inhibiting angiogenesis), by causing production of other factors, agents or cell types which inhibit tumor growth, or by suppressing, eliminating or inhibiting factors, agents or cell types which promote tumor growth.
  • genes tumor inhibition activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
  • a protein of the invention may also exhibit one or more of the following additional activities or effects: inhibiting the growth, infection or function of, or killing, infectious agents, including, without limitation, bacteria, viruses, fungi and other parasites; effecting (suppressing or enhancing) bodily characteristics, including, without limitation, height, weight, hair color, eye color, skin, fat to lean ratio or other tissue pigmentation, or organ or body part size or shape (such as, for example, breast augmentation or diminution, change in bone form or shape); effecting biorhythms or circadian cycles or rhythms; effecting the fertility of male or female subjects; effecting the metabolism, catabolism, anabolism, processing, utilization, storage or elimination of dietary fat, lipid, protein, carbohydrate, vitamins, minerals, cofactors or other nutritional factors or component(s); effecting behavioral characteristics, including, without limitation, appetite, libido, stress, cognition (including cognitive disorders), depression (including depressive disorders) and violent behaviors; providing analgesic effects or other pain reducing effects; promoting differentiation and growth
  • Proteins which interact with the polypeptides encoded by cDNAs derived from the 5' ESTs or fragments thereof, such as receptor proteins, may be identified using two hybrid systems such as the Matchmaker Two Hybrid System 2 (Catalog No. K 1604-1, Clontech). As described in the manual accompanying the kit which is inco ⁇ orated herein by reference, the the cDNAs derived from 5' ESTs, or fragments thereof, are inserted into an expression vector such that they are in frame with DNA encoding the DNA binding domain of the yeast transcriptional activator GAL4.
  • cDNAs in a cDNA library which encode proteins which might interact with the polypeptides encoded by the extended cDNAs or portions thereof are inserted into a second expression vector such that they are in frame with DNA encoding the activation domain of GAL4.
  • the two expression plasmids are transformed into yeast and the yeast are plated on selection medium which selects for expression of selectable markers on each of the expression vectors as well as GAL4 dependent expression of the HIS3 gene.
  • Transformants capable of growing on medium lacking histidine are screened for GAL4 dependent lacZ expression. Those cells which are positive in both the histidine selection and the lacZ assay contain plasmids encoding proteins which interact with the polypeptide encoded by the extended cDNAs or portions thereof.
  • the system described in Lustig et al, Methods in Enzymology 283: 83- 99, 1997, and in U.S. Patent No. 5,654,150, the disclosure of which is inco ⁇ orated herein by reference, may be used for identifying molecules which interact with the polypeptides encoded by extended cDNAs.
  • in vitro transcription reactions are performed on a pool of vectors containing extended cDNA inserts cloned downstream of a promoter which drives in vitro transcription.
  • the resulting pools of mRNAs are introduced into Xenopus laevis oocytes.
  • the oocytes are then assayed for a desired activity.
  • pooled in vitro transcription products produced as described above may be translated in vitro.
  • the pooled in vitro translation products can be assayed for a desired activity or for interaction with a known polypeptide.
  • Proteins or other molecules interacting with polypeptides encoded by extended cDNAs can be found by a variety of additional techniques.
  • affinity columns containing the polypeptide encoded by the extended cDNA or a portion thereof can be constructed.
  • the affinity column contains chimeric proteins in which the protein encoded by the extended cDNA or a portion thereof is fused to glutathione S-transferase. A mixture of cellular proteins or pool of expressed proteins as described above and is applied to the affinity column.
  • Proteins interacting with the polypeptide attached to the column can then be isolated and analyzed on 2-D electrophoresis gel as described in Ramunsen et al, Electrophoresis 18:588-598, 1997, the disclosure of which is inco ⁇ orated herein by reference.
  • the proteins retained on the affinity column can be purified by electrophoresis based methods and sequenced. The same method can be used to isolate antibodies, to screen phage display products, or to screen phage display human antibodies.
  • Proteins interacting with polypeptides encoded by extended cDNAs or portions thereof can also be screened by using an Optical Biosensor as described in Edwards and Leatherbarrow, Analytical Biochemistry 246:1-6, 1997, the disclosure of which is incorporated herein by reference.
  • the main advantage of the method is that it allows the determination of the association rate between the protein and other interacting molecules. Thus, it is possible to specifically select interacting molecules with a high or low association rate.
  • a target molecule is linked to the sensor surface (through a carboxymethl dextran matrix) and a sample of test molecules is placed in contact with the target molecules. The binding of a test molecule to the target molecule causes a change in the refractive index and/ or thickness.
  • the target molecule can be one of the polypeptides encoded by extended cDNAs or a portion thereof and the test sample can be a collection of proteins extracted from tissues or cells, a pool of expressed proteins, combinatorial peptide and/ or chemical libraries, or phage displayed peptides.
  • the tissues or cells from which the test proteins are extracted can originate from any species.
  • a target protein is immobilized and the test population is a collection of unique polypeptides encoded by the extended cDNAs or portions thereof.
  • the proteins expressed from the extended cDNAs or portions may be assayed for numerous activities in addition to those specifically enumerated above.
  • the expressed proteins may be evaluated for applications involving control and regulation of inflammation, tumor proliferation or metastasis, infection, or other clinical conditions.
  • the proteins expressed from the extended cDNAs or portions thereof may be useful as nutritional agents or cosmetic agents.
  • the proteins expressed from the cDNAs or portions thereof may be used to generate antibodies capable of specifically binding to the expressed protein or fragments thereof as described in Example 40 below.
  • the antibodies may capable of binding a full length protein encoded by a cDNA derived from a 5' EST, a mature protein (i.e. the protein generated by cleavage of the signal peptide) encoded by a cDNA derived from a 5' EST, or a signal peptide encoded by a cDNA derived from a 5' EST.
  • the antibodies may be capable of binding fragments of at least 10 amino acids of the proteins encoded by the above cDNAs.
  • the antibodies may be capable of binding fragments of at least 15 amino acids of the proteins encoded by the above cDNAs. In other embodiments, the antibodies may be capable of binding fragments of at least 25 amino acids of the proteins expressed from the extended cDNAs which comprise at least 25 amino acids of the proteins encoded by the above cDNAs. In further embodiments, the antibodies may be capable of binding fragments of at least 40 amino acids of the proteins encoded by the above cDNAs.
  • Substantially pure protein or polypeptide is isolated from the transfected or transformed cells as described in Example 30.
  • concentration of protein in the final preparation is adjusted, for example, by concentration on an Amicon filter device, to the level of a few ⁇ g/ml.
  • Monoclonal or polyclonal antibody to the protein can then be prepared as follows:
  • Monoclonal Antibody Production by Hvbridoma Fusion Monoclonal antibody to epitopes of any of the peptides identified and isolated as described can be prepared from murine hybridomas according to the classical method of Kohler, and Milstein, Nature 256:495, 1975 or derivative methods thereof. Briefly, a mouse is repetitively inoculated with a few micrograms of the selected protein or peptides derived therefrom over a period of a few weeks. The mouse is then sacrificed, and the antibody producing cells of the spleen isolated. The spleen cells are fused by means of polyethylene glycol with mouse myeloma cells, and the excess unf ⁇ ised cells destroyed by growth of the system on selective media comprising aminopterin (HAT media).
  • HAT media aminopterin
  • the successfully fused cells are diluted and aliquots of the dilution placed in wells of a microtiter plate where growth of the culture is continued.
  • Antibody-producing clones are identified by detection of antibody in the supernatant fluid of the wells by immunoassay procedures, such as ELISA, as originally described by Engvall, Meth. Enzymol. 70:419, 1980, the disclosure of which is inco ⁇ orated herein by reference and derivative methods thereof.
  • Selected positive clones can be expanded and their monoclonal antibody product harvested for use. Detailed procedures for monoclonal antibody production are described in Davis et al. in Basic Methods in Molecular Biology Elsevier, New York. Section 21-2, the disclosure of which is inco ⁇ orated herein by reference.
  • Polyclonal antiserum containing antibodies to heterogenous epitopes of a single protein can be prepared by immunizing suitable animals with the expressed protein or peptides derived therefrom, which can be unmodified or modified to enhance immunogenicity. Effective polyclonal antibody production is affected by many factors related both to the antigen and the host species. For example, small molecules tend to be less immunogenic than others and may require the use of carriers and adjuvant. Also, host animals response vary depending on site of inoculations and doses, with both inadequate or excessive doses of antigen resulting in low titer antisera. Small doses (ng level) of antigen administered at multiple intradermal sites appears to be most reliable.
  • Booster injections can be given at regular intervals, and antiserum harvested when antibody titer thereof, as determined semi-quantitatively, for example, by double irnmunodifiiision in agar against known concentrations of the antigen, begins to fall. See, for example, Ouchterlony, et al, Chap. 19 in: Handbook of Experimental Immunology D. Wier (ed) Blackwell (1973) , the disclosure of which is inco ⁇ orated herein by reference. Plateau concentration of antibody is usually in the range of 0.1 to 0.2 mg/ml of serum (about 12 ⁇ M).
  • Affinity of the antisera for the antigen is determined by preparing competitive binding curves, as described, for example, by Fisher, D., Chap. 42 in: Manual of Clinical Immunology, 2d Ed. (Rose and Friedman, Eds.) Amer. Soc. For Microbiol., Washington, D.C. (1980) , the disclosure of which is inco ⁇ orated herein by reference.
  • Antibody preparations prepared according to either protocol are useful in quantitative immunoassays which determine concentrations of antigen-bearing substances in biological samples; they are also used semi-quantitatively or qualitatively to identify the presence of antigen in a biological sample.
  • the antibodies may also be used in therapeutic compositions for killing cells expressing the protein or reducing the levels of the protein in the body. V. Use of 5' ESTs or Sequences Obtainable Therefrom or Portions Thereof as Reagents
  • the 5' ESTs of the present invention may be used as reagents in isolation procedures, diagnostic assays, and forensic procedures.
  • sequences from the 5' ESTs may be detectably labeled and used as probes to isolate other sequences capable of hybridizing to them.
  • sequences from 5' ESTs may be used to design PCR primers to be used in isolation, diagnostic, or forensic procedures.
  • the 5' EST sequences may be used to prepare PCR primers for a variety of applications, including isolation procedures for cloning nucleic acids capable of hybridizing to such sequences, diagnostic techniques and forensic techniques.
  • the PCR primers are at least 10 bases, and preferably at least 12, 15, or 17 bases in length. More preferably, the PCR primers are at least 20-30 bases in length. In some embodiments, the PCR primers may be more than 30 bases in length. It is preferred that the primer pairs have approximately the same G/C ratio, so that melting temperatures are approximately the same.
  • a variety of PCR techniques are familiar to those skilled in the art. For a review of PCR technology, see Molecular Cloning to Genetic Engineering, White Ed.
  • PCR primers on either side of the nucleic acid sequences to be amplified are added to a suitably prepared nucleic acid sample along with dNTPs and a thermostable polymerase such as Taq polymerase, Pfu polymerase, or Vent polymerase.
  • a thermostable polymerase such as Taq polymerase, Pfu polymerase, or Vent polymerase.
  • the nucleic acid in the sample is denatured and the PCR primers are specifically hybridized to complementary nucleic acid sequences in the sample.
  • the hybridized primers are extended. Thereafter, another cycle of denaturation, hybridization, and extension is initiated. The cycles are repeated multiple times to produce an amplified fragment containing the nucleic acid sequence between the primer sites.
  • Probes derived from 5' ESTs (or cDNAs or genomic DNAs obtainable therefrom), including full length cDNAs or genomic sequences, may be labeled with detectable labels familiar to those skilled in the art, including radioisotopes and non-radioactive labels, to provide a detectable probe.
  • the detectable probe may be single stranded or double stranded and may be made using techniques known in the art, including in vitro transcription, nick translation, or kinase reactions.
  • a nucleic acid sample containing a sequence capable of hybridizing to the labeled probe is contacted with the labeled probe. If the nucleic acid in the sample is double stranded, it may be denatured prior to contacting the probe.
  • the nucleic acid sample may be immobilized on a surface such as a nitrocellulose or nylon membrane.
  • the nucleic acid sample may comprise nucleic acids obtained from a variety of sources, including genomic DNA, cDNA libraries, RNA, or tissue samples.
  • Procedures used to detect the presence of nucleic acids capable of hybridizing to the detectable probe include well known techniques such as Southern blotting, Northern blotting, dot blotting, colony hybridization, and plaque hybridization.
  • the nucleic acid capable of hybridizing to the labeled probe may be cloned into vectors such as expression vectors, sequencing vectors, or in vitro transcription vectors to facilitate the characterization and expression of the hybridizing nucleic acids in the sample.
  • vectors such as expression vectors, sequencing vectors, or in vitro transcription vectors to facilitate the characterization and expression of the hybridizing nucleic acids in the sample.
  • such techniques may be used to isolate and clone sequences in a genomic library or cDNA library which are capable of hybridizing to the detectable probe as described in Example 30 above.
  • PCR primers made as described in Example 44 above may be used in forensic analyses, such as the DNA finge ⁇ rinting techniques described in Examples 46-50 below. Such analyses may utilize detectable probes or primers based on the sequences of the the 5' ESTs or of cDNAs or genomic DNAs isolated using the 5' ESTs.
  • DNA samples are isolated from forensic specimens of, for example, hair, semen, blood or skin cells by conventional methods.
  • a panel of PCR primers based on a number of the 5' ESTs of Example 25, or cDNAs or genomic DNAs isolated therefrom as described above, is then utilized in accordance with Example 44 to amplify DNA of approximately 100-200 bases in length from the forensic specimen.
  • Corresponding sequences are obtained from a test subject.
  • Each of these identification DNAs is then sequenced using standard techniques, and a simple database comparison determines the differences, if any, between the sequences from the subject and those from the sample.
  • primers are prepared from a large number of 5'EST sequences from Example 25, or cDNA or genomic DNA sequences obtainable therefrom. Preferably, 20 to 50 different primers are used. These primers are used to obtain a corresponding number of PCR-generated DNA segments from the individual in question in accordance with Example 44. Each of these DNA segments is sequenced, using the methods set forth in Example 46. The database of sequences generated through this procedure uniquely identifies the individual from whom the sequences were obtained. The same panel of primers may then be used at any later time to absolutely correlate tissue or other biological specimen with that individual.
  • EXAMPLE 48
  • Example 47 Southern Blot Forensic Identification The procedure of Example 47 is repeated to obtain a panel of at least 10 amplified sequences from an individual and a specimen.
  • the panel contains at least 50 amplified sequences. More preferably, the panel contains 100 amplified sequences. In some embodiments, the panel contains 200 amplified sequences.
  • This PCR-generated DNA is then digested with one or a combination of, preferably, four base specific restriction enzymes. Such enzymes are commercially available and known to those of skill in the art. After digestion, the resultant gene fragments are size separated in multiple duplicate wells on an agarose gel and transferred to nitrocellulose using Southern blotting techniques well known to those with skill in the art. For a review of Southern blotting see Davis et al. (Basic Methods in Molecular Biology, 1986, Elsevier Press, pp 62-65) , the disclosure of which is inco ⁇ orated herein by reference..
  • a panel of probes based on the sequences of 5' ESTs (or cDNAs or genomic DNAs obtainable therefrom), or fragments thereof of at least 10 bases, are radioactively or colorimetrically labeled using methods known in the art, such as nick translation or end labeling, and hybridized to the Southern blot using techniques known in the art (Davis et al, supra).
  • the probe comprises at least 12, 15, or 17 consecutive nucleotides from the 5' EST (or cDNAs or genomic DNAs obtainable therefrom). More preferably, the probe comprises at least 20-30 consecutive nucleotides from the 5' EST (or cDNAs or genomic DNAs obtainable therefrom). In some embodiments, the probe comprises more than 30 nucleotides from the 5' EST (or cDNAs or genomic DNAs obtainable therefrom).
  • At least 5 to 10 of these labeled probes are used, and more preferably at least about 20 or 30 are used to provide a unique pattern.
  • the resultant bands appearing from the hybridization of a large sample of 5' EST (or cDNAs or genomic DNAs obtainable therefrom) will be a unique identifier. Since the restriction enzyme cleavage will be different for every individual, the band pattern on the Southern blot will also be unique. Increasing the number of 5' EST (or cDNAs or genomic DNAs obtainable therefrom) probes will provide a statistically higher level of confidence in the identification since there will be an increased number of sets of bands used for identification.
  • Dot Blot Identification Procedure Another technique for identifying individuals using the 5' EST sequences disclosed herein utilizes a dot blot hybridization technique. Genomic DNA is isolated from nuclei of subject to be identified. Oligonucleotide probes of approximately 30 bp in length are synthesized that correspond to at least 10, preferably 50 sequences from the 5' ESTs or cDNAs or genomic DNAs obtainable therefrom. The probes are used to hybridize to the genomic DNA through conditions known to those in the art. The oligonucleotides are end labeled with P 32 using polynucleotide kinase (Pharmacia).
  • Dot Blots are created by spotting the genomic DNA onto nitrocellulose or the like using a vacuum dot blot manifold (BioRad, Richmond California).
  • the nitrocellulose filter containing the genomic sequences is baked or UV linked to the filter, prehybridized and hybridized with labeled probe using techniques known in the art (Davis et al, supra).
  • the 32 P labeled DNA fragments are sequentially hybridized with successively stringent conditions to detect minimal differences between the 30 bp sequence and the DNA.
  • Tetramethylammonium chloride is useful for identifying clones containing small numbers of nucleotide mismatches (Wood et al, Proc. Natl. Acad. Sci. USA 82(6): 1585-1588, 1985) which is hereby inco ⁇ orated by reference. A unique pattern of dots distinguishes one individual from another individual.
  • 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom) or oligonucleotides containing at least 10 consecutive bases from these sequences can be used as probes in the following alternative finge ⁇ rinting technique.
  • the probe comprises at least 12, 15, or 17 consecutive nucleotides from the 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom).
  • the probe comprises at least 20-30 consecutive nucleotides from the 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom). In some embodiments, the probe comprises more than 30 nucleotides from the 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom).
  • Example 50 provides a representative alternative finge ⁇ rinting procedure in which the probes are derived from 5 'EST. EXAMPLE 50
  • oligonucleotides are prepared from a large number, e.g. 50, 100, or 200, of 5'EST using commercially available oligonucleotide services such as Genset, Paris, France.
  • Cell samples from the test subject are processed for DNA using techniques well known to those with skill in the art.
  • the nucleic acid is digested with restriction enzymes such as EcoRI and Xbal. Following digestion, samples are applied to wells for electrophoresis.
  • the procedure as known in the art, may be modified to accommodate polyacrylamide electrophoresis, however in this example, samples containing 5 ug of DNA are loaded into wells and separated on 0.8% agarose gels. The gels are transferred onto nitrocellulose using standard Southern blotting techniques.
  • the proteins encoded by the extended cDNAs may also be used to generate antibodies as explained in Examples 30 and 43 in order to identify the tissue type or cell species from which a sample is derived as described in example 51.
  • tissue specific antigens are accomplished by the visualization of tissue specific antigens by means of antibody preparations according to Examples 30 and 43 which are conjugated, directly or indirectly to a detectable marker.
  • Selected labeled antibody species bind to their specific antigen binding partner in tissue sections, cell suspensions, or in extracts of soluble proteins from a tissue sample to provide a pattern for qualitative or semi-qualitative inte ⁇ retation.
  • Antisera for these procedures must have a potency exceeding that of the native preparation, and for that reason, antibodies are concentrated to a mg/ml level by isolation of the gamma globulin fraction, for example, by ion-exchange chromatography or by ammonium sulfate fractionation.
  • a fluorescent marker either fluorescein or rhodamine
  • antibodies can also be labeled with an enzyme that supports a color producing reaction with a substrate, such as horseradish peroxidase. Markers can be added to tissue-bound antibody in a second step, as described below.
  • the specific antitissue antibodies can be labeled with fe ⁇ itin or other electron dense particles, and localization of the fe ⁇ itin coupled antigen-antibody complexes achieved by means of an electron microscope.
  • the antibodies are radiolabeled, with, for example 125 I, and detected by overlaying the antibody treated preparation with photographic emulsion.
  • Preparations to carry out the procedures can comprise monoclonal or polyclonal antibodies to a single protein or peptide identified as specific to a tissue type, for example, brain tissue, or antibody preparations to several antigenically distinct tissue specific antigens can be used in panels, independently or in mixtures, as required.
  • Tissue sections and cell suspensions are prepared for immunohistochemical examination according to common histological techniques. Multiple cryostat sections (about 4 ⁇ m, unfixed) of the unknown tissue and known control, are mounted and each slide covered with different dilutions of the antibody preparation. Sections of known and unknown tissues should also be treated with preparations to provide a positive control, a negative control, for example, pre-immune sera, and a control for non-specific staining, for example, buffer.
  • Treated sections are incubated in a humid chamber for 30 min at room temperature, rinsed, then washed in buffer for 30-45 min. Excess fluid is blotted away, and the marker developed.
  • tissue specific antibody was not labeled in the first incubation, it can be labeled at this time in a second antibody-antibody reaction, for example, by adding fluorescein- or enzyme-conjugated antibody against the immunoglobulin class of the antiserum-producing species, for example, fluorescein labeled antibody to mouse IgG.
  • fluorescein- or enzyme-conjugated antibody against the immunoglobulin class of the antiserum-producing species for example, fluorescein labeled antibody to mouse IgG.
  • fluorescein labeled antibody to mouse IgG.
  • tissue specific soluble proteins The visualization of tissue specific proteins and identification of unknown tissues from that procedure is carried out using the labeled antibody reagents and detection strategy as described for immunohistochemistry; however the sample is prepared according to an electrophoretic technique to distribute the proteins extracted from the tissue in an orderly array on the basis of molecular weight for detection.
  • a tissue sample is homogenized using a Virtis apparatus; cell suspensions are disrupted by Dounce homogenization or osmotic lysis, using detergents in either case as required to disrupt cell membranes, as is the practice in the art.
  • Insoluble cell components such as nuclei, microsomes, and membrane fragments are removed by ultracentrifugation, and the soluble protein-containing fraction concentrated if necessary and reserved for analysis.
  • a sample of the soluble protein solution is resolved into individual protein species by conventional SDS polyacrylamide electrophoresis as described, for example, by Davis, et al, Section 19-2 in: Basic Methods in Molecular Biology, Leder ed., Elsevier, New York, 1986, the disclosure of which is inco ⁇ orated herein by reference, using a range of amounts of polyacrylamide in a set of gels to resolve the entire molecular weight range of proteins to be detected in the sample.
  • a size marker is run in parallel for pu ⁇ oses of estimating molecular weights of the constituent proteins.
  • Sample size for analysis is a convenient volume of from 5 to 55 ⁇ l, and containing from about 1 to 100 ⁇ g protein.
  • An aliquot of each of the resolved proteins is transferred by blotting to a nitrocellulose filter paper, a process that maintains the pattern of resolution. Multiple copies are prepared.
  • the procedure, known as Western Blot Analysis, is well described in Davis, L. et al, supra Section 19-3.
  • One set of nitrocellulose blots is stained with Coomassie blue dye to visualize the entire set of proteins for comparison with the antibody bound proteins.
  • the remaining nitrocellulose filters are then incubated with a solution of one or more specific antisera to tissue specific proteins prepared as described in Examples 30 and 43.
  • a detectable label can be attached to the primary tissue antigen-primary antibody complex according to various strategies and permutations thereof.
  • the primary specific antibody can be labeled; alternatively, the unlabeled complex can be bound by a labeled secondary anti-IgG antibody.
  • either the primary or secondary antibody is conjugated to a biotin molecule, which can, in a subsequent step, bind an avidin conjugated marker.
  • enzyme labeled or radioactive protein A which has the property of binding to any IgG, is bound in a final step to either the primary or secondary antibody.
  • tissue specific antigen binding at levels above those seen in control tissues to one or more tissue specific antibodies, prepared from the gene sequences identified from extended cDNA sequences, can identify tissues of unknown origin, for example, forensic samples, or differentiated tumor tissue that has metastasized to foreign bodily sites.
  • 5' ESTs may be mapped to their chromosomal locations.
  • Example 52 below describes radiation hybrid (RH) mapping of human chromosomal regions using 5 'ESTs.
  • Example 53 describes a representative procedure for mapping an 5' EST to its location on a human chromosome.
  • Example 54 below describes mapping of 5' ESTs on metaphase chromosomes by Fluorescence In Situ Hybridization (FISH).
  • FISH Fluorescence In situ Hybridization
  • Radiation hybrid mapping of 5'ESTs to the human genome Radiation hybrid (RH) mapping is a somatic cell genetic approach that can be used for high resolution mapping of the human genome.
  • RH Radiation hybrid
  • cell lines containing one or more human chromosomes are lethally irradiated, breaking each chromosome into fragments whose size depends on the radiation dose. These fragments are rescued by fusion with cultured rodent cells, yielding subclones containing different portions of the human genome.
  • This technique is described by Benham et al, Genomics 4:509-517, 1989; and Cox et al, Science 250:245-250, 1990, the entire contents of which are hereby inco ⁇ orated by reference.
  • RH mapping has been used to generate a high-resolution whole genome radiation hybrid map of human chromosome 17q22-q25.3 across the genes for growth hormone (GH) and thymidine kinase (TK) (Foster et al, Genomics 33:185-192, 1996), the region surrounding the Gorlin syndrome gene (Obermayr et al, Eur. J. Hum. Genet.
  • oligonucleotide primer pairs are designed from the 5' ESTs (or cDNAs or genomic DNAs obtainable therefrom) to minimize the chance of amplifying through an intron.
  • the oligonucleotide primers are 18-23 bp in length and are designed for PCR amplification.
  • the creation of PCR primers from known sequences is well known to those with skill in the art. For a review of PCR technology see Erlich in PCR Technology; Principles and Applications or DNA Amplification, Freeman and Co., New York, 1992, the disclosure of which is inco ⁇ orated herein by reference.
  • the primers are used in polymerase chain reactions (PCR) to amplify templates from total human genomic DNA.
  • PCR conditions are as follows: 60 ng of genomic DNA is used as a template for PCR with 80 ng of each oligonucleotide primer, 0.6 unit of Taq polymerase, and 1 ⁇ Cu of a 32 P-labeled deoxycytidine triphosphate.
  • the PCR is performed in a microplate thermocycler (Techne) under the following conditions: 30 cycles of 94°C, 1.4 min; 55°C, 2 min; and 72°C, 2 min; with a final extension at 72°C for 10 min.
  • the amplified products are analyzed on a 6%> polyacrylamide sequencing gel and visualized by autoradiography.
  • the PCR reaction is repeated with DNA templates from two panels of human- rodent somatic cell hybrids, BIOS PCRable DNA (BIOS Co ⁇ oration) and NIGMS Human- Rodent Somatic Cell Hybrid Mapping Panel Number 1 (NIGMS, Camden, NJ).
  • PCR is used to screen a series of somatic cell hybrid cell lines containing defined sets of human chromosomes for the presence of a given 5' EST (or cDNA or genomic DNA obtainable therefrom).
  • DNA is isolated from the somatic hybrids and used as starting templates for PCR_reactions using the primer pairs from the 5' EST (or cDNA or genomic DNA obtainable therefrom). Only those somatic cell hybrids with chromosomes containing the human gene corresponding to the 5' EST (or cDNA or genomic DNA obtainable therefrom) will yield an amplified fragment.
  • the 5' EST (or cDNA or genomic DNA obtainable therefrom) are assigned to a chromosome by analysis of the segregation pattern of PCR products from the somatic hybrid DNA templates.
  • the single human chromosome present in all cell hybrids that give rise to an amplified fragment is the chromosome containing that 5'EST (or cDNA or genomic DNA obtainable therefrom).
  • 5'EST or cDNA or genomic DNA obtainable therefrom.
  • Hybridization Fluorescence in situ hybridization allows the 5'EST (or cDNA or genomic DNA obtainable therefrom) to be mapped to a particular location on a given chromosome.
  • the chromosomes to be used for fluorescence in situ hybridization techniques may be obtained from a variety of sources including cell cultures, tissues, or whole blood.
  • chromosomal localization of an 5'EST is obtained by FISH as described by Cherif et al. (Proc. Natl. Acad Sci. U.S.A., 87:6639-6643, 1990) , the disclosure of which is inco ⁇ orated herein by reference.
  • Metaphase chromosomes are prepared from phytohemagglutinin (PHA)- stimulated blood cell donors. PHA-stimulated lymphocytes from healthy males are cultured for 72 h in RPMI-1640 medium.
  • methotrexate (10 ⁇ M) is added for 17 h, followed by addition of 5-bromodeoxyuridine (5-BrdU, 0.1 mM) for 6 h.
  • Colcemid (1 ⁇ g/ml) is added for the last 15 min before harvesting the cells.
  • Cells are collected, washed in RPMI, incubated with a hypotonic solution of KCl (75 mM) at 37°C for 15 min and fixed in three changes of methanol: acetic acid (3:1). The cell suspension is dropped onto a glass slide and air dried.
  • the 5'EST (or cDNA or genomic DNA obtainable therefrom) is labeled with biotin- 16 dUTP by nick translation according to the manufacturer's instructions (Bethesda Research Laboratories, Bethesda, MD), purified using a Sephadex G-50 column (Pharmacia, Upsala, Sweden) and precipitated.
  • the DNA pellet is dissolved in hybridization buffer (50% formamide, 2 X SSC, 10% dextran sulfate, 1 mg/ml sonicated salmon sperm DNA, pH 7) and the probe is denatured at 70°C for 5-10 min.
  • Slides kept at -20°C are treated for 1 h at 37°C with RNase A (100 ⁇ g/ml), rinsed three times in 2 X SSC and dehydrated in an ethanol series. Chromosome preparations are denatured in 70% formamide, 2 X SSC for 2 min at 70°C, then dehydrated at 4°C.
  • the slides are treated with proteinase K (10 ⁇ g/100 ml in 20 mM Tris-HCl, 2 mM CaCl 2 ) at 37°C for 8 min and dehydrated.
  • the hybridization mixture containing the probe is placed on the slide, covered with a coverslip, sealed with rubber cement and incubated overnight in a humid chamber at 37°C.
  • the biotinylated probe is detected by avidin-FITC and amplified with additional layers of biotinylated goat anti-avidin and avidin-FITC.
  • fluorescent R-bands are obtained as previously described (Cherif et al, supra.). The slides are observed under a LEICA fluorescence microscope (DMRXA). Chromosomes are counterstained with propidium iodide and the fluorescent signal of the probe appears as two symmetrical yellow-green spots on both chromatids of the fluorescent R-band chromosome (red).
  • DMRXA LEICA fluorescence microscope
  • the 5'EST (or cDNA or genomic DNA obtainable therefrom) have been assigned to particular chromosomes using the techniques described in Examples 52-54 above, they may be utilized to construct a high resolution map of the chromosomes on which they are located or to identify the chromosomes in a sample.
  • Chromosome mapping involves assigning a given unique sequence to a particular chromosome as described above. Once the unique sequence has been mapped to a given chromosome, it is ordered relative to other unique sequences located on the same chromosome.
  • One approach to chromosome mapping utilizes a series of yeast artificial chromosomes (YACs) bearing several thousand long inserts derived from the chromosomes of the organism from which the extended cDNAs (or genomic DNAs obtainable therefrom) are obtained. This approach is described in Nagaraja et al, Genome Research 7:210-222, 1997, the disclosure of which is inco ⁇ orated herein by reference.
  • each chromosome is broken into overlapping pieces which are inserted into the YAC vector.
  • the YAC inserts are screened using PCR or other methods to determine whether they include the 5'EST (or cDNA or genomic DNA obtainable therefrom) whose position is to be determined. Once an insert has been found which includes the 5'EST (or cDNA or genomic DNA obtainable therefrom), the insert can be analyzed by PCR or other methods to determine whether the insert also contains other sequences known to be on the chromosome or in the region from which the 5'EST (or cDNA or genomic DNA obtainable therefrom) was derived.
  • This process can be repeated for each insert in the YAC library to determine the location of each of the extended cDNAs (or genomic DNAs obtainable therefrom) relative to one another and to other known chromosomal markers. In this way, a high resolution map of the distribution of numerous unique markers along each of the organisms chromosomes may be obtained.
  • extended cDNAs may also be used to identify genes associated with a particular phenotype, such as hereditary disease or drug response.
  • EXAMPLE 56 Identification of genes associated with hereditary diseases or drug response This example illustrates an approach useful for the association of 5'ESTs (or cDNA or genomic DNA obtainable therefrom) with particular phenotypic characteristics.
  • a particular 5'EST or cDNA or genomic DNA obtainable therefrom is used as a test probe to associate that 5'EST (or cDNA or genomic DNA obtainable therefrom) with a particular phenotypic characteristic.
  • 5'ESTs (or cDNA or genomic DNA obtainable therefrom) are mapped to a particular location on a human chromosome using techniques such as those described in Examples 52 and 53 or other techniques known in the art.
  • a search of Mendelian Inheritance in Man (McKusick in Mendelian Inheritance in Man (available on line through Johns Hopkins University Welch Medical Library) reveals the region of the human chromosome which contains the 5'EST (or cDNA or genomic DNA obtainable therefrom) to be a very gene rich region containing several known genes and several diseases or phenotypes for which genes have not been identified.
  • the gene corresponding to this 5'EST (or cDNA or genomic DNA obtainable therefrom) thus becomes an immediate candidate for each of these genetic diseases.
  • PCR primers from the 5'EST are used to screen genomic DNA, mRNA or cDNA obtained from the patients.
  • 5'ESTs or cDNA or genomic DNA obtainable therefrom
  • the PCR analysis may yield fragments of different lengths when the samples are derived from an individual having the phenotype associated with the disease than when the sample is derived from a healthy individual, indicating that the gene containing the 5'EST may be responsible for the genetic disease.
  • 5'EST or cDNA or Genomic DNA Obtainable Therefrom
  • the present 5'ESTs may also be used to construct secretion vectors capable of directing the secretion of the proteins encoded by genes therein.
  • secretion vectors may facilitate the purification or enrichment of the proteins encoded by genes inserted therein by reducing the number of background proteins from which the desired protein must be purified or enriched.
  • Exemplary secretion vectors are described in Example 57 below.
  • the secretion vectors include a promoter capable of directing gene expression in the host cell, tissue, or organism of interest.
  • promoters include the Rous Sarcoma Virus promoter, the SV40 promoter, the human cytomegalovirus promoter, and other promoters familiar to those skilled in the art.
  • a signal sequence from a 5' EST (or cDNAs or genomic DNAs obtainable therefrom) is operably linked to the promoter such that the mRNA transcribed from the promoter will direct the translation of the signal peptide.
  • the host cell, tissue, or organism may be any cell, tissue, or organism which recognizes the signal peptide encoded by the signal sequence in the 5' EST (or cDNA or genomic DNA obtainable therefrom). Suitable hosts include mammalian cells, tissues or organisms, avian cells, tissues, or organisms, insect cells, tissues or organisms, or yeast.
  • the secretion vector contains cloning sites for inserting genes encoding the proteins which are to be secreted.
  • the cloning sites facilitate the cloning of the insert gene in frame with the signal sequence such that a fusion protein in which the signal peptide is fused to the protein encoded by the inserted gene is expressed from the mRNA transcribed from the promoter.
  • the signal peptide directs the extracellular secretion of the fusion protein.
  • the secretion vector may be DNA or RNA and may integrate into the chromosome of the host, be stably maintained as an extrachromosomal replicon in the host, be an artificial chromosome, or be transiently present in the host.
  • Many nucleic acid backbones suitable for use as secretion vectors are known to those skilled in the art, including retroviral vectors, SV40 vectors, Bovine Papilloma Virus vectors, yeast integrating plasmids, yeast episomal plasmids, yeast artificial chromosomes, human artificial chromosomes, P element vectors, baculovirus vectors, or bacterial plasmids capable of being transiently introduced into the host.
  • the secretion vector may also contain a polyA signal such that the polyA signal is located downstream of the gene inserted into the secretion vector.
  • the secretion vector is introduced into the host cell, tissue, or organism using calcium phosphate precipitation, DEAE-Dextran, electroporation, liposome-mediated transfection, viral particles or as naked DNA.
  • the protein encoded by the inserted gene is then purified or enriched from the supernatant using conventional techniques such as ammonium sulfate precipitation, immunoprecipitation, immunochromatography, size exclusion chromatography, ion exchange chromatography, and HPLC.
  • the secreted protein may be in a sufficiently enriched or pure state in the supernatant or growth media of the host to permit it to be used for its intended pu ⁇ ose without further enrichment.
  • the signal sequences may also be inserted into vectors designed for gene therapy.
  • the signal sequence is operably linked to a promoter such that mRNA transcribed from the promoter encodes the signal peptide.
  • a cloning site is located downstream of the signal sequence such that a gene encoding a protein whose secretion is desired may readily be inserted into the vector and fused to the signal sequence.
  • the vector is introduced into an appropriate host cell.
  • the protein expressed from the promoter is secreted extracellularly, thereby producing a therapeutic effect.
  • the 5' ESTs may also be used to clone sequences located upstream of the 5' ESTs which are capable of regulating gene expression, including promoter sequences, enhancer sequences, and other upstream sequences which influence transcription or translation levels. Once identified and cloned, these upstream regulatory sequences may be used in expression vectors designed to direct the expression of an inserted gene in a desired spatial, temporal, developmental, or quantitative fashion.
  • Example 58 describes a method for cloning sequences upstream of the extended cDNAs or 5' ESTs.
  • Sequences derived from extended cDNAs or 5' ESTs may be used to isolate the promoters of the corresponding genes using chromosome walking techniques.
  • chromosome walking technique which utilizes the GenomeWalkerTM kit available from Clontech, five complete genomic DNA samples are each digested with a different restriction enzyme which has a 6 base recognition site and leaves a blunt end. Following digestion, oligonucleotide adapters are ligated to each end of the resulting genomic DNA fragments.
  • a first PCR reaction is performed according to the manufacturer's instructions (which are inco ⁇ orated herein by reference) using an outer adaptor primer provided in the kit and an outer gene specific primer.
  • the gene specific primer should be selected to be specific for the extended cDNA or 5' EST of interest and should have a melting temperature, length, and location in the extended cDNA or 5'EST which is consistent with its use in PCR reactions.
  • Each first PCR reaction contains 5 ng of genomic DNA, 5 ⁇ l of 10X Tth reaction buffer, 0.2 mM of each dNTP, 0.2 ⁇ M each of outer adaptor primer and outer gene specific primer, 1.1 mM of Mg(OAc) 2 , and 1 ⁇ l of the Tth polymerase 50X mix in a total volume of 50 ⁇ l.
  • the reaction cycle for the first PCR reaction is as follows: 1 min - 94°C / 2 sec - 94°C, 3 min - 72°C (7 cycles) / 2 sec - 94°C, 3 min - 67°C (32 cycles) / 5 min - 67°C.
  • the product of the first PCR reaction is diluted and used as a template for a second PCR reaction according to the manufacturer's instructions using a pair of nested primers which are located internally on the amplicon resulting from the first PCR reaction.
  • 5 ⁇ l of the reaction product of the first PCR reaction mixture may be diluted 180 times. Reactions are made in a 50 ⁇ l volume having a composition identical to that of the first PCR reaction except the nested primers are used.
  • the first nested primer is specific for the adaptor, and is provided with the GenomeWalkerTM kit.
  • the second nested primer is specific for the particular extended cDNA or 5' EST for which the promoter is to be cloned and should have a melting temperature, length, and location in the extended cDNA or 5' EST which is consistent with its use in PCR reactions.
  • the reaction parameters of the second PCR reaction are as follows: 1 min - 94°C / 2 sec - 94°C, 3 min - 72°C (6 cycles) / 2 sec - 94°C, 3 min - 67°C (25 cycles) / 5 min - 67°C.
  • the product of the second PCR reaction is purified, cloned, and sequenced using standard techniques.
  • two or more human genomic DNA libraries can be constructed by using two or more restriction enzymes.
  • the digested genomic DNA is cloned into vectors which can be converted into single stranded, circular, or linear DNA.
  • a biotinylated oligonucleotide comprising at least 15 nucleotides from the extended cDNA or 5' EST sequence is hybridized to the single stranded DNA. Hybrids between the biotinylated oligonucleotide and the single stranded DNA containing the extended cDNA or EST sequence are isolated as described in Example 29 above.
  • the single stranded DNA containing the extended cDNA or EST sequence is released from the beads and converted into double stranded DNA using a primer specific for the extended cDNA or 5' EST sequence or a primer corresponding to a sequence included in the cloning vector.
  • the resulting double stranded DNA is transformed into bacteria.
  • DNAs containing the 5' EST or extended cDNA sequences are identified by colony PCR or colony hybridization.
  • promoters and transcription start sites within the upstream sequences may be identified by comparing the sequences upstream of the extended cDNAs or 5' ESTs with databases containing known transcription start sites, transcription factor binding sites, or promoter sequences.
  • promoters in the upstream sequences may be identified using promoter reporter vectors as described in Example .
  • each of these promoter reporter vectors include multiple cloning sites positioned upstream of a reporter gene encoding a readily assayable protein such as secreted alkaline phosphatase, ⁇ galactosidase, or green fluorescent protein.
  • the sequences upstream of the extended cDNAs or 5' ESTs are inserted into the cloning sites upstream of the reporter gene in both orientations and introduced into an appropriate host cell.
  • the level of reporter protein is assayed and compared to the level obtained from a vector which lacks an insert in the cloning site.
  • the presence of an elevated expression level in the vector containing the insert with respect to the control vector indicates the presence of a promoter in the insert.
  • the upstream sequences can be cloned into vectors which contain an enhancer for augmenting transcription levels from weak promoter sequences. A significant level of expression above that observed with the vector lacking an insert indicates that a promoter sequence is present in the inserted upstream sequence.
  • Appropriate host cells for the promoter reporter vectors may be chosen based on the results of the above described determination of expression patterns of the extended cDNAs and ESTs. For example, if the expression pattern analysis indicates that the mRNA corresponding to a particular extended cDNA or 5' EST is expressed in fibroblasts, the promoter reporter vector may be introduced into a human fibroblast cell line.
  • Promoter sequences within the upstream genomic DNA may be further defined by constructing nested deletions in the upstream DNA using conventional techniques such as Exonuclease HI digestion. The resulting deletion fragments can be inserted into the promoter reporter vector to determine whether the deletion has reduced or obliterated promoter activity. In this way, the boundaries of the promoters may be defined. If desired, potential individual regulatory sites within the promoter may be identified using site directed mutagenesis or linker scanning to obliterate potential transcription factor binding sites within the promoter individually or in combination. The effects of these mutations on transcription levels may be determined by inserting the mutations into the cloning sites in the promoter reporter vectors.
  • the promoter having the internal designation P13H2 (SEQ ID NO:31) was obtained.
  • Figure 4 provides a schematic description of the promoters isolated and the way they are assembled with the corresponding 5' tags.
  • the upstream sequences were screened for the presence of motifs resembling transcription factor binding sites or known transcription start sites using the computer program Matlnspector release 2.0, August 1996.
  • Table VII describes the transcription factor binding sites present in each of these promoters.
  • the columns labeled matrice provides the name of the Matlnspector matrix used.
  • the column labeled position provides the 5' position of the promoter site. Numeration of the sequence starts from the transcription site as determined by matching the genomic sequence with the 5' EST sequence.
  • the column labeled "orientation” indicates the DNA strand on which the site is found, with the + strand being the coding strand as determined by matching the genomic sequence with the sequence of the 5' EST.
  • the column labeled “score” provides the Matlnspector score found for this site.
  • the column labeled "length” provides the length of the site in nucleotides.
  • the column labeled "sequence” provides the sequence of the site found.
  • Bacterial clones containing plasmids containing the promoter sequences described above described above are presently stored in the inventor's laboratories under the internal identification numbers provided above.
  • the inserts may be recovered from the deposited materials by growing an aliquot of the appropriate bacterial clone in the appropriate medium.
  • the plasmid DNA can then be isolated using plasmid isolation procedures familiar to those skilled in the art such as alkaline lysis minipreps or large scale alkaline lysis plasmid isolation procedures. If desired the plasmid DNA may be further enriched by centrUugation on a cesium chloride gradient, size exclusion chromatography, or anion exchange chromatography.
  • the plasmid DNA obtained using these procedures may then be manipulated using standard cloning techniques familiar to those skilled in the art.
  • a PCR can be done with primers designed at both ends of the EST insertion.
  • the PCR product which corresponds to the 5' EST can then be manipulated using standard cloning techniques familiar to those skilled in the art.
  • the promoters and other regulatory sequences located upstream of the extended cDNAs or 5' ESTs may be used to design expression vectors capable of directing the expression of an inserted gene in a desired spatial, temporal, developmental, or quantitative manner.
  • a promoter capable of directing the desired spatial, temporal, developmental, and quantitative patterns may be selected using the results of the expression analysis described in Example 26 above. For example, if a promoter which confers a high level of expression in muscle is desired, the promoter sequence upstream of an extended cDNA or 5' EST derived from an mRNA which is expressed at a high level in muscle, as determined by the method of Example 26, may be used in the expression vector.
  • the desired promoter is placed near multiple restriction sites to facilitate the cloning of the desired insert downstream of the promoter, such that the promoter is able to drive expression of the inserted gene.
  • the promoter may be inserted in conventional nucleic acid backbones designed for extrachromosomal replication, integration into the host chromosomes or transient expression.
  • Suitable backbones for the present expression vectors include retroviral backbones, backbones from eukaryotic episomes such as SV40 or Bovine Papilloma Virus, backbones from bacterial episomes, or artificial chromosomes.
  • the expression vectors also include a polyA signal downstream of the multiple restriction sites for directing the polyadenylation of mRNA transcribed from the gene inserted into the expression vector.
  • Sequences within the promoter region which are likely to bind transcription factors may be identified by homology to known transcription factor binding sites or through conventional mutagenesis or deletion analyses of reporter plasmids containing the promoter sequence. For example, deletions may be made in a reporter plasmid containing the promoter sequence of interest operably linked to an assayable reporter gene. The reporter plasmids carrying various deletions within the promoter region are transfected into an appropriate host cell and the effects of the deletions on expression levels is assessed. Transcription factor binding sites within the regions in which deletions reduce expression levels may be further localized using site directed mutagenesis, linker scanning analysis, or other techniques familiar to those skilled in the art.
  • Nucleic acids encoding proteins which interact with sequences in the promoter may be identified using one-hybrid systems such as those described in the manual accompanying the Matchmaker One-Hybrid System kit available from Clontech (Catalog No. K1603-1), the disclosure of which is inco ⁇ orated herein by reference. Briefly, the Matchmaker One-hybrid system is used as follows. The target sequence for which it is desired to identify binding proteins is cloned upstream of a selectable reporter gene and integrated into the yeast genome. Preferably, multiple copies of the target sequences are inserted into the reporter plasmid in tandem.
  • the yeast are plated on selective media to select cells expressing the selectable marker linked to the promoter sequence.
  • the colonies which grow on the selective media contain genes encoding proteins which bind the target sequence.
  • the inserts in the genes encoding the fusion proteins are further characterized by sequencing.
  • the inserts may be inserted into expression vectors or in vitro transcription vectors. Binding of the polypeptides encoded by the inserts to the promoter DNA may be confirmed by techniques familiar to those skilled in the art, such as gel shift analysis or DNAse protection analysis.
  • the present invention also comprises the use of 5'ESTs (or cDNA or genomic DNA obtainable therefrom) in gene therapy strategies, including antisense and triple helix strategies as described in Examples 62 and 63 below.
  • antisense approaches nucleic acid sequences complementary to an mRNA are hybridized to the mRNA intracellularly, thereby blocking the expression of the protein encoded by the mRNA.
  • the antisense sequences may prevent gene expression through a variety of mechanisms.
  • the antisense sequences may inhibit the ability of ribosomes to translate the mRNA.
  • the antisense sequences may block transport of the mRNA from the nucleus to the cytoplasm, thereby limiting the amount of mRNA available for translation.
  • antisense sequences may inhibit gene expression is by interfering with mRNA splicing.
  • the antisense nucleic acid may be inco ⁇ orated in a ribozyme capable of specifically cleaving the target mRNA.
  • the antisense nucleic acid molecules to be used in gene therapy may be either DNA or RNA sequences. They may comprise a sequence complementary to the sequence of the 5'EST (or cDNA or genomic DNA obtainable therefrom).
  • the antisense nucleic acids should have a length and melting temperature sufficient to permit formation of an intracellular duplex with sufficient stability to inhibit the expression of the mRNA in the duplex.
  • Strategies for designing antisense nucleic acids suitable for use in gene therapy are disclosed in Green et al, Ann. Rev. Biochem. 55:569-597, 1986; and Izant and Weintraub, Cell 36:1007-1015, 1984, which are hereby inco ⁇ orated by reference.
  • antisense molecules are obtained from a nucleotide sequence encoding a protein by reversing the orientation of the coding region with respect to a promoter so as to transcribe the opposite strand from that which is normally transcribed in the cell.
  • the antisense molecules may be transcribed using in vitro transcription systems such as those which employ T7 or SP6 polymerase to generate the transcript.
  • Another approach involves transcription of the antisense nucleic acids in vivo by operably linking DNA containing the antisense sequence to a promoter in an expression vector.
  • oligonucleotides which are complementary to the strand normally transcribed in the cell may be synthesized in vitro.
  • the antisense nucleic acids are complementary to the corresponding mRNA and are capable of hybridizing to the mRNA to create a duplex.
  • the antisense sequences may contain modified sugar phosphate backbones to increase stability and make them less sensitive to RNase activity. Examples of modifications suitable for use in antisense strategies are described by Rossi et al, Pharmacol. Ther. 50(2):245-254, 1991, which is hereby inco ⁇ orated by reference.
  • antisense oligonucleotides complementary to the sequence of the 5'EST may be used.
  • stable and semi-stable antisense oligonucleotides described in International Application No. PCT WO94/23026, hereby inco ⁇ orated by reference are used.
  • the 3' end or both the 3' and 5' ends are engaged in intramolecular hydrogen bonding between complementary base pairs.
  • these molecules are better able to withstand exonuclease attacks and exhibit increased stability compared to conventional antisense oligonucleotides.
  • the antisense oligodeoxynucleotides against he ⁇ es simplex virus types 1 and 2 described in International Application No. WO 95/04141, hereby inco ⁇ orated by reference are used.
  • the covalently cross-linked antisense oligonucleotides described in International Application No. WO 96/31523, hereby inco ⁇ orated by reference are used.
  • These double- or single-stranded oligonucleotides comprise one or more, respectively, inter- or intra-oligonucleotide covalent cross-linkages, wherein the linkage consists of an amide bond between a primary amine group of one strand and a carboxyl group of the other strand or of the same strand, respectively, the primary amine group being directly substituted in the 2' position of the strand nucleotide monosaccharide ring, and the carboxyl group being carried by an aliphatic spacer group substituted on a nucleotide or nucleotide analog of the other strand or the same strand, respectively.
  • the antisense oligodeoxynucleotides and oligonucleotides disclosed in International Application No. WO 92/18522, inco ⁇ orated by reference, may also be used. These molecules are stable to degradation and contain at least one transcription control recognition sequence which binds to control proteins and are effective as decoys therefore. These molecules may contain "hairpin” structures, “dumbbell” structures, “modified dumbbell” structures, "cross-linked” decoy structures and "loop” structures.
  • the cyclic double-stranded oligonucleotides described in European Patent Application No. 0 572 287 A2, hereby inco ⁇ orated by reference are used.
  • These ligated oligonucleotide "dumbbells" contain the binding site for a transcription factor and inhibit expression of the gene under control of the transcription factor by sequestering the factor.
  • oligonucleotides disclosed in International Application No. WO 92/19732, hereby inco ⁇ orated by reference, is also contemplated. Because these molecules have no free ends, they are more resistant to degradation by exonucleases than are conventional oligonucleotides. These oligonucleotides may be multifunctional, interacting with several regions which are not adjacent to the target mRNA.
  • the appropriate level of antisense nucleic acids required to inhibit gene expression may be determined using in vitro expression analysis.
  • the antisense molecule may be introduced into the cells by diffusion, injection, infection, transfection or h-region-mediated import using procedures known in the art.
  • the antisense nucleic acids can be introduced into the body as a bare or naked oligonucleotide, oligonucleotide encapsulated in lipid, oligonucleotide sequence encapsidated by viral protein, or as an oligonucleotide operably linked to a promoter contained in an expression vector.
  • the expression vector may be any of a variety of expression vectors known in the art, including retroviral or viral vectors, vectors capable of extrachromosomal replication, or integrating vectors
  • the vectors may be DNA or RNA.
  • the antisense molecules are introduced onto cell samples at a number of different concentrations preferably between lxlO "10 M to lxlO ⁇ M. Once the minimum concentration that can adequately control gene expression is identified, the optimized dose is translated into a dosage suitable for use in vivo. For example, an inhibiting concentration in culture of lxlO "7 translates into a dose of approximately 0 6 mg/kg bodyweight. Levels of oligonucleotide approaching 100 mg/kg bodyweight or higher may be possible after testing the toxicity of the oligonucleotide in laboratory animals It is additionally contemplated that cells from the vertebrate are removed, treated with the antisense oligonucleotide, and reintroduced into the vertebrate.
  • the antisense oligonucleotide sequence is inco ⁇ orated into a ribozyme sequence to enable the antisense to specifically bind and cleave its target mRNA.
  • ribozyme and antisense oligonucleotides see Rossi et al, supra.
  • the polypeptide encoded by the gene is first identified, so that the effectiveness of antisense inhibition on translation can be monitored using techniques that include but are not limited to antibody-mediated tests such as RIAs and ELISA, functional assays, or radiolabeling
  • the 5' ESTs of the present invention may also be used in gene therapy approaches based on intracellular triple helix formation.
  • Triple helix oligonucleotides are used to inhibit transcription from a genome. They are particularly useful for studying alterations in cell activity as it is associated with a particular gene.
  • the 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom) of the present invention or, more preferably, a portion of those sequences, can be used to inhibit gene expression in individuals having diseases associated with expression of a particular gene.
  • a portion of 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom) can be used to study the effect of inhibiting transcription of a particular gene within a cell.
  • homopurine sequences were considered the most useful for triple helix strategies.
  • homopyrimidine sequences can also inhibit gene expression Such homopyrimidine oligonucleotides bind to the major groove at homopurine:homopyrimidine sequences.
  • both types of sequences from the 5'EST or from the gene corresponding to the 5'EST are contemplated within the scope of this invention.
  • oligonucleotides containing the candidate sequences into tissue culture cells which normally express the target gene.
  • the oligonucleotides may be prepared on an oligonucleotide synthesizer or they may be purchased commercially from a company specializing in custom oligonucleotide synthesis, such as GENSET, Paris, France.
  • oligonucleotides may be introduced into the cells using a variety of methods known to those skilled in the art, including but not limited to calcium phosphate precipitation, DEAE-Dextran, electroporation, liposome-mediated transfection or native uptake.
  • Treated cells are monitored for altered cell function or reduced gene expression using techniques such as Northern blotting, RNase protection assays, or PCR based strategies to monitor the transcription levels of the target gene in cells which have been treated with the oligonucleotide.
  • the cell functions to be monitored are predicted based upon the homologies of the target gene corresponding to the extended cDNA from which the oligonucleotide was derived with known gene sequences that have been associated with a particular function.
  • the cell functions can also be predicted based on the presence of abnormal physiologies within cells derived from individuals with a particular inherited disease, particularly when the extended cDNA is associated with the disease using techniques described in Example 56.
  • the oligonucleotides which are effective in inhibiting gene expression in tissue culture cells may then be introduced in vivo using the techniques described above and in Example 62 at a dosage calculated based on the in vitro results, as described in Example 62.
  • the natural (beta) anomers of the oligonucleotide units can be replaced with alpha anomers to render the oligonucleotide more resistant to nucleases.
  • an intercalating agent such as ethidium bromide, or the like, can be attached to the 3' end of the alpha oligonucleotide to stabilize the triple helix.
  • the cDNAs obtained as described above using the 5' ESTs of the present invention may also be used to express an encoded protein in a host organism to produce a beneficial effect.
  • the encoded protein may be transiently expressed in the host organism or stably expressed in the host organism.
  • the encoded protein may have any of the activities described above.
  • the encoded protein may be a protein which the host organism lacks or, alternatively, the encoded protein may augment the existing levels of the protein in the host organism.
  • a full length extended cDNA encoding the signal peptide and the mature protein, or an extended cDNA encoding only the mature protein is introduced into the host organism.
  • the extended cDNA may be introduced into the host organism using a variety of techniques known to those of skill in the art.
  • the extended cDNA may be injected into the host organism as naked DNA such that the encoded protein is expressed in the host organism, thereby producing a beneficial effect.
  • the extended cDNA may be cloned into an expression vector downstream of a promoter which is active in the host organism.
  • the expression vector may be any of the expression vectors designed for use in gene therapy, including viral or retroviral vectors.
  • the expression vector may be directly introduced into the host organism such that the encoded protein is expressed in the host organism to produce a beneficial effect.
  • the expression vector may be introduced into cells in vitro. Cells containing the expression vector are thereafter selected and introduced into the host organism, where they express the encoded protein to produce a beneficial effect.
  • the short core hydrophobic region (h) of signal peptides encoded by the 5'ESTS or extended cDNAs derived from SEQ ID NOs: 38-305 may also be used as a carrier to import a peptide or a protein of interest, so-called cargo, into tissue culture cells (Lin et al, J. Biol.
  • nucleic acids can be genetically engineered, using techniques familiar to those skilled in the art, in order to link the extended cDNA sequence encoding the h region to the 5' or the 3' end of a DNA sequence coding for a cargo polypeptide.
  • Such genetically engineered nucleic acids are then translated either in vitro or in vivo after transfection into appropriate cells, using conventional techniques to produce the resulting cell permeable polypeptide. Suitable hosts cells are then simply incubated with the cell permeable polypeptide which is then translocated across the membrane.
  • This method may be applied to study diverse intracellular functions and cellular processes. For instance, it has been used to probe functionally relevant domains of intracellular proteins and to examine protein-protein interactions involved in signal transduction pathways (Lin et al, supra; Lin et al, J. Biol. Chem., 271: 5305-5308, 1996; Rojas et al, Biol Chem., 271: 27456-27461, 1996; Liu et al, Proc.
  • Such techniques may be used in cellular therapy to import proteins producing therapeutic effects. For instance, cells isolated from a patient may be treated with imported therapeutic proteins and then re-introduced into the host organism.
  • the h region of signal peptides of the present invention could be used in combination with a nuclear localization signal to deliver nucleic acids into cell nucleus.
  • oligonucleotides may be antisense oligonucleotides or oligonucleotides designed to form triple helixes, as described in examples 62 and 63 respectively, in order to inhibit processing and/or maturation of a target cellular RNA.
  • the cDNAs or portions thereof obtained using the 5' ESTs of the present invention can be used for various pu ⁇ oses.
  • the polynucleotides can be used to express recombinant protein for analysis, characterization or therapeutic use; as markers for tissues in which the corresponding protein is preferentially expressed (either constitutively or at a particular stage of tissue differentiation or development or in disease states); as molecular weight markers on Southern gels; as chromosome markers or tags (when labeled) to identify chromosomes or to map related gene positions; to compare with endogenous DNA sequences in patients to identify potential genetic disorders; as probes to hybridize and thus discover novel, related DNA sequences; as a source of information to derive PCR primers for genetic finge ⁇ rinting; for selecting and making oligomers for attachment to a "gene chip” or other support, including for examination for expression patterns; to raise anti-protein antibodies using DNA immunization techniques; and as an antigen to raise anti-DNA antibodies or elicit another immune response.
  • the polynucleotide encodes a protein which binds or potentially binds to another protein (such as, for example, in a receptor-ligand interaction)
  • the polynucleotide can also be used in interaction trap assays (such as, for example, that described in Gyuris et al, Cell 75:791-803, 1993, the disclosure of which is hereby inco ⁇ orated by reference) to identify polynucleotides encoding the other protein with which binding occurs or to identify inhibitors of the binding interaction.
  • the proteins or polypeptides provided by the present invention can similarly be used in assays to determine biological activity, including in a panel of multiple proteins for high- throughput screening; to raise antibodies or to elicit another immune response; as a reagent (including the labeled reagent) in assays designed to quantitatively determine levels of the protein (or its receptor) in biological fluids; as markers for tissues in which the corresponding protein is preferentially expressed (either constitutively or at a particular stage of tissue differentiation or development or in a disease state); and, of course, to isolate correlative receptors or ligands.
  • the protein binds or potentially binds to another protein (such as, for example, in a receptor-ligand interaction)
  • the protein can be used to identify the other protein with which binding occurs or to identify inhibitors of the binding interaction. Proteins involved in these binding interactions can also be used to screen for peptide or small molecule inhibitors or agonists of the binding interaction.
  • Polynucleotides and proteins of the present invention can also be used as nutritional sources or supplements. Such uses include without limitation use as a protein or amino acid supplement, use as a carbon source, use as a nitrogen source and use as a source of carbohydrate.
  • the protein or polynucleotide of the invention can be added to the feed of a particular organism or can be administered as a separate solid or liquid preparation, such as in the form of powder, pills, solutions, suspensions or capsules.
  • the protein or polynucleotide of the invention can be added to the medium in or on which the microorganism is cultured.
  • ID128 new 5 Fetal kidney 58-41-3-B4-PU
  • ID139 new 4.7 Fetal kidney- 58-8-1-G7-PU
  • ID171 new 4.1 Fetal kidney- 58-53-3-G4-PU
  • ID209 new 3.5 Fetal kidney 58-41-4-G9-PU
  • ID237 est-not-ext 11.2 Fetal kidney 58-7-2-A7-PU
  • ID241 est-not-ext 9.6 Fetal kidney 58-40-1-F5-PU
  • ID242 est-not-ext 9.5 Fetal kidney 58-6-4-G2-PU
  • ID244 est-not-ext 8.9 Fetal kidney 58-48-1-Al l-PU
  • ID245 est-not-ext 8.8 Fetal kidney 58-35-2-B6-PU
  • ID247 est-not-ext 8.4 Fetal kidney 58-45-1-E6-PU
  • ID248 est-not-ext 8.1 Fetal kidney 58-39-1-A12-PU
  • ID251 est-not-ext 7.9 Fetal kidney 58-17-2-D9-PU
  • ID252 est-not-ext 7.9 Fetal kidney 58-52-3-B7-PU
  • ID262 est-not-ext 6.6 Fetal kidney 58-9-4-F6-PU
  • ID263 est-not-ext 6.4 Fetal kidney 58-1-1-E3-PU
  • ID268 est-not-ext 6.2 Fetal kidney 58-33-1-Fl-PU
  • ID269 est-not-ext 5.9 Fetal kidney 58-48-4-H2-PU
  • ID274 est-not-ext 5.5 Fetal kidney 58-54-1-D 11-PU
  • ID276 est-not-ext 5.4 Fetal kidney 58-30-2-H 10-PU
  • ID277 est-not-ext 5.3 Fetal kidney 58-29-1-Hl-PU
  • ID280 est-not-ext 5 Fetal kidney 58-34-3-H10-PU
  • ID282 est-not-ext 4.9 Fetal kidney 58-4-2-D 12-PU
  • ID284 est-not-ext 4.7 Fetal kidney 58-37-3-C10-PU
  • ID286 est-not-ext 4.6 Fetal kidney 58-52-1-Al l-PU
  • ID292 est-not-ext 4.1 Fetal kidney 58-33-2-C6-PU
  • ID302 est-not-ext 3.5 Fetal kidney 58-54-1-E6-PU
  • ID304 est-not-ext 3.5 Fetal kidney- 58-23-4-F4-PU

Abstract

The sequences of 5' ESTs derived from mRNAs encoding secreted proteins are disclosed. The 5' ESTs may be to obtain cDNAs and genomic DNAs corresponding to the 5' ESTs. The 5' ESTs may also be used in diagnostic, forensic, gene therapy, and chromosome mapping procedures. Upstream regulatory sequences may also be obtained using the 5' ESTs. The 5' ESTs may also be used to design expression vectors and secretion vectors.

Description

5' ESTs FOR SECRETED PROTEINS EXPRESSED IN MUSCLE AND OTHER MESODERMAL TISSUES
Background of the Invention The estimated 50,000-100,000 genes scattered along the human chromosomes offer tremendous promise for the understanding, diagnosis, and treatment of human diseases. In addition, probes capable of specifically hybridizing to loci distributed throughout the human genome find applications in the construction of high resolution chromosome maps and in the identification of individuals.
In the past, the characterization of even a single human gene was a painstaking process, requiring years of effort. Recent developments in the areas of cloning vectors, DNA sequencing, and computer technology have merged to greatly accelerate the rate at which human genes can be isolated, sequenced, mapped, and characterized. Cloning vectors such as yeast artificial chromosomes (YACs) and bacterial artificial chromosomes (BACs) are able to accept DNA inserts ranging from 300 to 1000 kilobases (kb) or 100-400 kb in length respectively, thereby facilitating the manipulation and ordering of DNA sequences distributed over great distances on the human chromosomes. Automated DNA sequencing machines permit the rapid sequencing of human genes. Bioinformatics software enables the comparison of nucleic acid and protein sequences, thereby assisting in the characterization of human gene products. Currently, two different approaches are being pursued for identifying and characterizing the genes distributed along the human genome. In one approach, large fragments of genomic DNA are isolated, cloned, and sequenced. Potential open reading frames in these genomic sequences are identified using bioinformatics software. However, this approach entails sequencing large stretches of human DNA which do not encode proteins in order to find the protein encoding sequences scattered throughout the genome. In addition to requiring extensive sequencing, the bioinformatics software may mischaracterize the genomic sequences obtained. Thus, the software may produce false positives in which non- coding DNA is mischaracterized as coding DNA or false negatives in which coding DNA is mislabeled as non-coding DNA. An alternative approach takes a more direct route to identifying and characterizing human genes. In this approach, complementary DNAs (cDNAs) are synthesized from isolated messenger RNAs (mRNAs) which encode human proteins. Using this approach, sequencing is only performed on DNA which is derived from protein coding portions of the genome. Often, only short stretches of the cDNAs are sequenced to obtain sequences called expressed sequence tags (ESTs). The ESTs may then be used to isolate or purify extended cDNAs which include sequences adjacent to the EST sequences. The extended cDNAs may contain all of the sequence of the EST which was used to obtain them or only a portion of the sequence of the EST which was used to obtain them. In addition, the extended cDNAs may contain the full coding sequence of the gene from which the EST was derived or, alternatively, the extended cDNAs may include portions of the coding sequence of the gene from which the EST was derived. It will be appreciated that there may be several extended cDNAs which include the EST sequence as a result of alternate splicing or the activity of alternative promoters.
In the past, these short EST sequences were often obtained from oligo-dT primed cDNA libraries. Accordingly, they mainly corresponded to the 3' untranslated region of the mRNA. In part, the prevalence of EST sequences derived from the 3' end of the mRNA is a result of the fact that typical techniques for obtaining cDNAs are not well suited for isolating cDNA sequences derived from the 5' ends of mRNAs. (Adams et al., Nature 377:3-174, 1996; Hfflier e/ ar/., Genome Res. 6:807-828, 1996).
In addition, in those reported instances where longer cDNA sequences have been obtained, the reported sequences typically correspond to coding sequences and do not include the full 5' untranslated region of the mRNA from which the cDNA is derived. Such incomplete sequences may not include the first exon of the mRNA, particularly in situations where the first exon is short. Furthermore, they may not include some exons, often short ones, which are located upstream of splicing sites. Thus, there is a need to obtain sequences derived from the 5' ends of mRNAs.
While many sequences derived from human chromosomes have practical applications, approaches based on the identification and characterization of those chromosomal sequences which encode a protein product are particularly relevant to diagnostic and therapeutic uses. Of the 50,000-100,000 protein coding genes, those genes encoding proteins which are secreted from the cell in which they are synthesized, as well as the secreted proteins themselves, are particularly valuable as potential therapeutic agents. Such proteins are often involved in cell to cell communication and may be responsible for producing a clinically relevant response in their target cells.
In fact, several secretory proteins, including tissue plasminogen activator, G-CSF, GM-CSF, erythropoietin, human growth hormone, insulin, interferon-α, interferon-β, interferon-γ, and interleukin-2, are currently in clinical use. These proteins are used to treat a wide range of conditions, including acute myocardial infarction, acute ischemic stroke, anemia, diabetes, growth hormone deficiency, hepatitis, kidney carcinoma, chemotherapy induced neutropenia and multiple sclerosis. For these reasons, extended cDNAs encoding secreted proteins or portions thereof represent a particularly valuable source of therapeutic agents. Thus, there is a need for the identification and characterization of secreted proteins and the nucleic acids encoding them.
In addition to being therapeutically useful themselves, secretory proteins include short peptides, called signal peptides, at their amino termini which direct their secretion. These signal peptides are encoded by the signal sequences located at the 5' ends of the coding sequences of genes encoding secreted proteins. Because these signal peptides will direct the extracellular secretion of any protein to which they are operably linked, the signal sequences may be exploited to direct the efficient secretion of any protein by operably linking the signal sequences to a gene encoding the protein for which secretion is desired. In addition, portions of signal sequences may also be used to direct the intracellular import of a peptide or protein of interest. This may prove beneficial in gene therapy strategies in which it is desired to deliver a particular gene product to cells other than the cell in which it is produced. Signal sequences encoding signal peptides also find application in simplifying protein purification techniques. In such applications, the extracellular secretion of the desired protein greatly facilitates purification by reducing the number of undesired proteins from which the desired protein must be selected. Thus, there exists a need to identify and characterize the 5' portions of the genes for secretory proteins which encode signal peptides.
Public information on the number of human genes for which the promoters and upstream regulatory regions have been identified and characterized is quite limited. In part, this may be due to the difficulty of isolating such regulatory sequences. Upstream regulatory sequences such as transcription factor binding sites are typically too short to be utilized as probes for isolating promoters from human genomic libraries. Recently, some approaches have been developed to isolate human promoters. One of them consists of making a CpG island library (Cross, et al., Nature Genetics 6: 236-244, 1994). The second consists of isolating human genomic DNA sequences containing Spel binding sites by the use of Spel binding protein. (Mortlock et al., Genome Res. 6:327-335, 1996). Both of these approaches have their limits due to a lack of specificity or of comprehensiveness.
The present 5' ESTs may be used to efficiently identify and isolate upstream regulatory regions which control the location, developmental stage, rate, and quantity of protein synthesis, as well as the stability of the mRNA. (Theil, BioFactors 4:87-93, 1993). Once identified and characterized, these regulatory regions may be utilized in gene therapy or protein purification schemes to obtain the desired amount and locations of protein synthesis or to inhibit, reduce, or prevent the synthesis of undesirable gene products.
In addition, ESTs containing the 5' ends of secretory protein genes may include sequences useful as probes for chromosome mapping and the identification of individuals. Thus, there is a need to identify and characterize the sequences upstream of the 5' coding sequences of genes encoding secretory proteins.
Summary of the Invention The present invention relates to purified, isolated, or recombinant ESTs which include sequences derived from the authentic 5' ends of their corresponding mRNAs. The term "corresponding mRNA" refers to the mRNA which was the template for the cDNA synthesis which produced the 5' EST. These sequences will be referred to hereinafter as "5' ESTs." As used herein, the term "purified" does not require absolute purity; rather, it is intended as a relative definition. Individual 5' EST clones isolated from a cDNA library have been conventionally purified to electrophoretic homogeneity. The sequences obtained from these clones could not be obtained directly either from the library or from total human DNA. The cDNA clones are not naturally occurring as such, but rather are obtained via manipulation of a partially purified naturally occurring substance (messenger RNA). The conversion of mRNA into a cDNA library involves the creation of a synthetic substance (cDNA) and pure individual cDNA clones can be isolated from the synthetic library by clonal selection. Thus, creating a cDNA library from messenger RNA and subsequently isolating individual clones from that library results in an approximately 104- 106 fold purification of the native message. Purification of starting material or natural material to at least one order of magnitude, preferably two or three orders, and more preferably four or five orders of magnitude is expressly contemplated.
As used herein, the term "isolated" requires that the material be removed from its original environment (e.g., the natural environment if it is naturally occurring). For example, a naturally-occurring polynucleotide present in a living animal is not isolated, but the same polynucleotide, separated from some or all of the coexisting materials in the natural system, is isolated.
As used herein, the term "recombinant" means that the 5' EST is adjacent to "backbone" nucleic acid to which it is not adjacent in its natural environment. Additionally, to be "enriched" the 5' ESTs will represent 5% or more of the number of nucleic acid inserts in a population of nucleic acid backbone molecules. Backbone molecules according to the present invention include nucleic acids such as expression vectors, self-replicating nucleic acids, viruses, integrating nucleic acids, and other vectors or nucleic acids used to maintain or manipulate a nucleic acid insert of interest. Preferably, the enriched 5' ESTs represent 15% or more of the number of nucleic acid inserts in the population of recombinant backbone molecules. More preferably, the enriched 5' ESTs represent 50% or more of the number of nucleic acid inserts in the population of recombinant backbone molecules. In a highly preferred embodiment, the enriched 5' ESTs represent 90% or more of the number of nucleic acid inserts in the population of recombinant backbone molecules.
"Stringent", moderate," and "low" hybridization conditions are as defined in Example 29.
Unless otherwise indicated, a "complementary" sequence is fully complementary. Thus, 5' ESTs in cDNA libraries in which one or more 5' ESTs make up 5% or more of the number of nucleic acid inserts in the backbone molecules are "enriched recombinant 5' ESTs" as defined herein. Likewise, 5' ESTs in a population of plasmids in which one or more 5' EST of the present invention have been inserted such that they represent 5% or more of the number of inserts in the plasmid backbone are " enriched recombinant 5' ESTs" as defined herein. However, 5' ESTs in cDNA libraries in which 5' ESTs constitute less than 5% of the number of nucleic acid inserts in the population of backbone molecules, such as libraries in which backbone molecules having a 5' EST insert are extremely rare, are not "enriched recombinant 5' ESTs."
In particular, the present invention relates to 5' ESTs which are derived from genes encoding secreted proteins. As used herein, a "secreted" protein is one which, when expressed in a suitable host cell, is transported across or through a membrane, including transport as a result of signal peptides in its amino acid sequence. "Secreted" proteins include without limitation proteins secreted wholly (e.g. soluble proteins), or partially (e.g. receptors) from the cell in which they are expressed. "Secreted" proteins also include without limitation proteins which are transported across the membrane of the endoplasmic reticulum. Such 5' ESTs include nucleic acid sequences, called signal sequences, which encode signal peptides which direct the extracellular secretion of the proteins encoded by the genes from which the 5' ESTs are derived. Generally, the signal peptides are located at the amino termini of secreted proteins.
Secreted proteins are translated by ribosomes associated with the "rough" endoplasmic reticulum. Generally, secreted proteins are co-translationally transferred to the membrane of the endoplasmic reticulum. Association of the ribosome with the endoplasmic reticulum during translation of secreted proteins is mediated by the signal peptide. The signal peptide is typically cleaved following its co-translational entry into the endoplasmic reticulum.
After delivery to the endoplasmic reticulum, secreted proteins may proceed through the Golgi apparatus. In the Golgi apparatus, the proteins may undergo post-translational modification before entering secretory vesicles which transport them across the cell membrane.
The 5' ESTs of the present invention have several important applications. For example, they may be used to obtain and express cDNA clones which include the full protein coding sequences of the corresponding gene products, including the authentic translation start sites derived from the 5' ends of the coding sequences of the mRNAs from which the 5' ESTs are derived. These cDNAs will be referred to hereinafter as "full length cDNAs." These cDNAs may also include DNA derived from mRNA sequences upstream of the translation start site. The full length cDNA sequences may be used to express the proteins corresponding to the 5' ESTs. As discussed above, secreted proteins are therapeutically important. Thus, the proteins expressed from the cDNAs may be useful in treating or controlling a variety of human conditions. The 5' ESTs may also be used to obtain the corresponding genomic DNA. The term "corresponding genomic DNA" refers to the genomic DNA which encodes the mRNA from which the 5' EST was derived.
Alternatively, the 5' ESTs may be used to obtain and express extended cDNAs encoding portions of the secreted protein. The portions may comprise the signal peptides of the secreted proteins or the mature proteins generated when the signal peptide is cleaved off. The portions may also comprise polypeptides having at least 10 consecutive amino acids encoded by the extended cDNAs or full length cDNAs. Alternatively, the portions may comprise at least 15 consecutive amino acids encoded by the extended cDNAs or full length cDNAs. In some embodiments, the portions may comprise at least 25 consecutive amino acids encoded by the extended cDNAs or full length cDNAs. In other embodiments, the portions may comprise at least 40 amino acids encoded by the extended cDNAs or full length cDNAs.
Antibodies which specifically recognize the entire secreted proteins encoded by the extended cDNAs, full length cDNAs, or fragments thereof having at least 10 consecutive amino acids, at least 15 consecutive amino acids, at least 25 consecutive amino acids, or at least 40 consecutive amino acids may also be obtained as described below. Antibodies which specifically recognize the mature protein generated when the signal peptide is cleaved may also be obtained as described below. Similarly, antibodies which specifically recognize the signal peptides encoded by the extended cDNAs or full length cDNAs may also be obtained.
In some embodiments, the extended cDNAs obtained using the 5' ESTs include the signal sequence. In other embodiments, the extended cDNAs obtained using the 5' ESTs may include the full coding sequence for the mature protein (i.e. the protein generated when the signal polypeptide is cleaved off). In addition, the extended cDNAs obtained using the 5' ESTs may include regulatory regions upstream of the translation start site or downstream of the stop codon which control the amount, location, or developmental stage of gene expression.
As discussed above, secreted proteins are therapeutically important. Thus, the proteins expressed from the extended cDNAs or full length cDNAs obtained using the 5' ESTs may be useful in treating or controlling a variety of human conditions. The 5' ESTs (or cDNAs or genomic DNAs obtained therefrom) may be used in forensic procedures to identify individuals or in diagnostic procedures to identify individuals having genetic diseases resulting from abnormal expression of the genes corresponding to the 5' ESTs. In addition, the present invention is useful for constructing a high resolution map of the human chromosomes.
The present invention also relates to secretion vectors capable of directing the secretion of a protein of interest. Such vectors may be used in gene therapy strategies in which it is desired to produce a gene product in one cell which is to be delivered to another location in the body. Secretion vectors may also facilitate the purification of desired proteins. The present invention also relates to expression vectors capable of directing the expression of an inserted gene in a desired spatial or temporal manner or at a desired level. Such vectors may include sequences upstream of the 5' ESTs, such as promoters or upstream regulatory sequences.
Finally, the present invention may also be used for gene therapy to control or treat genetic diseases. Signal peptides may also be fused to heterologous proteins to direct their extracellular secretion.
Bacterial clones containing Bluescript plasmids having inserts containing the 5' ESTs of the present invention (SEQ ID NOs: 38-305 are presently stored at 80°C in 4% (v/v) glycerol in the inventor's laboratories under the designations listed next to the SEQ ID NOs in LI). The inserts may be recovered from the deposited materials by growing the appropriate clones on a suitable medium. The Bluescript DNA can then be isolated using plasmid isolation procedures familiar to those skilled in the art such as alkaline lysis minipreps or large scale alkaline lysis plasmid isolation procedures. If desired the plasmid DNA may be further enriched by centriftigation on a cesium chloride gradient, size exclusion chromatography, or anion exchange chromatography. The plasmid DNA obtained using these procedures may then be manipulated using standard cloning techniques familiar to those skilled in the art. Alternatively, a PCR can be done with primers designed at both ends of the EST insertion. The PCR product which corresponds to the 5' EST can then be manipulated using standard cloning techniques familiar to those skilled in the art. One aspect of the present invention is a purified or isolated nucleic acid having the sequence of one of SEQ ED NOs: 38-305 or having a sequence complementary thereto. In one embodiment, the nucleic acid is recombinant.
Another aspect of the present invention is a purified or isolated nucleic acid comprising at least 10 consecutive bases of the sequence of one of SEQ ID NOs: 38-305 or one of the sequences complementary thereto.
Yet another aspect of the present invention is a purified or isolated nucleic acid comprising at least 15 consecutive bases of one of the sequences of SEQ LD NOs: 38-305 or one of the sequences complementary thereto. In one embodiment, the nucleic acid is recombinant.
A further aspect of the present invention is a purified or isolated nucleic acid of at least 15 bases capable of hybridizing under stringent conditions to the sequence of one of SEQ ID NOs: 38-305 or one of the sequences complementary to the sequences of SEQ ID NOs: 38-305. In one embodiment, the nucleic acid is recombinant. Another aspect of the present invention is a purified or isolated nucleic acid encoding a human gene product, said human gene product having a sequence partially encoded by one of the sequences of SEQ ID NO: 38-305.
Still another aspect of the present invention is a method of making a cDNA encoding a human secretory protein, said human secretory protein being partially encoded by one of SEQ ID NOs 38-305, comprising the steps of contacting a collection of mRNA molecules from human cells with a primer comprising at least 15 consecutive nucleotides of a sequence complementary to one of SEQ ID NOs: 38-305; hybridizing said primer to an mRNA in said collection that encodes said protein; reverse transcribing said hybridized primer to make a first cDNA strand from said mRNA; making a second cDNA strand complementary to said first cDNA strand; and isolating the resulting cDNA encoding said protein comprising said first cDNA strand and said second cDNA strand.
Another aspect of the invention is an isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein encoded by one of SEQ ID NOs 38-305 or a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method described in the preceding paragraph. In one embodiment, the cDNA comprises the full protein coding sequence of said protein which sequence is partially included in one of the sequences of SEQ ID NOs: 38-305.
Another aspect of the present invention is a method of making a cDNA encoding a human secretory protein that is partially encoded by one of SEQ ID NOs 38-305, comprising the steps of obtaining a cDNA comprising one of the sequences of SEQ ID NOs: 38-305; contacting said cDNA with a detectable probe comprising at least 15 consecutive nucleotides of said sequence of SEQ LD NO: 38-305 or a sequence complementary thereto under conditions which permit said probe to hybridize to said cDNA; identifying a cDNA which hybridizes to said detectable probe; and isolating said cDNA which hybridizes to said probe. Another aspect of the present invention is an isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein encoded by one of SEQ ID NOs 38-305 or a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method described in the preceding paragraph. In one embodiment, the cDNA comprises the full protein coding sequence partially included in one of the sequences of SEQ ID NOs: 38-305.
Another aspect of the present invention is a method of making a cDNA comprising one of the sequence of SEQ ID NOs: 38-305, comprising the steps of contacting a collection of mRNA molecules from human cells with a first primer capable of hybridizing to the polyA tail of said mRNA; hybridizing said first primer to said polyA tail; reverse transcribing said mRNA to make a first cDNA strand; making a second cDNA strand complementary to said first cDNA strand using at least one primer comprising at least 15 nucleotides of one of the sequences of SEQ ED NOs 38-305; and isolating the resulting cDNA comprising said first cDNA strand and said second cDNA strand.
Another aspect of the present invention is an isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein encoded by one of SEQ ED NOs 38-305 or a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method described in the preceding paragraph. In one embodiment, the cDNA comprises the full protein coding sequence partially included in one of the sequences of SEQ ED NOs: 38-305. In one embodiment of the method described in the two paragraphs above, the second cDNA strand is made by contacting said first cDNA strand with a first pair of primers, said first pair 'of primers comprising a second primer comprising at least 15 consecutive nucleotides of one of the sequences of SEQ ED NOs 38-305 and a third primer having a sequence therein which is included within the sequence of said first primer; performing a first polymerase chain reaction with said first pair of nested primers to generate a first PCR product; contacting said first PCR product with a second pair of primers, said second pair of primers comprising a fourth primer, said fourth primer comprising at least 15 consecutive nucleotides of said sequence of one of SEQ ID NOs: 38-305 , and a fifth primer, said fourth and fifth primers being capable of hybridizing to sequences within said first PCR product; and performing a second polymerase chain reaction, thereby generating a second PCR product. One aspect of the present invention is an isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein encoded by one of SEQ ED NOs 38-305, or a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method of the preceding paragraph. In one embodiment, the cDNA comprises the full protein coding sequence partially included in one of the sequences of SEQ ED NOs: 38-305.
Another aspect of the present invention is the method described four paragraphs above in which the second cDNA strand is made by contacting said first cDNA strand with a second primer comprising at least 15 consecutive nucleotides of the sequences of SEQ ID NOs: 38-305; hybridizing said second primer to said first strand cDNA; and extending said hybridized second primer to generate said second cDNA strand.
Another aspect of the present invention is an isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein partially encoded by one of SEQ ED NOs 38-305 or comprising a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method described in the preceding paragraph. En one embodiment, the cDNA comprises the full protein coding sequence partially included in of one of the sequences of SEQ ED NOs: 38-305.
Another aspect of the present invention is a method of making a protein comprising one of the sequences of SEQ ED NOs: 306-573, comprising the steps of obtaining a cDNA encoding the full protein sequence partially included in one of the sequences of sequence of SEQ ED NOs: 38-305; inserting said cDNA in an expression vector such that said cDNA is operably linked to a promoter; introducing said expression vector into a host cell whereby said host cell produces the protein encoded by said cDNA; and isolating said protein.
Another aspect of the present invention is an isolated protein obtainable by the method described in the preceding paragraph. Another aspect of the present invention is a method of obtaining a promoter DNA comprising the steps of obtaining DNAs located upstream of the nucleic acids of SEQ ID NOs: 38-305 or the sequences complementary thereto; screening said upstream DNAs to identify a promoter capable of directing transcription initiation; and isolating said DNA comprising said identified promoter. In one embodiment, the obtaining step comprises chromosome walking from said nucleic acids of SEQ ID NOs: 38-305 or sequences complementary thereto. In another embodiment, the screening step comprises inserting said upstream sequences into a promoter reporter vector. In another embodiment, the screening step comprises identifying motifs in said upstream DNAs which are transcription factor binding sites or transcription start sites. Another aspect of the present invention is an isolated promoter obtainable by the method described above.
Another aspect of the present invention is an isolated or purified protein comprising one of the sequences of SEQ ID NOs: 306-573.
Another aspect of the present invention is the inclusion of at least one of the sequences of SEQ ED NOs: 38-305, or one of the sequences complementary to the sequences of SEQ ED NOs: 38-305, or a fragment thereof of at least 15 consecutive nucleotides in an array of discrete ESTs or fragments thereof of at least 15 nucleotides in length. In one embodiment, the array includes at least two of the sequences of SEQ ED NOs: 38-305, the sequences complementary to the sequences of SEQ ED NOs: 38-305, or fragments thereof of at least 15 consecutive nucleotides. In another embodiment, the array includes at least five of the sequences of SEQ ID NOs: 38-305, the sequences complementary to the sequences of SEQ ID NOs: 38-305, or fragments thereof of at least 15 consecutive nucleotides.
Another aspect of the present invention is a promoter having a sequence selected from the group consisting of SEQ ED NOs: 31, 34, and 37. Brief Description of the Drawings
Figure 1 is a summary of a procedure for obtaining cDNAs which have been selected to include the 5' ends of the mRNAs from which they derived.
Figure 2 shows the distribution of Von Heijne scores for 5' ESTs in each of the categories described herein and the probability that these 5' ESTs encode a signal peptide.
Figure 3 summarizes a general method used to clone and sequence extended cDNAs containing sequences adjacent to 5' ESTs.
Figure 4 (description of promoters structure isolated from SignalTag 5' ESTs) provides a schematic description of promoters isolated and the way they are assembled with the corresponding 5' tags.
Detailed Description of the Preferred Embodiment Table IV is an analysis of the 43 amino acids located at the N terminus of all human SwissProt proteins to determine the frequency of false positives and false negatives using the techniques for signal peptide identification described herein. Table V shows the distribution of 5' ESTs in each category described herein and the number of 5' ESTs in each category having a given minimum Von Heijne's score.
Table VI shows the distribution of 5' ESTs in each category described herein with respect to the tissue from which the 5' ESTs of the corresponding mRNA were obtained.
Table VII describes the transcription factor binding sites present in each of these promoters.
I. General Methods for Obtaining 5' ESTs derived from mRNAs with intact 5* ends
In order to obtain the 5' ESTs of the present invention, mRNAs with intact 5' ends must be obtained. Currently, there are two approaches for obtaining such mRNAs with intact 5' ends as described below: either chemical (1) or enzymatic (2).
1. Chemical Methods for Obtaining mRNAs having Intact 5' Ends
One of these approaches is a chemical modification method involving derivatization of the 5' ends of the mRNAs and selection of the derivatized mRNAs. The 5' ends of eukaryotic mRNAs possess a structure referred to as a "cap" which comprises a guanosine methylated at the 7 position. The cap is joined to the first transcribed base of the mRNA by a 5', 5 '-triphosphate bond. In some instances, the 5' guanosine is methylated in both the 2 and 7 positions. Rarely, the 5' guanosine is trimethylated at the 2, 7 and 7 positions. In the chemical method for obtaining mRNAs having intact 5' ends, the 5' cap is specifically derivatized and coupled to a reactive group on an immobilizing substrate. This specific derivatization is based on the fact that only the ribose linked to the methylated guanosine at the 5' end of the mRNA and the ribose linked to the base at the 3' tei inus of the mRNA, possess 2', 3'-cis diols.
Optionally, the 2', 3'-cis diol of the 3' terminal ribose may be chemically modified, substituted, converted, or eliminated, leaving only the ribose linked to the methylated guanosine at the 5' end of the mRNA with a 2', 3'-cis diol. A variety of techniques are available for eliminating the 2', 3'-cis diol on the 3' terminal ribose. For example, controlled alkaline hydrolysis may be used to generate mRNA fragments in which the 3' terminal ribose is a 3 '-phosphate, 2'-phosphate or (2', 3')-cyclophosphate. Thereafter, the fragment which includes the original 3' ribose may be eliminated from the mixture through chromatography on an oligodT column. Alternatively, a base which lacks the 2', 3'-cis diol may be added to the 3' end of the mRNA using an RNA ligase such as T4 RNA ligase. Example 1 below describes a method for ligation of a nucleoside diphosphate to the 3' end of messenger RNA.
EXAMPLE 1 Ligation of the Nucleoside Diphosphate pCp to the 3' End of mRNA
One μg of RNA was incubated in a final reaction medium of 10 μl in the presence of 5 U of T phage RNA ligase in the buffer provided by the manufacturer (Gibco - BRL), 40 U of the RNase inhibitor RNasin (Promega) and, 2 μl of 32pCp (Amersham #PB 10208). The incubation was performed at 37°C for 2 hours or overnight at 7-8°C.
Following modification or elimination of the 2', 3'-cis diol at the 3' ribose, the 2', 3'- cis diol present at the 5' end of the mRNA may be oxidized using reagents such as NaBH,, NaBH3CN, or sodium periodate, thereby converting the 2', 3'-cis diol to a dialdehyde. Example 2 describes the oxidation of the 2', 3'-cis diol at the 5' end of the mRNA with sodium periodate.
EXAMPLE 2 Oxidation of 2'. 3'-cis diol at the 5' End of the mRNA with Sodium Periodate
0.1 OD unit of either a capped oligoribonucleotide of 47 nucleotides (including the cap) or an uncapped oligoribonucleotide of 46 nucleotides were treated as follows. The oligoribonucleotides were produced by in vitro transcription using the transcription kit "AmpliScribe T7" (Epicentre Technologies). As indicated below, the DNA template for the RNA transcript contained a single cytosine. To synthesize the uncapped RNA, all four NTPs were included in the in vitro transcription reaction. To obtain the capped RNA, GTP was replaced by an analogue of the cap, m7G(5')ppp(5')G. This compound, recognized by the polymerase, was incoφorated into the 5' end of the nascent transcript during the initiation of transcription but was not incoφorated during the extension step. Consequently, the resulting RNA contained a cap at its 5' end. The sequences of the oligoribonucleotides produced by the in vitro transcription reaction were: +Cap:
5'm7GpppGCAUCCUACUCCCAUCCAAUUCCACCCUAACUCCUCCCAUCUCCAC- 3' (SEQ ED NO: 1) -Cap:
5'-pppGCAUCCUACUCCCAUCCAAUUCCACCCUAACUCCUCCCAUCUCCAC-3' (SEQ ED NO:2)
The oligoribonucleotides were dissolved in 9 μl of acetate buffer (0.1 M sodium acetate, pH 5.2) and 3 μl of freshly prepared 0.1 M sodium periodate solution. The mixture was incubated for 1 hour in the dark at 4°C or room temperature. Thereafter, the reaction was stopped by adding 4 μl of 10% ethylene glycol. The product was ethanol precipitated, resuspended in at least 10 μl of water or appropriate buffer and dialyzed against water.
The resulting aldehyde groups may then be coupled to molecules having a reactive amine group, such as hydrazine, carbazide, thiocarbazide or semicarbazide groups, in order to facilitate enrichment of the 5' ends of the mRNAs. Molecules having reactive amine groups which are suitable for use in selecting mRNAs having intact 5' ends include avidin, proteins, antibodies, vitamins, ligands capable of specifically binding to receptor molecules, or oligonucleotides. Example 3 below describes the coupling of the resulting dialdehyde to biotin.
EXAMPLE 3 Coupling of the Dialdehyde at the 5' End of Transcripts with Biotin The oxidation product obtained in Example 2 was dissolved in 50 μl of sodium acetate at a pH between 5 and 5.2 and 50 μl of freshly prepared 0.02 M solution of biotin hydrazide in a methoxyethanol/water mixture ( 1 : 1 ) of formula:
Ln the compound used in these experiments, n=5. However, it will be appreciated that other commercially available hydrazides may also be used, such as molecules of the above formula in which n varies from 0 to 5. The mixture was then incubated for 2 hours at 37°C, precipitated with ethanol and dialyzed against distilled water. Example 4 demonstrates the specificity of the biotinylation reaction.
EXAMPLE 4 Specificity of Biotinylation of Capped Transcripts
The specificity of the biotinylation for capped mRNAs was evaluated by gel electrophoresis of the following samples:
Sample 1. The 46 nucleotide uncapped in vitro transcript prepared as in Example 2 and labeled with j2pCp as described in Example 1. Sample 2. The 46 nucleotide uncapped in vitro transcript prepared as in Example 2, labeled with o2pCp as described in Example 1, treated with the oxidation reaction of Example 2, and subjected to the biotinylation conditions of Example 3. Sample 3. The 47 nucleotide capped in vitro transcript prepared as in Example 2 and labeled with 32pCp as described in Example 1.
Sample 4. The 47 nucleotide capped in vitro transcript prepared as in Example 2, labeled with 32pCp as described in Example 1, treated with the oxidation reaction of Example 2, and subjected to the biotinylation conditions of Example 3.
Samples 1 and 2 had identical migration rates, demonstrating that the uncapped RNAs were not oxidized and biotinylated. Sample 3 migrated more slowly than Samples 1 and 2, while Sample 4 exhibited the slowest migration. The difference in migration of the RNAs in Samples 3 and 4 demonstrates that the capped RNAs were specifically biotinylated.
Ln some cases, mRNAs having intact 5' ends may be enriched by binding the molecule containing a reactive amine group to a suitable solid phase substrate such as the inside of the vessel containing the mRNAs, magnetic beads, chromatography matrices, or nylon or nitrocellulose membranes. For example, where the molecule having a reactive amine group is biotin, the solid phase substrate may be coupled to avidin or streptavidin. Alternatively, where the molecule having the reactive amine group is an antibody or receptor ligand, the solid phase substrate may be coupled to the cognate antigen or receptor. Finally, where the molecule having a reactive amine group comprises an oligonucleotide, the solid phase substrate may comprise a complementary oligonucleotide. The mRNAs having intact 5' ends may be released from the solid phase following the enrichment procedure. For example, where the dialdehyde is coupled to biotin hydrazide and the solid phase comprises streptavidin, the mRNAs may be released from the solid phase by simply heating to 95 degrees Celsius in 2% SDS. In some methods, the molecule having a reactive amine group may also be cleaved from the mRNAs having intact 5' ends following enrichment. Example 5 describes the capture of biotinylated mRNAs with streptavidin coated beads and the release of the biotinylated mRNAs from the beads following enrichment.
EXAMPLE 5
Capture and Release of Biotinylated mRNAs Using Streptavidin Coated Beads The streptavidin coated magnetic beads were prepared according to the manufacturer's instructions (CPG Inc., USA). The biotinylated mRNAs were added to a hybridization buffer (1.5 M NaCl, pH 5 - 6). After incubating for 30 minutes, the unbound and nonbiotinylated material was removed. The beads were then washed several times in water with 1% SDS. The beads thus obtained were incubated for 15 minutes at 95°C in water containing 2% SDS. Example 6 demonstrates the efficiency with which biotinylated mRNAs were recovered from the streptavidin coated beads.
EXAMPLE 6
Efficiency of Recovery of Biotinylated mRNAs The efficiency of the recovery procedure was evaluated as follows. Capped RNAs were labeled with 32pCp, oxidized, biotinylated and bound to streptavidin coated beads as described above. Subsequently, the bound RNAs were incubated for 5, 15 or 30 minutes at 95°C in the presence of 2% SDS.
The products of the reaction were analyzed by electrophoresis on 12% polyacrylamide gels under denaturing conditions (7 M urea). The gels were subjected to autoradiography. During this manipulation, the hydrazone bonds were not reduced.
Increasing amounts of nucleic acids were recovered as incubation times in 2% SDS increased, demonstrating that biotinylated mRNAs were efficiently recovered.
In an alternative method for obtaining mRNAs having intact 5' ends, an oligonucleotide which has been derivatized to contain a reactive amine group is specifically coupled to mRNAs having an intact cap. Preferably, the 3' end of the mRNA is blocked prior to the step in which the aldehyde groups are joined to the derivatized oligonucleotide, as described above, so as to prevent the derivatized oligonucleotide from being joined to the 3' end of the mRNA. For example, pCp may be attached to the 3' end of the mRNA using T4 RNA ligase as described in example 1. However, as discussed above, blocking the 3' end of the mRNA is an optional step. Derivatized oligonucleotides may be prepared as described in Example 7. EXAMPLE 7
Derivatization of Oligonucleotides
An oligonucleotide phosphorylated at its 3' end was converted to a 3' hydrazide in 3' by treatment with an aqueous solution of hydrazine or of dihydrazide of the formula H2N(R1)NH2 at about 1 to 3 M, and at pH 4 5 at a temperature of 8°C overnight This incubation was performed in the presence of a carbodiimide type agent soluble in water such as l-ethyl-3-(3-dimethylaminopropyl)carbodii ide at a final concentration of 0 3 M
The derivatized oligonucleotide was then separated from the other agents and products using a standard technique for isolating oligonucleotides As discussed above, the mRNAs to be enriched may be treated to eliminate the 3' OH groups which may be present thereon This may be accomplished by enzymatic ligation of sequences lacking a 3' OH, such as pCp, as descπbed in Example 1 Alternatively, the 3' OH groups may be eliminated by alkaline hydrolysis as descnbed in Example 8 below
EXAMPLE 8
Elimination of 3' OH Groups of mRNA Using Alkaline Hydrolysis In a total volume of 100 μl of 0 1 N sodium hydroxide, 1 5 μg mRNA is incubated for 40 to 60 minutes at 4°C The solution is neutralized with acetic acid and precipitated with ethanol Following the optional elimination of the 3' OH groups, the diol groups at the 5' ends of the mRNAs are oxidized as described below in Example 9
EXAMPLE 9
Oxidation of Diols of mRNA Up to 1 OD unit of RNA was dissolved in 9 μl of buffer (0 1 M sodium acetate, pH
6-7) or water and 3 μl of freshly prepared 0 1 M sodium periodate solution The reaction was incubated for 1 h in the dark at 4°C or room temperature Following the incubation, the reaction was stopped by adding 4 μl of 10% ethylene glycol Thereafter the mixture was incubated at room temperature for 15 minutes After ethanol precipitation, the product was resuspended in at least 10 μl of water or appropriate buffer and dialyzed against water Following oxidation of the diol groups at the 5' ends of the mRNAs, the derivatized oligonucleotide was joined to the resulting aldehydes as described in Example 10.
EXAMPLE 10 Ligature of Aldehydes of mRNA to Derivatized Oligonucleotides
The oxidized mRNA was dissolved in an acidic medium such as 50 μl of sodium acetate pH 4-6. Fifty μl of a solution of the derivatized oligonucleotide were added in order to obtain an mRNA: derivatized oligonucleotide ratio of 1:20. The mixture was reduced with a borohydride and incubated for 2 h at 37°C or overnight (14 h) at 10°C. The mixture was then ethanol precipitated, resuspended in 10 μl or more of water or appropriate buffer and dialyzed against distilled water. If desired, the resulting product may be analyzed using acrylamide gel electrophoresis, HPLC analysis, or other conventional techniques.
Following the attachment of the derivatized oligonucleotide to the mRNAs, a reverse transcription reaction may be performed as described in Example 11 below.
EXAMPLE 11
Reverse Transcription of mRNAs Ligatured to Derivatized Oligonucleotides An oligodeoxyribonucleotide was derivatized as follows. Three OD units of an oligodeoxyribonucleotide of sequence 5ΑTCAAGAATTCGCACGAGACCATTA3' (SEQ ED NO:3) having 5'-OH and 3'-P ends were dissolved in 70 μl of a 1.5 M hydroxybenzotriazole solution, pH 5.3, prepared in dimethylformamide/water (75:25) containing 2 μg of l-ethyl-3-(3-dimethylanτinopropyl)carbodiimide. The mixture was incubated for 2 h 30 min at 22°C and then precipitated twice in LiClθ4/acetone. The pellet was resuspended in 200 μl of 0.25 M hydrazine and incubated at 8°C from 3 to 14 h. Following the hydrazine reaction, the mixture was precipitated twice in LiClO acetone.
The messenger RNAs to be reverse transcribed were extracted from blocks of placenta having sides of 2 cm which had been stored at -80°C. The total RNA was extracted using conventional acidic phenol techniques. Oligo-dT chromatography was used to purify the mRNAs. The integrity of the mRNAs was checked by Northern-blotting. The diol groups on 7 μg of the placental mRNAs were oxidized as described above in Example 9. The derivatized oligonucleotide was joined to the mRNAs as described in Example 10 above except that the precipitation step was replaced by an exclusion chromatography step to remove derivatized oligodeoxyribonucleotides which were not joined to mRNAs. Exclusion chromatography was performed as follows:
Ten ml of Ultrogel AcA34 (BioSepra#230151) gel, a mix of agarose and acrylamide, were equilibrated in 50 ml of a solution of 10 mM Tris pH 8.0, 300 mM NaCl, 1 mM EDTA, and 0.05% SDS. The mixture was allowed to sediment. The supernatant was eliminated and the gel was resuspended in 50 ml of buffer. This procedure was repeated 2 or 3 times. A glass bead (diameter 3 mm) was introduced into a 2 ml disposable pipette (length
25 cm). The pipette was filled with the gel suspension until the height of the gel stabilized at 1 cm from the top of the pipette. The column was then equilibrated with 20 ml of equilibration buffer (10 mM Tris HCl pH 7.4, 20 mM NaCl).
Ten μl of the mRNA which had reacted with the derivatized oligonucleotide were mixed in 39 μl of 10 mM urea and 2 μl of blue-glycerol buffer, which had been prepared by dissolving 5 mg of bromophenol blue in 60% glycerol (v/v), and passing the mixture through a 0.45 μm diameter filter.
The column was then loaded with the mRNAs coupled to the oligonucleotide. As soon as the sample had penetrated, equilibration buffer was added. Hundred μl fractions were then collected. Derivatized oligonucleotide which had not been attached to mRNA appeared in fraction 16 and later fractions. Thus, fractions 3 to 15 were combined and precipitated with ethanol.
To determine whether the derivatized oligonucleotide was actually linked to mRNA, one tenth of the combined fractions were spotted twice on a nylon membrane and hybridized to a radioactive probe using conventional techniques. The 32P labeled probe used in these hybridizations was an oligodeoxyribonucleotide of sequence
5'TAATGGTCTCGTGCGAATTCTTGAT3' (SEQ ID NO:4) anticomplementary to the derivatized oligonucleotide. A signal observed after autoradiography, indicated that the derivatized oligonucleotide had been truly joined to the mRNA. The remaining nine tenth of the mRNAs which had reacted with the derivatized oligonucleotide was reverse transcribed as follows. A reverse transcription reaction was carried out with reverse transcriptase following the manufacturer's instructions and 50 pmol of nonamers with random sequence as primers.
To ensure that reverse transcription had been carried out through the cap structure, two types of experiments were performed. In the first approach, after eumination of RNA of the cDNA:RNA heteroduplexes obtained from the reverse transcription reaction by an alkaline hydrolysis, a portion of the resulting single stranded cDNAs was spotted on a positively charged membrane and hybridized, using conventional methods, to a 32P labeled probe having a sequence identical to that of the derivatized oligonucleotide. Control spots containing, 1 pmol, 100 finol, 50 frnol, 10 frnol and 1 frnol of a control oligodeoxyribonucleotide of sequence identical to that of the derivatized oligonucleotide were included. The signal observed in the spots containing the cDNA indicated that approximately 15 frnol of the derivatized oligonucleotide had been reverse transcribed. These results demonstrate that the reverse transcription can be performed through the cap and, in particular, that reverse transcriptase crosses the 5'-P-P-P- 5' bond of the cap of eukaryotic messenger RNAs.
In the second type of experiment, the single stranded cDNAs obtained from the above first strand synthesis were used as template for PCR reactions. Two types of reactions were carried out. First, specific amplification of the mRNAs for alpha globin, dehydrogenase, ppl5 and elongation factor E4 were carried out using the following pairs of oligodeoxyribonucleotide primers.
alpha-globin GLO-S: 5'CCG ACA AGA CCA ACG TCA AGG CCG C3' (SEQ ED NO:5) GLO-As: 5'TCA CCA GCA GGC AGT GGC TTA GGA G 3' (SEQ ED NO:6)
dehydrogenase 3 DH-S: 5'AGT GAT TCC TGC TAC TTT GGA TGG C3' (SEQ LD NO:7) 3 DH-As: 5'GCT TGG TCT TGT TCT GGA GTT TAG A3' (SEQ ED NO:8)
ppl5 PP15-S: 5'TCC AGA ATG GGA GAC AAG CCA ATT T3' (SEQ ED NO:9) PP15-As:-5'AGG GAG GAG GAA ACA GCG TGA GTC C3' (SEQ ID NO: 10)
Elongation factor E4 EFA1-S: 5'ATG GGA AAG GAA AAG ACT CAT ATC A3' (SEQ ID NO:l 1) EFIA-As: 5'AGC AGC AAC AAT CAG GAC AGC ACA G3' (SEQ ID NO.T2)
Second, non specific amplifications were also carried out with the antisense oligodeoxyribonucleotides of the pairs described above and with a primer derived from the sequence of the derivatized oligodeoxyribonucleotide (5' ATCAAGAATTCGCACGAGACCATTA3 ') (SEQ ED NO: 13).
One twentieth of the following RT-PCR product samples were run on a 1.5% agarose gel and stained with ethidium bromide.
Sample 1 : The products of a PCR reaction using the globin primers of SEQ ID NOs 5 and 6 in the presence of cDNA Sample 2: The products of a PCR reaction using the globin primers of SEQ ED NOs
5 and 6 in the absence of added cDNA.
Sample 3: The products of a PCR reaction using the dehydrogenase primers of SEQ ED NOs 7 and 8 in the presence of cDNA.
Sample 4: The products of a PCR reaction using the dehydrogenase primers of SEQ ID NOs 7 and 8 in the absence of added cDNA.
Sample 5: The products of a PCR reaction using the ppl5 primers of SEQ ED NOs 9 and 10 in the presence of cDNA.
Sample 6: The products of a PCR reaction using the ppl5 primers of SEQ ID NOs 9 and 10 in the absence of added cDNA. Sample 7: The products of a PCR reaction using the EEF4 primers of SEQ ID NOs
11 and 12 in the presence of added cDNA.
Sample 8: The products of a PCR reaction using the EEF4 primers of SEQ ID NOs 11 and 12 in the absence of added cDNA.
A band of the size expected for the PCR product was observed only in samples 1, 3, 5 and 7, thus indicating the presence of the corresponding sequence in the cDNA population. PCR reactions were also carried out with the antisense oligonucleotides of the globin and dehydrogenase primers (SEQ ED NOs 6 and 8) and an oligonucleotide whose sequence corresponds to that of the derivatized oligonucleotide. The presence of PCR products of the expected size in the samples equivalent to above samples 1 and 3 indicated that the derivatized oligonucleotide had been linked to mRNA.
The above examples summarize the chemical procedure for enriching mRNAs for those having intact 5' ends as illustrated in Figure 1. Further detail regarding the chemical approaches for obtaining such mRNAs are disclosed in International Application No. W096/34981, published November 7, 1996, which is incoφorated herein by reference. Strategies based on the above chemical modifications to the 5' cap structure may be utilized to generate cDNAs selected to include the 5' ends of the mRNAs from which they derived. In one version of such procedures, the 5' ends of the mRNAs are modified as described above. Thereafter, a reverse transcription reaction is conducted to extend a primer complementary to the 5' end of the mRNA. Single stranded RNAs are eliminated to obtain a population of cDNA/mRNA heteroduplexes in which the mRNA includes an intact 5' end. The resulting heteroduplexes may be captured on a solid phase coated with a molecule capable of interacting with the molecule used to derivatize the 5' end of the mRNA. Thereafter, the strands of the heteroduplexes are separated to recover single stranded first cDNA strands which include the 5' end of the mRNA. Second strand cDNA synthesis may then proceed using conventional techniques. For example, the procedures disclosed in WO 96/34981 or in Caminci. et al., Genomics 37:327-336, 1996, the disclosures of which are incoφorated herein by reference, may be employed to select cDNAs which include the sequence derived from the 5' end of the coding sequence of the mRNA. Following ligation of the oligonucleotide tag to the 5' cap of the mRNA, a reverse transcription reaction is conducted to extend a primer complementary to the mRNA to the 5' end of the mRNA. Following elimination of the RNA component of the resulting heteroduplex using standard techniques, second strand cDNA synthesis is conducted with a primer complementary to the oligonucleotide tag. 2. Enzymatic Methods for Obtaining mRNAs having Intact 5' Ends
Other techniques for selecting cDNAs extending to the 5' end of the mRNA from which they are derived are fully enzymatic. Some versions of these techniques are disclosed in Dumas Milne Edwards J.B. (Doctoral Thesis of Paris VI University, Le clonage des ADNc complets: difficultes et perspectives nouvelles. Apports pour l'etude de la regulation de l'expression de la tryptophane hydroxylase de rat, 20 Dec. 1993), EPO 625572 and Kato et al, Gene 150:243-250, 1994, the disclosures of which are incoφorated herein by reference.
Briefly, in such approaches, isolated mRNA is treated with alkaline phosphatase to remove the phosphate groups present on the 5' ends of uncapped incomplete mRNAs. Following this procedure, the cap present on full length mRNAs is enzymatically removed with a decapping enzyme such as T4 polynucleotide kinase or tobacco acid pyrophosphatase.
An oligonucleotide, which may be either a DNA oligonucleotide or a DNA-RNA hybrid oligonucleotide having RNA at its 3' end, is then ligated to the phosphate present at the 5' end of the decapped mRNA using T4 RNA ligase. The oligonucleotide may include a restriction site to facilitate cloning of the cDNAs following their synthesis. Example 12 below describes one enzymatic method based on the doctoral thesis of Dumas.
EXAMPLE 12
Enzymatic Approach for Obtaining 5' ESTs Twenty micrograms of PolyA+ RNA were dephosphorylated using Calf Intestinal
Phosphatase (Biolabs). After a phenol chloroform extraction, the cap structure of mRNA was hydrolysed using the Tobacco Acid Pyrophosphatase (purified as described by Shinshi et al.., Biochemistry 15: 2185-2190, 1976) and a hemi 5ONA/RNA-3' oligonucleotide having an unphosphorylated 5' end, a stretch of adenosine ribophosphate at the 3' end, and an EcoRl site near the 5' end was ligated to the 5'P ends of mRNA using the T4 RNA ligase (Biolabs). Oligonucleotides suitable for use in this procedure are preferably 30 to 50 bases in length. Oligonucleotides having an unphosphorylated 5' end may be synthesized by adding a fluorochrome at the 5' end. The inclusion of a stretch of adenosine ribophosphates at the 3' end of the oligonucleotide increases ligation efficiency. It will be appreciated that the oligonucleotide may contain cloning sites other than EcoRl. Following ligation of the oligonucleotide to the phosphate present at the 5' end of the decapped mRNA, first and second strand cDNA synthesis is carried out using conventional methods or those specified in EPO 625,572 and Kato et al. supra, and Dumas Milne Edwards, supra, the disclosures of which are incoφorated herein by reference. The resulting cDNA may then be ligated into vectors such as those disclosed in Kato et al., supra or other nucleic acid vectors known to those skilled in the art using techniques such as those described in Sambrook et al., Molecular Cloning: A Laboratory Manual 2d Ed., Cold Spring Harbor Laboratory Press, 1989, the disclosure of which is incoφorated herein by reference.
LT. Obtention and Characterization of the 5' ESTs of the Present Invention
The 5' ESTs of the present invention were obtained using the aforementioned chemical and enzymatic approaches for enriching mRNAs for those having intact 5' ends as decribed below.
1. Obtention of 5* ESTS Using mRNAs with Intact 5' Ends
First, mRNAs were prepared as described in Example 13 below.
EXAMPLE 13 Preparation of mRNA With Intact 5' Ends
Total human RNAs or polyA+ RNAs derived from 29 different tissues were respectively purchased from LABIMO and CLONTECH and used to generate 44 cDNA libraries as follows. The purchased RNA had been isolated from cells or tissues using acid guanidium thiocyanate-phenol-chloroform extraction (Chomczyniski and Sacchi, Analytical Biochemistry 162:156-159, 1987). PolyA+ RNA was isolated from total RNA (LABIMO) by two passes of oligo dT chromatography, as described by Aviv and Leder, Proc. Natl.
Acad Sci. USA 69:1408-1412, 1972 in order to eliminate ribosomal RNA.
The quality and the integrity of the polyA+ RNAs were checked. Northern blots hybridized with a globin probe were used to confirm that the mRNAs were not degraded. Contamination of the polyA" mRNAs by ribosomal sequences was checked using Northern blots and a probe derived from the sequence of the 28 S rRNA. Preparations of mRNAs with less than 5% of rRNAs were used in library construction. To avoid constructing libraries with RNAs contaminated by exogenous sequences (prokaryotic or fungal), the presence of bacterial 16S ribosomal sequences or of two highly expressed fungal mRNAs was examined using PCR. Following preparation of the mRNAs, the above described chemical and/or the enzymatic procedures for enriching mRNAs for thoses having intact 5' ends were employed to obtain 5' ESTs from various tissues. In both approaches, an oligonucleotide tag was attached to the 5' ends of the mRNAs. The oligonucleotide tag had an EcoRl site therein to facilitate later cloning procedures. To facilitate the processing of single stranded and double stranded cDNA obtained in the construction of the librairies, the same nucleotidic sequence was used to design the ligated oligonucleotide in both chemical and enzymatic approaches. Nevertheless, in the chemical procedure, the tag used was an oligodeoxyribonucleotide which was linked to the cap of the mRNA whereas in the enzymatic ligation, the tag was a chimeric hemi 5'DNA/RNA3' oligonucleotide which was ligated to the 5' end of decapped mRNA as described in example 12.
Following attachment of the oligonucleotide tag to the mRNA by either the chemical or enzymatic methods, the integrity of the mRNA was examined by performing a Northern blot with 200 to 500 ng of mRNA using a probe complementary to the oligonucleotide tag before performing the first strand synthesis as described in example 14.
EXAMPLE 14 cDNA Synthesis Using mRNA Templates Having Intact 5' Ends For the mRNAs joined to oligonucleotide tags using both the chemical and enzymatic methods, first strand cDNA synthesis was performed using the Superscript II (Gibco BRL) or the Rnase H Minus M-MLV (Promega) reverse transcriptase with random nonamers as primers. In order to protect internal EcoRl sites in the cDNA from digestion at later steps in the procedure, methylated dCTP was used for first strand synthesis. After removal of RNA by an alkaline hydrolysis, the first strand of cDNA was precipitated using isopropanol in order to eliminate residual primers. For both the chemical and the enzymatic methods, the second strand of the cDNA was synthesized with a Klenow fragment using a primer corresponding to the 5' end of the ligated oligonucleotide described in Example 12. Preferably, the primer is 20-25 bases in length. Methylated dCTP was also used for second strand synthesis in order to protect internal EcoRI sites in the cDNA from digestion during the cloning process.
Following cDNA synthesis, the cDNAs were cloned into pBlueScript as described in Example 15 below.
EXAMPLE 15
Cloning of cDNAsderived from mRNA with intact 5' ends into BlueScript
Following second strand synthesis, the ends of the cDNA were blunted with T4 DNA polymerase (Biolabs) and the cDNA was digested with EcoRI. Since methylated dCTP was used during cDNA synthesis, the EcoRI site present in the tag was the only hemi-methylated site, hence the only site susceptible to EcoRI digestion. The cDNA was then size fractionated using exclusion chromatography (Ac A, Biosepra) and fractions corresponding to cDNAs of more than 150 bp were pooled and ethanol precipitated. The cDNA was directionally cloned into the Smal and EcoRI ends of the phagemid pBlueScript vector (Stratagene). The ligation mixture was electroporated into bacteria and propagated under appropriate antibiotic selection.
Clones containing the oligonucleotide tag attached were then selected as described in Example 16 below.
EXAMPLE 16 Selection of Clones Having the Oligonucleotide Tag Attached Thereto The plasmid DNAs containing 5' EST libraries made as described above were purified (Qiagen). A positive selection of the tagged clones was performed as follows. Briefly, in this selection procedure, the plasmid DNA was converted to single stranded DNA using gene II endonuclease of the phage FI in combination with an exonuclease (Chang et al, Gene 127:95-8, 1993) such as exonuclease HI or T7 gene 6 exonuclease. The resulting single stranded DNA was then purified using paramagnetic beads as described by Fry et al, Biotechniques, 13: 124-131, 1992. In this procedure, the single stranded DNA was hybridized with a biotinylated oligonucleotide having a sequence corresponding to the 3' end of the oligonucleotide described in Example 13. Preferably, the primer has a length of 20-25 bases. Clones including a sequence complementary to the biotinylated oligonucleotide were captured by incubation with streptavidin coated magnetic beads followed by magnetic selection. After capture of the positive clones, the plasmid DNA was released from the magnetic beads and converted into double stranded DNA using a DNA polymerase such as the ThermoSequenase obtained from Amersham Pharmacia Biotech. Alternatively, protocoles such as the one described in the Gene Trapper kit available from Gibco BRL may be used. The double stranded DNA was then electroporated into bacteria. The percentage of positive clones having the 5' tag oligonucleotide was estimated to typically rank between 90 and 98%o using dot blot analysis. Following electroporation, the libraries were ordered in 384-microtiter plates (MTP).
A copy of the MTP was stored for future needs. Then the libraries were transferred into 96
MTP and sequenced as described below.
EXAMPLE 17 Sequencing of Inserts in Selected Clones
Plasmid inserts were first amplified by PCR on PE 9600 thermocyclers (Perkin-Elmer, Applied Biosystems Division, Foster City, CA), using standard SETA-A and SETA-B primers (Genset SA), AmpliTaqGold (Perkin-Elmer), dNTPs (Boehringer), buffer and cycling conditions as recommended by the Perkin-Elmer Coφoration. PCR products were then sequenced using automatic ABI Prism 377 sequencers
(Perkin Elmer). Sequencing reactions were performed using PE 9600 thermocyclers with standard dye-primer chemistry and ThermoSequenase (Amersham Pharmacia Biotech). The primers used were either T7 or 21M13 (available from Genset SA) as appropriate. The primers were labeled with the JOE, FAM, ROX and TAMRA dyes. The dNTPs and ddNTPs used in the sequencing reactions were purchased from Boehringer. Sequencing buffer, reagent concentrations and cycling conditions were as recommended by Amersham.
Following the sequencing reaction, the samples were precipitated with ethanol, resuspended in formamide loading buffer, and loaded on a standard 4% acrylamide gel. Electrophoresis was performed for 2.5 hours at 3000V on an ABI 377 sequencer, and the sequence data were collected and analyzed using the ABI Prism DNA Sequencing Analysis Software, version 2.1.2. 2. Computer analysis of the Obtained 5' ESTs: Construction of NetGene and SignalTag databases
The sequence data from the 44 cDNA libraries made as described above were transferred to a proprietary database, where quality control and validation steps were performed. A proprietary base-caller, working using a Unix system, automatically flagged suspect peaks, taking into account the shape of the peaks, the inter-peak resolution, and the noise level. The proprietary base-caller also performed an automatic trimming. Any stretch of 25 or fewer bases having more than 4 suspect peaks was considered unreliable and was discarded. Sequences corresponding to cloning vector or ligation oligonucleotides were automatically removed from the EST sequences. However, the resulting EST sequences may contain 1 to 5 bases belonging to the above mentioned sequences at their 5' end. If needed, these can easily be removed on a case to case basis.
Following sequencing as described above, the sequences of the 5' ESTs were entered in NetGene™, a proprietary database called for storage and manipulation as described below. It will be appreciated by those skilled in the art that the data could be stored and manipulated on any medium which can be read and accessed by a computer. Computer readable media include magnetically, optically, or electronically readable media. For example, the computer readable media may be a hard disc, a floppy disc, a magnetic tape, CD-ROM RAM, or ROM as well as other types of other media known to those skilled in the art. In addition, the sequence data may be stored and manipulated in a variety of data processor programs in a diversity of formats. For instance, the sequence data may be stored as text in a word processing file, such as Microsoft WORD or WORDPERFECT or as an ASCII file in a variety of database programs familiar to those of skill in the art, such as DB2, SYBASE, or ORACLE. The computer readable media on which the sequence information is stored may be in a personal computer, a network, a server or other computer systems known to those skilled in the art. The computer or other system preferably includes the storage media described above, and a processor for accessing and manipulating the sequence data. Once the sequence data has been stored, it may be manipulated and searched to locate those stored sequences which contain a desired nucleic acid sequence or which encode a protein having a particular functional domain. For example, the stored sequence information may be compared to other known sequences to identify homologies, motifs implicated in biological function, or structural motifs.
Programs which may be used to search or compare the stored sequences include the
MacPattern (EMBL), BLAST, and BLAST2 program series (NCBI), basic local alignment search tool programs for nucleotide (BLASTN) and peptide (BLASTX) comparisons
(Altschul et al, J. Mol. Biol. 215: 403, 1990) and FASTA (Pearson and Lipman, Proc. Natl.
Acad. Sci. USA 85: 2444, 1988). The BLAST programs then extend the alignments on the basis of defined match and mismatch criteria.
Motifs which may be detected using the above programs and those described in Example 28 include sequences encoding leucine zippers, helix-turn-helix motifs, glycosylation sites, ubiquitination sites, alpha helices, and beta sheets, signal sequences encoding signal peptides which direct the secretion of the encoded proteins, sequences implicated in transcription regulation such as homeoboxes, acidic stretches, enzymatic active sites, substrate binding sites, and enzymatic cleavage sites. Before searching the cDNAs in the NetGene™ database for sequence motifs of interest, cDNAs derived from mRNAs which were not of interest were identified and eliminated from further consideration as described in Example 18 below.
EXAMPLE 18 Elimination of Undesired Sequences from Further Consideration
5' ESTs in the NetGene™ database which were derived from undesired sequences such as transfer RNAs, ribosomal RNAs, mitochondrial RNAs, prokaryotic RNAs, fungal RNAs, Alu sequences, Ll sequences, .or repeat sequences were identified using the FASTA and BLASTN programs with the parameters listed in Table I. To eliminate 5' ESTs encoding tRNAs from further consideration, the 5' EST sequences were compared to the sequences of 1190 known tRNAs obtained from EMBL release 38, of which 100 were human. The comparison was performed using FASTA on both strands of the 5' ESTs. Sequences having more than 80% homology over more than 60 nucleotides were identified as tRNA. Of the 144,341 sequences screened, 26 were identified as tRNAs and eliminated from further consideration. To eliminate 5' ESTs encoding rRNAs from further consideration, the 5' EST sequences were compared to the sequences of 2497 known rRNAs obtained from EMBL release 38, of which 73 were human. The comparison was performed using BLASTN on both strands of the 5' ESTs with the parameter S=108. Sequences having more than 80% homology over stretches longer than 40 nucleotides were identified as rRNAs. Of the 144,341 sequences screened, 3,312 were identified as rRNAs and eliminated from further consideration.
To eliminate 5' ESTs encoding mtRNAs from further consideration, the 5' EST sequences were compared to the sequences of the two known mitochondrial genomes for which the entire genomic sequences are available and all sequences transcribed from these mitochondrial genomes including tRNAs, rRNAs, and mRNAs for a total of 38 sequences. The comparison was performed using BLASTN on both strands of the 5' ESTs with the parameter S=108. Sequences having more than 80% homology over stretches longer than 40 nucleotides were identified as mtRNAs. Of the 144,341 sequences screened, 6,110 were identified as mtRNAs and eliminated from further consideration.
Sequences which might have resulted from exogenous contaminants were eliminated from further consideration by comparing the 5' EST sequences to release 46 of the EMBL bacterial and fungal divisions using BLASTN with the parameter S=144. All sequences having more than 90% homology over at least 40 nucleotides were identified as exogenous contaminants. Of the 42 cDNA libraries examined, the average percentages of prokaryotic and fungal sequences contained therein were 0.2% and 0.5% respectively. Among these sequences, only one could be identified as a sequence specific to fungi. The others were either fungal or prokaryotic sequences having homologies with vertebrate sequences or including repeat sequences which had not been masked during the electronic comparison. In addition, the 5' ESTs were compared to 6093 Alu sequences and 1115 Ll sequences to mask 5' ESTs containing such repeat sequences. 5' ESTs including THE and MER repeats, SSTR sequences or satellite, micro-satellite, or telomeric repeats were also eliminated from further consideration. On average, 11.5% of the sequences in the libraries contained repeat sequences. Of this 11.5%, 7% contained Alu repeats, 3.3% contained Ll repeats and the remaining 1.2% were derived from the other screened types of repetitive sequences. These percentages are consistent with those found in cDNA libraries prepared by other groups. For example, the cDNA libraries of Adams et al. contained between 0% and 7.4% Alu repeats depending on the source of the RNA which was used to prepare the cDNA library (Adams et al, Nature 377:174, 1996).
The sequences of those 5' ESTs remaining after the elimination of undesirable sequences were compared with the sequences of known human mRNAs to determine the accuracy of the sequencing procedures described above.
EXAMPLE 19 Measurement of Sequencing Accuracy by Comparison to Known Sequences
To further determine the accuracy of the sequencing procedure described above, the sequences of 5' ESTs derived from known sequences were identified and compared to the original known sequences. First, a FASTA analysis with overhangs shorter than 5 bp on both ends was conducted on the 5' ESTs to identify those matching an entry in the public human mRNA database. The 6655 5' ESTs which matched a known human mRNA were then realigned with their cognate mRNA and dynamic programming was used to include substitutions, insertions, and deletions in the list of "errors" which would be recognized. Errors occurring in the last 10 bases of the 5' EST sequences were ignored to avoid the inclusion of spurious cloning sites in the analysis of sequencing accuracy. This analysis revealed that the sequences incoφorated in the NetGene™ database had an accuracy of more than 99.5%.
To determine the efficiency with which the above selection procedures select cDNAs which include the 5' ends of their corresponding mRNAs, the following analysis was performed.
EXAMPLE 20
Determination of Efficiency of 5' EST Selection To determine the efficiency at which the above selection procedures isolated 5' ESTs which included sequences close to the 5' end of the mRNAs from which they derived, the sequences of the ends of the 5' ESTs derived from the elongation factor 1 subunit α and ferritin heavy chain genes were compared to the known cDNA sequences of these genes. Since the transcription start sites of both genes are well characterized, they may be used to determine the percentage of derived 5' ESTs which included the authentic transcription start sites. For both genes, more than 95% of the obtained 5' ESTs actually included sequences close to or upstream of the 5' end of the corresponding mRNAs.
To extend the analysis of the reliability of the procedures for isolating 5' ESTs from ESTs in the NetGene™ database, a similar analysis was conducted using a database composed of human mRNA sequences extracted from GenBank database release 97 for comparison. The 5' ends of more than 85% of 5' ESTs derived from mRNAs included in the GeneBank database were located close to the 5' ends of the known sequence. As some of the mRNA sequences available in the GenBank database are deduced from genomic sequences, a 5' end matching with these sequences will be counted as an internal match. Thus, the method used here underestimates the yield of ESTs including the authentic 5' ends of their corresponding mRNAs.
The EST libraries made above included multiple 5' ESTs derived from the same mRNA. The sequences of such 5' ESTs were compared to one another and the longest 5'
ESTs for each mRNA were identified. Overlapping cDNAs were assembled into continuous sequences (contigs). The resulting continuous sequences were then compared to public databases to gauge their similarity to known sequences, as described in Example 21 below.
EXAMPLE 21
Clustering of the 5' ESTs and Calculation of Novelty Indices for cDNA Libraries For each sequenced EST library, the sequences were clustered by the 5' end. Each sequence in the library was compared to the others with BLASTN2 (direct strand, parameters S=107). ESTs with High Scoring Segment Pairs (HSPs) at least 25 bp long, having 95% identical bases and beginning closer than 10 bp from each EST 5' end were grouped. The longest sequence found in the cluster was used as representative of the group. A global clustering between libraries was then performed leading to the definition of super-contigs. To assess the yield of new sequences within the EST libraries, a novelty rate (NR) was defined as: NR= 100 X (Number of new unique sequences found in the library/Total number of sequences from the library). Typically, novelty rating ranged between 10%) and 41%) depending on the tissue from which the EST library was obtained. For most of the libraries, the random sequencing of 5' EST libraries was pursued until the novelty rate reached 20%.
Following characterization as described above, the collection of 5' ESTs in NetGene™ was screened to identify those 5' ESTs bearing potential signal sequences as described in Example 22 below.
EXAMPLE 22
Identification of Potential Signal Sequences in 5' ESTs The 5' ESTs in the NetGene™ database were screened to identify those having an uninterrupted open reading frame (ORF) longer than 45 nucleotides beginning with an ATG codon and extending to the end of the EST. Approximately half of the cDNA sequences in NetGene™ contained such an ORF. The ORFs of these 5' ESTs were then searched to identify potential signal motifs using slight modifications of the procedures disclosed in Von Heijne, Nucleic Acids Res. 14:4683-4690, 1986, the disclosure of which is incoφorated herein by reference. Those 5' EST sequences encoding a stretch of at least 15 amino acid long with a score of at least 3.5 in the Von Heijne signal peptide identification matrix were considered to possess a signal sequence. Those 5' ESTs which matched a known human mRNA or EST sequence and had a 5' end more than 20 nucleotides downstream of the known 5' end were excluded from further analysis. The remaining cDNAs having signal sequences therein were included in a database called SignalTag™.
To confirm the accuracy of the above method for identifying signal sequences, the analysis of Example 23 was performed. EXAMPLE 23
Confirmation of Accuracy of Identification of Potential Signal Sequences in 5' ESTs The accuracy of the above procedure for identifying signal sequences encoding signal peptides was evaluated by applying the method to the 43 amino acids located at the N tcπninus of all human SwissProt proteins. The computed Von Heijne score for each protein was compared with the known characterization of the protein as being a secreted protein or a non-secreted protein. In this manner, the number of non-secreted proteins having a score higher than 3.5 (false positives) and the number of secreted proteins having a score lower than 3.5 (false negatives) could be calculated. Using the results of the above analysis, the probability that a peptide encoded by the
5' region of the mRNA is in fact a genuine signal peptide based on its Von Heijne's score was calculated based on either the assumption that 10% of human proteins are secreted or the assumption that 20% of human proteins are secreted. The results of this analysis are shown in Figure 2 and table IV. Using the above method of identification of secretory proteins, 5' ESTs of the following polypeptides known to be secreted were obtained: human glucagon, gamma interferon induced monokine precursor, secreted cyclophilin-like protein, human pleiotropin, and human biotinidase precursor. Thus, the above method successfully identified those 5' ESTs which encode a signal peptide.
To confirm that the signal peptide encoded by the 5' ESTs actually functions as a signal peptide, the signal sequences from the 5' ESTs may be cloned into a vector designed for the identification of signal peptides. Such vectors are designed to confer the ability to grow in selective medium only to host cells containing a vector with an operably linked signal sequence. For example, to confirm that a 5' EST encodes a genuine signal peptide, the signal sequence of the 5' EST may be inserted upstream and in frame with a non-secreted form of the yeast invertase gene in signal peptide selection vectors such as those described in U.S. Patent No. 5,536,637, the disclosure of which is incoφorated herein by reference. Growth of host cells containing signal sequence selection vectors with the correctly inserted 5' EST signal sequence confirms that the 5' EST encodes a genuine signal peptide. Alternatively, the presence of a signal peptide may be confirmed by cloning the extended cDNAs obtained using the ESTs into expression vectors such as pXTl (as described below in example 30), or by constructing promoter-signal sequence-reporter gene vectors which encode fusion proteins between the signal peptide and an assayable reporter protein. After introduction of these vectors into a suitable host cell, such as COS cells or NLH 3T3 cells, the growth medium may be harvested and analyzed for the presence of the secreted protein. The medium from these cells is compared to the medium from control cells containing vectors lacking the signal sequence or extended cDNA insert to identify vectors which encode a functional signal peptide or an authentic secreted protein. Those 5' ESTs which encoded a signal peptide, as determined by the method of
Example 22 above, were further grouped into four categories based on their homology to known sequences as described in Example 24 below.
EXAMPLE 24 Categorization of 5' ESTs Encoding a Signal Peptide
Those 5' ESTs having a sequence not matching any known vertebrate sequence nor any publicly available EST sequence were designated "new." Of the sequences in the SignalTag™ database, 947 of the 5' ESTs having a Von Heijne's score of at least 3.5 fell into this category. Those 5' ESTs having a sequence not matching any vertebrate sequence but matching a publicly known EST were designated "EST-ext", provided that the known EST sequence was extended by at least 40 nucleotides in the 5' direction. Of the sequences in the SignalTag™ database, 150 of the 5' ESTs having a Von Heijne's score of at least 3.5 fell into this category. Those ESTs not matching any vertebrate sequence but matching a publicly known
EST without extending the known EST by at least 40 nucleotides in the 5' direction were designated "EST." Of the sequences in the SignalTag™ database, 599 of the 5' ESTs having a Von Heijne's score of at least 3.5 fell into this category.
Those 5' ESTs matching a human mRNA sequence but extending the known sequence by at least 40 nucleotides in the 5' direction were designated "VERT-ext." Of the sequences in the SignalTag™ database, 23 of the 5' ESTs having a Von Heijne's score of at least 3.5 fell into this category. Included in this category was a 5' EST which extended the known sequence of the human translocase mRNA by more than 200 bases in the 5' direction. A 5' EST which extended the sequence of a human tumor suppressor gene in the 5' direction was also identified. Table V shows the distribution of 5' ESTs in each category and the number of 5'
ESTs in each category having a given minimum von Heijne's score.
3. Evaluation of Spatial and Temporal Expression of mRNAs Corresponding to the SΕSTs or Extended cDNAs
Each of the 5' ESTs was also categorized based on the tissue from which its corresponding mRNA was obtained, as described below in Example 25.
EXAMPLE 25 Categorization of Expression Patterns
Table VI shows the distribution of 5' ESTs in each of the above defined category with respect to the tissue from which the 5'ESTs of the corresponding mRNA were obtained. Table II provides the sequence identification numbers of 5' EST sequences derived from muscle and other mesodermal tissues, the categories in which these sequences fall, and the von Heijne's score of the signal peptides which they encode. The 5' EST sequences and the amino acid sequences they encode are provided in the appended sequence listings. Table III provides the sequence ED numbers of the 5' ESTs and the sequences of the signal peptides which they encode. The sequences of the 5' ESTs and the polypeptides they encode are provided in the sequence listing appended hereto. The sequences of DNA SEQ ID NOs: 38-305 can readily be screened for any errors therein and any sequence ambiguities can be resolved by resequencing a fragment containing such errors or amibiguities on both strands. Such fragments may be obtained from the plasmids stored in the inventors' laboratory or can be isolated using the techniques described herein. Resolution of any such ambiguities or errors may be facilitated by using primers which hybridize to sequences located close to the ambiguous or erroneous sequences. For example, the primers may hybridize to sequences within 50-75 bases of the amibiguity or error. Upon resolution of an error or ambiguity, the corresponding corrections can be made in the protein sequences encoded by the DNA containing the error or amibiguity.
In addition to categorizing the 5' ESTs with respect to their tissue of origin, the spatial and temporal expression patterns of the mRNAs corresponding to the 5' ESTs, as well as their expression levels, may be determined as described in Example 26 below. Characterization of the spatial and temporal expression patterns and expression levels of these mRNAs is useful for constructing expression vectors capable of producing a desired level of gene product in a desired spatial or temporal manner, as will be discussed in more detail below. Furthermore, 5' ESTs whose corresponding mRNAs are associated with disease states may also be identified. For example, a particular disease may result from the lack of expression, over expression, or under expression of an mRNA corresponding to a 5' EST. By comparing mRNA expression patterns and quantities in samples taken from healthy individuals with those from individuals suffering from a particular disease, 5' ESTs responsible for the disease may be identified.
It will be appreciated that the results of the above characterization procedures for 5'
ESTs also apply to extended cDNAs (obtainable as described below) which contain sequences adjacent to the 5' ESTs. It will also be appreciated that if desired, characterization may be delayed until extended cDNAs have been obtained rather than characterizing the
ESTs themselves.
EXAMPLE 26
Evaluation of Expression Levels and Patterns of mRNAs Corresponding to 5' ESTs or Extended cDNAs
Expression levels and patterns of mRNAs corresponding to 5' ESTs or extended cDNAs (obtainable as described below in example 27) may be analyzed by solution hybridization with long probes as described in International Patent Application No. WO 97/05277, the entire contents of which are hereby incoφorated by reference. Briefly, a 5' EST, extended cDNA, or fragment thereof corresponding to the gene encoding the mRNA to be characterized is inserted at a cloning site immediately downstream of a bacteriophage (T3, T7 or SP6) RNA polymerase promoter to produce antisense RNA. Preferably, the 5' EST or extended cDNA has 100 or more nucleotides. The plasmid is linearized and transcribed in the presence of ribonucleotides comprising modified ribonucleotides (i.e. biotin-UTP and DIG- UTP). An excess of this doubly labeled RNA is hybridized in solution with mRNA isolated from cells or tissues of interest. The hybridizations are performed under standard stringent conditions (40-50°C for 16 hours in an 80% formamide, 0.4 M NaCl buffer, pH 7-8). The unhybridized probe is removed by digestion with ribonucleases specific for single-stranded RNA (i.e. RNases CL3, Tl, Phy M U2 or A). The presence of the biotin-UTP modification enables capture of the hybrid on a microtitration plate coated with streptavidin. The presence of the DIG modification enables the hybrid to be detected and quantified by ELISA using an anti-DIG antibody coupled to alkaline phosphatase.
The 5' ESTs, extended cDNAs, or fragments thereof may also be tagged with nucleotide sequences for the serial analysis of gene expression (SAGE) as disclosed in UK Patent Application No. 2 305 241 A, the entire contents of which are incoφorated by reference. In this method, cDNAs are prepared from a cell, tissue, organism or other source of nucleic acid for which gene expression patterns must be determined. The resulting cDNAs are separated into two pools. The cDNAs in each pool are cleaved with a first restriction endonuclease, called an anchoring enzyme, having a recognition site which is likely to be present at least once in most cDNAs. The fragments which contain the 5' or 3' most region of the cleaved cDNA are isolated by binding to a capture medium such as streptavidin coated beads. A first oligonucleotide linker having a first sequence for hybridization of an amplification primer and an internal restriction site for a so-called tagging endonuclease is ligated to the digested cDNAs in the first pool. Digestion with the second endonuclease produces short tag fragments from the cDNAs. A second oligonucleotide having a second sequence for hybridization of an amplification primer and an internal restriction site is ligated to the digested cDNAs in the second pool. The cDNA fragments in the second pool are also digested with the tagging endonuclease to generate short tag fragments derived from the cDNAs in the second pool. The tags resulting from digestion of the first and second pools with the anchoring enzyme and the tagging endonuclease are ligated to one another to produce so-called ditags. In some embodiments, the ditags are concatamerized to produce ligation products containing from 2 to 200 ditags. The tag sequences are then determined and compared to the sequences of the 5' ESTs or extended cDNAs to determine which 5' ESTs or extended cDNAs are expressed in the cell, tissue, organism, or other source of nucleic acids from which the tags were derived. In this way, the expression pattern of the 5' ESTs or extended cDNAs in the cell, tissue, organism, or other source of nucleic acids is obtained.
Quantitative analysis of gene expression may also be performed using arrays. As used herein, the term array means a one dimensional, two dimensional, or multidimensional arrangement of full length cDNAs (i.e. extended cDNAs which include the coding sequence for the signal peptide, the coding sequence for the mature protein, and a stop codon), extended cDNAs, 5' ESTs or fragments thereof of sufficient length to permit specific detection of gene expression. Preferably, the fragments are at least 15 nucleotides in length. More preferably, the fragments are at least 100 nucleotide long. More preferably, the fragments are more than 100 nucleotides in length. In some embodiments, the fragments may be more than 500 nucleotide long. For example, quantitative analysis of gene expression may be performed with full length cDNAs as defined below, extended cDNAs, 5' ESTs, or fragments thereof in a complementary DNA microarray as described by Schena et al. (Science 270:467-470, 1995; Proc. Natl. Acad Sci. U.S.A. 93:10614-10619, 1996). Full length cDNAs, extended cDNAs, 5' ESTs or fragments thereof are amplified by PCR and arrayed from 96-well microtiter plates onto sOylated microscope slides using high-speed robotics. Printed arrays are incubated in a humid chamber to allow rehydration of the array elements and rinsed, once in 0.2% SDS for 1 min, twice in water for 1 min and once for 5 min in sodium borohydride solution. The arrays are submerged in water for 2 min at 95°C, transferred into 0.2% SDS for 1 min, rinsed twice with water, air dried and stored in the dark at 25°C. Cell or tissue mRNA is isolated or commercially obtained and probes are prepared by a single round of reverse transcription. Probes are hybridized to 1 cm2 icroarrays under a 14 x 14 mm glass coverslip for 6-12 hours at 60°C. Arrays are washed for 5 min at 25°C in low stringency wash buffer (1 x SSC/0.2%> SDS), then for 10 min at room temperature in high stringency wash buffer (0.1 x SSC/0.2% SDS). Arrays are scanned in 0.1 x SSC using a fluorescence laser scarining device fitted with a custom filter set. Accurate differential expression measurements are obtained by taking the average of the ratios of two independent hybridizations.
Quantitative analysis of the expression of genes may also be performed with full length cDNAs, extended cDNAs, 5' ESTs, or fragments thereof in complementary DNA arrays as described by Pietu et al. (Genome Research 6:492-503, 1996). The full length cDNAs, extended cDNAs, 5' ESTs or fragments thereof are PCR amplified and spotted on membranes. Then, mRNAs originating from various tissues or cells are labeled with radioactive nucleotides. After hybridization and washing in controlled conditions, the hybridized mRNAs are detected by phospho-imaging or autoradiography. Duplicate experiments are performed and a quantitative analysis of differentially expressed mRNAs is then performed.
Alternatively, expression analysis of the 5' ESTs or extended cDNAs can be done through high density nucleotide arrays as described by Lockhart et al. (Nature Biotechnology 14: 1675-1680, 1996) and Sosnowsky et al (Proc. Natl. Acad Sci. 94:1119-1123, 1997). Oligonucleotides of 15-50 nucleotides corresponding to sequences of the 5' ESTs or extended cDNAs are synthesized directly on the chip (Lockhart et al, supra) or synthesized and then addressed to the chip (Sosnowsky et al, supra). Preferably, the oligonucleotides are about 20 nucleotides in length. cDNA probes labeled with an appropriate compound, such as biotin, digoxigenin or fluorescent dye, are synthesized from the appropriate mRNA population and then randomly fragmented to an average size of 50 to 100 nucleotides. The said probes are then hybridized to the chip. After washing as described in Lockhart et al, supra and application of different electric fields (Sonowsky et al, supra , the dyes or labeling compounds are detected and quantified. Duplicate hybridizations are performed. Comparative analysis of the intensity of the signal originating from cDNA probes on the same target oligonucleotide in different cDNA samples indicates a differential expression of the mRNA corresponding to the 5' EST or extended cDNA from which the oligonucleotide sequence has been designed. ELL Use of 5' ESTs to Clone Extended cDNAs and to Clone the Corresponding Genomic DNAs
Once 5' ESTs which include the 5' end of the corresponding mRNAs have been selected using the procedures described above, they can be utilized to isolate extended cDNAs which contain sequences adjacent to the 5' ESTs. The extended cDNAs may include the entire coding sequence of the protein encoded by the corresponding mRNA, including the authentic translation start site, the signal sequence, and the sequence encoding the mature protein remaining after cleavage of the signal peptide. Such extended cDNAs are referred to herein as "full length cDNAs." Alternatively, the extended cDNAs may include only the sequence encoding the mature protein remaining after cleavage of the signal peptide, or only the sequence encoding the signal peptide.
Example 27 below describes a general method for obtaining extended cDNAs using 5' ESTs. Example 28 below provides experimental results, using the method explained in example 27, describing several extended cDNAs including the entire coding sequence and authentic 5' end of the corresponding mRNA for several secreted proteins.
The methods of Examples 27, 28, and 29 can also be used to obtain extended cDNAs which encode less than the entire coding sequence of the secreted proteins encoded by the genes corresponding to the 5' ESTs. In some embodiments, the extended cDNAs isolated using these methods encode at least 10 amino acids of one of the proteins encoded by the sequences of SEQ ID NOs: 38-305. Ln further embodiments, the extended cDNAs encode at least 20 amino acids of the proteins encoded by the sequences of SEQ ED NOs: 38-305. In further embodiments, the extended cDNAs encode at least 30 amino amino acids of the sequences of SEQ ID NOs: 38-305. In a preferred embodiment, the extended cDNAs encode a full length protein sequence, which includes the protein coding sequences of SEQ ED NOs: 38-305.
EXAMPLE 27
General Method for Using 5' ESTs to Clone and Sequence cDNAs which Include the Entire Coding Region and the Authentic 5' End of the Corresponding mRNA The following general method has been used to quickly and efficiently isolate extended cDNAs having the authentic 5' ends of their corresponding mRNAs as well as the full protein coding sequence and including sequence adjacent to the sequences of the 5' ESTs used to obtain them. This method may be applied to obtain extended cDNAs for any 5' EST in the NetGene™ database, including those 5' ESTs encoding polypeptides belonging to secreted proteins. The method is summarized in figure 3.
1 . Obtention of Extended cDNAs a) First strand synthesis
The method takes advantage of the known 5' sequence of the mRNA. A reverse transcription reaction is conducted on purified mRNA with a poly 14dT primer containing a 49 nucleotide sequence at its 5' end allowing the addition of a known sequence at the end of the cDNA which corresponds to the 3' end of the mRNA. For example, the primer may have the following sequence: 5'-ATC GTT GAG ACT CGT ACC AGC AGA GTC ACG AGA GAG ACT ACA CGG TAC TGG TTT TTT TTT TTT TTVN -3' (SEQ ID NO: 14). Those skilled in the art will appreciate that other sequences may also be added to the poly dT sequence and used to prime the first strand synthesis. Using this primer and a reverse transcriptase such as the Superscript II (Gibco BRL) or Rnase H Minus M-MLV (Promega) enzyme, a reverse transcript anchored at the 3' polyA site of the RNAs is generated.
After removal of the mRNA hybridized to the first cDNA strand by alkaline hydrolysis, the products of the alkaline hydrolysis and the residual poly dT primer are eliminated with an exclusion column such as an AcA34 (Biosepra) matrix as explained in Example 1 1. b) Second strand synthesis
A pair of nested primers on each end is designed based on the known 5' sequence from the 5' EST and the known 3' end added by the poly dT primer used in the first strand synthesis. Softwares used to design primers are either based on GC content and melting temperatures of oligonucleotides, such as OSP (Illier and Green, PCR Melh. Appl. 1:124- 128, 1991), or based on the octamer frequency disparity method (Griffais et al., Nucleic Acids Res. 19: 3887-3891, 1991) such as PC-Rare
(http:/^ioirιformatics.weizmann.ac.il/software/PC-Rjare/doo'manuel.htrnl). Preferably, the nested primers at the 5' end are separated from one another by four to nine bases. The 5' primer sequences may be selected to have melting temperatures and specificities suitable for use in PCR.
Preferably, the nested primers at the 3' end are separated from one another by four to nine bases. For example, the nested 3' primers may have the following sequences: (5'- CCA GCA GAG TCA CGA GAG AGA CTA CAC GG -3 '(SEQ ID NO: 15), and 5'- CAC GAG AGA GAC TAC ACG GTA CTG G -3' (SEQ ID NO: 16). These primers were selected because they have melting temperatures and specificities compatible with their use in PCR. However, those skilled in the art will appreciate that other sequences may also be used as primers.
The first PCR run of 25 cycles is performed using the Advantage Tth Polymerase Mix (Clontech) and the outer primer from each of the nested pairs. A second 20 cycle PCR using the same enzyme and the inner primer from each of the nested pairs is then performed on 1/2500 of the first PCR product. Thereafter, the primers and nucleotides are removed.
2. Sequencing of Full Length Extended cDNAs or Fragments Thereof
Due to the lack of position constraints on the design of 5' nested primers compatible for PCR use using the OSP software, amplicons of two types are obtained. Preferably, the second 5' primer is located upstream of the translation initiation codon thus yielding a nested PCR product containing the whole coding sequence. Such a full length extended cDNA undergoes a direct cloning procedure as described in section a.
However, in some cases, the second 5' primer is located downstream of the translation initiation codon, thereby yielding a PCR product containing only part of the ORF. Such incomplete PCR products are submitted to a modified procedure described in section b. a) Nested PCR products containing complete ORFs
When the resulting nested PCR product contains the complete coding sequence, as predicted from the 5'EST sequence, it is cloned in an appropriate vector such as pED6dpc2, as described in section 3. b) Nested PCR products containing incomplete ORFs When the amplicon does not contain the complete coding sequence, intermediate steps are necessary to obtain both the complete coding sequence and a PCR product containing the full coding sequence. The complete coding sequence can be assembled from several partial sequences determined directly from different PCR products as described in the following section.
Once the full coding sequence has been completely determined, new primers compatible for PCR use are designed to obtain amplicons containing the whole coding region. However, in such cases, 3' primers compatible for PCR use are located inside the 3' UTR of the corresponding mRNA, thus yielding amplicons which lack part of this region, i.e. the polyA tract and sometimes the polyadenylation signal, as illustrated in figure 3. Such full length extended cDNAs are then cloned into an appropriate vector as described in section 3. c) Sequencing extended cDNAs
Sequencing of extended cDNAs is performed using a Die Terminator approach with the AmpliTaq DNA polymerase FS kit available from Perkin Elmer.
In order to sequence PCR fragments, primer walking is performed using software such as OSP to choose primers and automated computer software such as ASMG (Sutton et al., Genome Science Technol. 1: 9-19, 1995) to construct contigs of walking sequences including the initial 5' tag using minimum overlaps of 32 nucleotides. Preferably, primer walking is performed until the sequences of full length cDNAs are obtained.
Completion of the sequencing of a given extended cDNA fragment is assessed as follows. Since sequences located after a polyA tract are difficult to determine precisely in the case of uncloned products, sequencing and primer walking processes for PCR products are interrupted when a polyA tract is identified in extended cDNAs obtained as described in case b. The sequence length is compared to the size of the nested PCR product obtained as described above. Due to the limited accuracy of the determination of the PCR product size by gel electrophoresis, a sequence is considered complete if the size of the obtained sequence is at least 70 % the size of the first nested PCR product. If the length of the sequence determined from the computer analysis is not at least 70% of the length of the nested PCR product, these PCR products are cloned and the sequence of the insertion is determined. When Northern blot data are available, the size of the mRNA detected for a given PCR product is used to finally assess that the sequence is complete. Sequences which do not fulfill the above criteria are discarded and will undergo a new isolation procedure.
Sequence data of all extended cDNAs are then transferred to a proprietary database, where quality controls and validation steps are carried out as described in example 15.
3. Cloning of Full Length Extended cDNAs
The PCR product containing the full coding sequence is then cloned in an appropriate vector. For example, the extended cDNAs can be cloned into the expression vector pED6dpc2 (DiscoverEase, Genetics Institute, Cambridge, MA) as follows. pED6dpc2 vector DNA is prepared with blunt ends by performing an EcoRI digestion followed by a fill in reaction. The blunt ended vector is dephosphorylated. After removal of PCR primers and ethanol precipitation, the PCR product containing the full coding sequence or the extended cDNA obtained as described above is phosphorylated with a kinase subsequently removed by phenol-Sevag extraction and precipitation. The double stranded extended cDNA is then ligated to the vector and the resulting expression plasmid introduced into appropriate host cells.
Since the PCR products obtained as described above are blunt ended molecules that can be cloned in either direction, the orientation of several clones for each PCR product is determined. Then, 4 to 10 clones are ordered in microtiter plates and subjected to a PCR reaction using a first primer located in the vector close to the cloning site and a second primer located in the portion of the extended cDNA corresponding to the 3' end of the mRNA. This second primer may be the antisense primer used in anchored PCR in the case of direct cloning (case a) or the antisense primer located inside the 3 'UTR in the case of indirect cloning (case b). Clones in which the start codon of the extended cDNA is operably linked to the promoter in the vector so as to permit expression of the protein encoded by the extended cDNA are conserved and sequenced. In addition to the ends of cDNA inserts, approximately 50 bp of vector DNA on each side of the cDNA insert are also sequenced.
The cloned PCR products are then entirely sequenced according to the aforementioned procedure. In this case, contigation of long fragments is then performed on walking sequences that have already contigated for uncloned PCR products during primer walking. Sequencing of cloned amplicons is complete when the resulting contigs include the whole coding region as well as overlapping sequences with vector DNA on both ends.
4. Computer analysis of Full Length Extended cDNA
Sequences of all full length extended cDNAs are then submitted to further analysis as described below. Before searching the extended full length cDNAs for sequences of interest, extended cDNAs which are not of interest (vector RNAs, transfer RNAs, ribosomal RNAs, mitochondrial RNAs, prokaryotic RNAs and fungal RNAs) are discarded using methods essentially similar to those described for 5 'ESTs in Example 18. a) Identification of structural features
Structural features, e.g. polyA tail and polyadenylation signal, of the sequences of full length extended cDNAs are subsequently determined as follows.
A polyA tail is defined as a homopolymeric stretch of at least 11 A with at most one alternative base within it. The polyA tail search is restricted to the last 100 nt of the sequence and limited to stretches of 11 consecutive As because sequencing reactions are often not readable after such a polyA stretch. Stretches having more than 90% homology over 8 nucleotides are identified as polyA tails using BLAST2N.
To search for a polyadenylation signal, the polyA tail is clipped from the full- length sequence. The 50 bp preceding the polyA tail are first searched for the canonic polyadenylation AAUAAA signal and, if the canonic signal is not detected, for the alternative AUUAAA signal (Sheets et al, Nuc. Acids Res. 18: 5799-5805, 1990). If neither of these consensus polyadenylation signals is found, the canonic motif is searched again allowing one mismatch to account for possible sequencing errors. More than 85 %> of identified polyadenylation signals of either type actually ends 10 to 30 bp from the polyA tail. Alternative AUUAAA signals represents approximately 15 % of the total number of identified polyadenylation signals. b) Identification of functional features
Functional features, e.g. ORFs and signal sequences, of the sequences of full length extended cDNAs were subsequently determined as follows. The 3 upper strand frames of extended cDNAs are searched for ORFs defined as the maximum length fragments beginning with a translation intiation codon and ending with a stop codon. ORFs encoding at least 20 amino acids are preferred.
Each found ORF is then scanned for the presence of a signal peptide in the first 50 amino-acids or, where appropriate, within shorter regions down to 20 amino acids or less in the ORF, using the matrix method of von Heijne (Niic. Acids Res. 14: 4683-4690, 1986), the disclosure of which is incoφorated herein by reference as described in Example 22. c) Homology to either nucleotidic orproteic sequences
Categorization of full-length sequences may be achieved using procedures essentially similar to those described for 5 'ESTs in Example 24.
Extended cDNAs prepared as described above may be subsequently engineered to obtain nucleic acids which include desired portions of the extended cDNA using conventional techniques such as subcloning, PCR, or in vitro oligonucleotide synthesis. For example, nucleic acids which include only the full coding sequences (i.e. the sequences encoding the signal peptide and the mature protein remaining after the signal peptide is cleaved oft) may be obtained using techniques known to those skilled in the art. Alternatively, conventional techniques may be applied to obtain nucleic acids which contain only the coding sequences for the mature protein remaining after the signal peptide is cleaved off or nucleic acids which contain only the coding sequences for the signal peptides.
Similarly, nucleic acids containing any other desired portion of the coding sequences for the secreted protein may be obtained. For example, the nucleic acid may contain at least 10 consecutive bases of an extended cDNA such as one of the extended cDNAs described below. In another embodiment, the nucleic acid may contain at least 15 consecutive bases of an extended cDNA such as one of the extended cDNAs described below. Alternatively, the nucleic acid may contain at least 20 consecutive bases of an extended cDNA such as one of the extended cDNAs described below. In another embodiment, the nucleic acid may contain at least 25 consecutive bases of an extended cDNA such as one of the extended cDNAs described below. In yet another embodiment, the nucleic acid may contain at least 40 consecutive bases of an extended cDNA such as one of the extended cDNAs described below. Once an extended cDNA has been obtained, it can be sequenced to determine the amino acid sequence it encodes. Once the encoded amino acid sequence has been determined, one can create and identify any of the many conceivable cDNAs that will encode that protein by simply using the degeneracy of the genetic code. For example, allelic variants or other homologous nucleic acids can be identified as described below. Alternatively, nucleic acids encoding the desired amino acid sequence can be synthesized in vitro.
In a preferred embodiment, the coding sequence may be selected using the known codon or codon pair preferences for the host organism in which the cDNA is to be expressed.
The extended cDNAs derived from the 5' ESTS of the present invention were obtained as described in Example 28 below.
EXAMPLE 28
Characterization of cloned extended cDNAs obtained using 5' ESTs The procedure described in Example 27 above was used to obtain the extended cDNAs derived from the 5' ESTs of the present invention in a variety of tissues. The following list provides a few examples of thus obtained extended cDNAs.
Using this approach, the full length cDNA of SEQ ID NO: 17 (internal identification number 48-19-3-G1-FL1) was obtained. This cDNA falls into the "EST-ext" category described above and encodes the signal peptide MKKVLLLITAELAVAVG (SEQ ED NO: 18) having a von Heijne score of 8.2.
The full length cDNA of SEQ ID NO: 19 (internal identification number 58-34-2-E7- FL2) was also obtained using this procedure. This cDNA falls into the "EST-ext" category described above and encodes the signal peptide MWWFQQGLSFLPSALVIWTSA (SEQ ID NO:20) having a von Heijne score of 5.5. Another f ll length cDNA obtained using the procedure described above has the sequence of SEQ ED NO:21 (internal identification number 51-27-1-E8-FL1). This cDNA, falls into the "EST-ext" category described above and encodes the signal peptide MVLTTLPSANSANSPVNMPTTGPNSLSYASSALSPCLT (SEQ ED NO:22) having a von Heijne score of 5.9. The above procedure was also used to obtain a full length cDNA having the sequence of SEQ ED NO:23 (internal identification number 76-4-1-G5-FL1). This cDNA falls into the "EST-ext" category described above and encodes the signal peptide ILSTVTALTFAXA (SEQ ID NO:24) having a von Heijne score of 5.5.
The full length cDNA of SEQ ID NO:25 (internal identification number 51-3-3-B10- FL3) was also obtained using this procedure. This cDNA falls into the "new" category described above and encodes a signal peptide LVLTLCTLPLAVA (SEQ ED NO: 26) having a von Heijne score of 10.1.
The full length cDNA of SEQ ED NO:27 (internal identification number 58-35-2-F10- FL2) was also obtained using this procedure. This cDNA falls into the "new" category described above and encodes a signal peptide LWLLFFLVTALHA (SEQ ED NO:28) having a von Heijne score of 10.7.
Bacterial clones containing plasmids containing the full length cDNAs described above are presently stored in the inventor's laboratories under the internal identification numbers provided above. The inserts may be recovered from the stored materials by growing an aliquot of the appropriate bacterial clone in the appropriate medium. The plasmid DNA can then be isolated using plasmid isolation procedures familiar to those skilled in the art such as alkaline lysis minipreps or large scale alkaline lysis plasmid isolation procedures. If desired the plasmid DNA may be further enriched by centrifugation on a cesium chloride gradient, size exclusion chromatography, or anion exchange chromatography. The plasmid DNA obtained using these procedures may then be manipulated using standard cloning techniques familiar to those skilled in the art. Alternatively, a PCR can be done with primers designed at both ends of the cDNA insertion. The PCR product which corresponds to the cDNA can then be manipulated using standard cloning techniques familiar to those skilled in the art.
The polypeptides encoded by the extended cDNAs may be screened for the presence of known structural or functional motifs or for the presence of signatures, small amino acid sequences which are well conserved amongst the members of a protein family. The conserved regions have been used to derive consensus patterns or matrices included in the PROSITE data bank, in particular in the file prosite.dat (Release 13.0 of November 1995, located at http://expasy.hcuge.ch/sprot/prosite.html. Prosite_convert and prosite_scan programs (http://ulrec3.unil.ch/ftpserveur/prosite_scan) may be used to find signatures on the extended cDNAs. For each pattern obtained with the prosite_convert program from the prosite.dat file, the accuracy of the detection on a new protein sequence may be assessed by evaluating the frequency of irrelevant hits on the population of human secreted proteins included in the data bank SWISSPROT. The ratio between the number of hits on shuffled proteins (with a window size of 20 amino acids) and the number of hits on native (unshuffled) proteins may be used as an index. Every pattern for which the ratio is greater than 20% (one hit on shuffled proteins for 5 hits on native proteins) may be skipped during the search with prosite_scan. The program used to shuffle protein sequences (db_shuffled) and the program used to determine the statistics for each pattern in the protein data banks (prosite_statistics) are available on the ftp site http://ulrec3.unil.ch/ftpserveur/prosite_scan.
In addition to PCR based methods for obtaining extended cDNAs, traditional hybridization based methods may also be employed. These methods may also be used to obtain the genomic DNAs which encode the mRNAs from which the 5' ESTs were derived, mRNAs corresponding to the extended cDNAs, or nucleic acids which are homologous to extended cDNAs or 5' ESTs. Example 29 below provides examples of such methods.
EXAMPLE 29
Methods for Obtaining cDNAs which include the Entire Coding Region and the Authentic 5 'End of the Corresponding mRNA
A full length cDNA library can be made using the strategies described in Examples 13, 14, 15, and 16 above by replacing the random nonamer used in Example 14 with an oligo- dT primer. For instance, the oligonucleotide of SEQ ED NO: 14 may be used.
Alternatively, a cDNA library or genomic DNA library may be obtained from a commercial source or made using techniques familiar to those skilled in the art. Such cDNA or genomic DNA librairies may be used to isolate extended cDNAs obtained from 5' EST or nucleic acids homologous to extended cDNAs or 5' EST as follows. The cDNA library or genomic DNA library is hybridized to a detectable probe comprising at least 10 consecutive nucleotides from the 5' EST or extended cDNA using conventional techniques. Preferably, the probe comprises at least 12, 15, or 17 consecutive nucleotides from the 5' EST or extended cDNA. More preferably, the probe comprises at least 20 to 30 consecutive nucleotides from the 5' EST or extended cDNA. In some embodiments, the probe comprises more than 30 nucleotides from the 5 ' EST or extended cDNA.
Techniques for identifying cDNA clones in a cDNA library which hybridize to a given probe sequence are disclosed in Sambrook et al, Molecular Cloning: A Laboratory Manual 2d Ed., Cold Spring Harbor Laboratory Press, 1989, the disclosure of which is incoφorated herein by reference. The same techniques may be used to isolate genomic DNAs.
Briefly, cDNA or genomic DNA clones which hybridize to the detectable probe are identified and isolated for further manipulation as follows. A probe comprising at least 10 consecutive nucleotides from the 5' EST or extended cDNA is labeled with a detectable label such as a radioisotope or a fluorescent molecule. Preferably, the probe comprises at least 12, 15, or 17 consecutive nucleotides from the 5' EST or extended cDNA. More preferably, the probe comprises 20 to 30 consecutive nucleotides from the 5' EST or extended cDNA. In some embodiments, the probe comprises more than 30 nucleotides from the 5' EST or extended cDNA. Techniques for labeling the probe are well known and include phosphorylation with polynucleotide kinase, nick translation, in vitro transcription, and non radioactive techniques. The cDNAs or genomic DNAs in the library are transferred to a nitrocellulose or nylon filter and denatured. After blocking of non specific sites, the filter is incubated with the labeled probe for an amount of time sufficient to allow binding of the probe to cDNAs or genomic DNAs containing a sequence capable of hybridizing thereto.
By varying the stringency of the hybridization conditions used to identify extended cDNAs or genomic DNAs which hybridize to the detectable probe, extended cDNAS having different levels of homology to the probe can be identified and isolated as described below.
1. Identification of Extended cDNA or Genomic cDNA Sequences Having a High Degree of Homology to the Labeled Probe
To identify extended cDNAs or genomic DNAs having a high degree of homology to the probe sequence, the melting temperature of the probe may be calculated using the following formulas: For probes between 14 and 70 nucleotides in length the melting temperature (Tm) is calculated using the formula: Tm=81.5+16.6(log [Na+])+0.41 (fraction G+C)-(600/N) where N is the length of the probe.
Lf the hybridization is carried out in a solution containing formamide, the melting temperature may be calculated using the equation Tm=81.5+16.6(log [Na+])+0.41 (fraction G+C)-(0.63% formamide)-(600/N) where N is the length of the probe.
Prehybridization may be carried out in 6X SSC, 5X Denhardt's reagent, 0.5% SDS, 100 μg denatured fragmented salmon sperm DNA or 6X SSC, 5X Denhardt's reagent, 0.5% SDS, 100 μg denatured fragmented salmon sperm DNA, 50%> formamide. The formulas for SSC and Denhardt's solutions are listed in Sambrook et al. , supra.
Hybridization is conducted by adding the detectable probe to the prehybridization solutions listed above. Where the probe comprises double stranded DNA, it is denatured before addition to the hybridization solution. The filter is contacted with the hybridization solution for a sufficient period of time to allow the probe to hybridize to extended cDNAs or genomic DNAs containing sequences complementary thereto or homologous thereto. For probes over 200 nucleotides in length, the hybridization may be carried out at 15-25°C below the Tm. For shorter probes, such as oligonucleotide probes, the hybridization may be conducted at 15-25°C below the Tm. Preferably, for hybridizations in 6X SSC, the hybridization is conducted at approximately 68°C. Preferably, for hybridizations in 50% formamide containing solutions, the hybridization is conducted at approximately 42°C.
All of the foregoing hybridizations would be considered to be under "stringent" conditions.
Following hybridization, the filter is washed in 2X SSC, 0.1% SDS at room temperature for 15 minutes. The filter is then washed with 0.1X SSC, 0.5% SDS at room temperature for 30 minutes to 1 hour. Thereafter, the solution is washed at the hybridization temperature in 0.1X SSC, 0.5% SDS. A final wash is conducted in 0.1X SSC at room temperature.
Extended cDNAs, nucleic acids homologous to extended cDNAs or 5' ESTs, or genomic DNAs which have hybridized to the probe are identified by autoradiography or other conventional techniques. 2. Obtention of Extended cDNA or Genomic cDNA Sequences Having Lower Degrees of Homology to the Labeled Probe
The above procedure may be modified to identify extended cDNAs, nucleic acids homologous to extended cDNAs, or genomic DNAs having decreasing levels of homology to the probe sequence. For example, to obtain extended cDNAs, nucleic acids homologous to extended cDNAs, or genomic DNAs of decreasing homology to the detectable probe, less stringent conditions may be used. For example, the hybridization temperature may be decreased in increments of 5°C from 68°C to 42°C in a hybridization buffer having a sodium concentration of approximately IM. Following hybridization, the filter may be washed with 2X SSC, 0.5% SDS at the temperature of hybridization. These conditions are considered to be "moderate" conditions above 50°C and "low" conditions below 50°C.
Alternatively, the hybridization may be carried out in buffers, such as 6X SSC, containing formamide at a temperature of 42°C. In this case, the concentration of formamide in the hybridization buffer may be reduced in 5% increments from 50% to 0%o to identify clones having decreasing levels of homology to the probe. Following hybridization, the filter may be washed with 6X SSC, 0.5% SDS at 50°C. These conditions are considered to be "moderate" conditions above 25% formamide and "low" conditions below 25%> formamide.
Extended cDNAs, nucleic acids homologous to extended cDNAs, or genomic DNAs which have hybridized to the probe are identified by autoradiography.
3. Determination of the Degree of Homology Between the Obtained Extended cDNAs and the Labeled Probe
If it is desired to obtain nucleic acids homologous to extended cDNAs, such as allelic variants thereof or nucleic acids encoding proteins related to the proteins encoded by the extended cDNAs, the level of homology between the hybridized nucleic acid and the extended cDNA or 5' EST used as the probe may be further determined using BLAST2N; parameters may be adapted depending on the sequence length and degree of homology studied. To determine the level of homology between the hybridized nucleic acid and the extended cDNA or 5'EST from which the probe was derived, the nucleotide sequences of the hybridized nucleic acid and the extended cDNA or 5'EST from which the probe was derived are compared. For example, using the above methods, nucleic acids having at least 95%> nucleic acid homology to the extended cDNA or 5'EST from which the probe was derived may be obtained and identified. Similarly, by using progressively less stringent hybridization conditions one can obtain and identify nucleic acids having at least 90%, at least 85%, at least 80%) or at least 75% homology to the extended cDNA or 5'EST from which the probe was derived.
To determine whether a clone encodes a protein having a given amount of homology to the protein encoded by the extended cDNA or 5' EST, the amino acid sequence encoded by the extended cDNA or 5' EST is compared to the amino acid sequence encoded by the hybridizing nucleic acid. Homology is determined to exist when an amino acid sequence in the extended cDNA or 5' EST is closely related to an amino acid sequence in the hybridizing nucleic acid. A sequence is closely related when it is identical to that of the extended cDNA or 5' EST or when it contains one or more amino acid substitutions therein in which amino acids having similar characteristics have been substituted for one another. Using the above methods and algorithms such as FASTA with parameters depending on the sequence length and degree of homology studied, one can obtain nucleic acids encoding proteins having at least 95%), at least 90%, at least 85%, at least 80% or at least 75% homology to the proteins encoded by the extended cDNA or 5'EST from which the probe was derived.
In addition to the above described methods, other protocols are available to obtain extended cDNAs using 5' ESTs as outlined in the following paragraphs.
Extended cDNAs may be prepared by obtaining mRNA from the tissue, cell, or organism of interest using mRNA preparation procedures utilizing polyA selection procedures or other techniques known to those skilled in the art. A first primer capable of hybridizing to the polyA tail of the mRNA is hybridized to the mRNA and a reverse transcription reaction is performed to generate a first cDNA strand.
The first cDNA strand is hybridized to a second primer containing at least 10 consecutive nucleotides of the sequences of SEQ ED NOs 38-305. Preferably, the primer comprises at least 12, 15, or 17 consecutive nucleotides from the sequences of SEQ ED NOs 38-305. More preferably, the primer comprises 20 to 30 consecutive nucleotides from the sequences of SEQ ED NOs 38-305. In some embodiments, the primer comprises more than 30 nucleotides from the sequences of SEQ ED NOs 38-305. If it is desired to obtain extended cDNAs containing the full protein coding sequence, including the authentic translation initiation site, the second primer used contains sequences located upstream of the translation initiation site. The second primer is extended to generate a second cDNA strand complementary to the first cDNA strand. Alternatively, RT-PCR may be performed as described above using primers from both ends of the cDNA to be obtained.
Extended cDNAs containing 5' fragments of the mRNA may be prepared by hybridizing an mRNA comprising the sequence of the 5'EST for which an extended cDNA is desired with a primer comprising at least 10 consecutive nucleotides of the sequences complementary to the 5'EST and reverse transcribing the hybridized primer to make a first cDNA strand from the mRNAs. Preferably, the primer comprises at least 12, 15, or 17 consecutive nucleotides from the 5'EST. More preferably, the primer comprises 20 to 30 consecutive nucleotides from the 5'EST.
Thereafter, a second cDNA strand complementary to the first cDNA strand is synthesized. The second cDNA strand may be made by hybridizing a primer complementary to sequences in the first cDNA strand to the first cDNA strand and extending the primer to generate the second cDNA strand.
The double stranded extended cDNAs made using the methods described above are isolated and cloned. The extended cDNAs may be cloned into vectors such as plasmids or viral vectors capable of replicating in an appropriate host cell. For example, the host cell may be a bacterial, mammalian, avian, or insect cell.
Techniques for isolating mRNA, reverse transcribing a primer hybridized to mRNA to generate a first cDNA strand, extending a primer to make a second cDNA strand complementary to the first cDNA strand, isolating the double stranded cDNA and cloning the double stranded cDNA are well known to those skilled in the art and are described in Current Protocols in Molecular Biology, John Wiley and Sons, Inc. 1997 and Sambrook et al, Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press, 1989, the entire disclosures of which are incoφorated herein by reference.
Alternatively, procedures such as the one described in Example 29 may be used for obtaining full length cDNAs or extended cDNAs. Ln this approach, full length or extended cDNAs are prepared from mRNA and cloned into double stranded phagemids as follows. The cDNA library in the double stranded phagemids is then rendered single stranded by treatment* with an endonuclease, such as the Gene El product of the phage FI, and an exonuclease (Chang et al, Gene 127:95-8, 1993). A biotinylated oligonucleotide comprising the sequence of a 5' EST, or a fragment containing at least 10 nucleotides thereof, is hybridized to the single stranded phagemids. Preferably, the fragment comprises at least 12, 15, or 17 consecutive nucleotides from the 5' EST. More preferably, the fragment comprises 20-30 consecutive nucleotides from the 5' EST. In some procedures, the fragment may comprise more than 30 consecutive nucleotides from the 5' EST.
Hybrids between the biotinylated oligonucleotide and phagemids having inserts containing the 5' EST sequence are isolated by incubating the hybrids with streptavidin coated paramagnetic beads and retrieving the beads with a magnet (Fry et al, Biotechniques, 13: 124-131, 1992). Therafter, the resulting phagemids containing the 5' EST sequence are released from the beads and converted into double stranded DNA using a primer specific for the 5' EST sequence. Alternatively, protocoles such as the Gene Trapper kit (Gibco BRL) may be used. The resulting double stranded DNA is transformed into bacteria. Extended cDNAs containing the 5' EST sequence are identified by colony PCR or colony hybridization.
Using any of the above described methods in section III, a plurality of extended cDNAs containing full length protein coding sequences or sequences encoding only the mature protein remaining after the signal peptide is cleaved off may be provided as cDNA libraries for subsequent evaluation of the encoded proteins or use in diagnostic assays as described below.
IV. Expression of Proteins Encoded by Extended cDNAs Isolated Using 5' ESTs
Extended cDNAs containing the full protein coding sequences of their corresponding mRNAs or portions thereof, such as cDNAs encoding the mature protein, may be used to express the encoded secreted proteins or portions thereof as described in Example 30 below. If desired, the extended cDNAs may contain the sequences encoding the signal peptide to facilitate secretion of the expressed protein. It will be appreciated that a plurality of extended cDNAs containing the full protein coding sequences or portions thereof may be simultaneously cloned into expression vectors to create an expression library for analysis of the encoded proteins as described below. EXAMPLE 30
Expression of the Proteins Encoded by the Genes Corresponding to 5ΕSTS or Portions Thereof To express the proteins encoded by the genes corresponding to 5' ESTs (or portions thereof), full length cDNAs containing the entire protein coding region or extended cDNAs containing sequences adjacent to the 5' ESTs (or portions thereof) are obtained as described in Examples 27-29 and cloned into a suitable expression vector. If desired, the nucleic acids may contain the sequences encoding the signal peptide to facilitate secretion of the expressed protein. The nucleic acids inserted into the expression vectors may also contain sequences upstream of the sequences encoding the signal peptide, such as sequences which regulate expression levels or sequences which confer tissue specific expression.
The nucleic acid encoding the protein or polypeptide to be expressed is operably linked to a promoter in an expression vector using conventional cloning technology. The expression vector may be any of the mammalian, yeast, insect or bacterial expression systems known in the art. Commercially available vectors and expression systems are available from a variety of suppliers including Genetics Institute (Cambridge, MA), Stratagene (La Jolla, California), Promega (Madison, Wisconsin), and Invitrogen (San Diego, California). If desired, to enhance expression and facilitate proper protein folding, the codon context and codon pairing of the sequence may be optimized for the particular expression organism in which the expression vector is introduced, as explained by Hatfield, et al, U.S. Patent No. 5,082,767, incoφorated herein by this reference.
The cDNA cloned into the expression vector may encode the entire protein (i.e. the signal peptide and the mature protein), the mature protein (i.e. the protein created by cleaving the signal peptide off), only the signal peptide or any other portion thereof. The following is provided as one exemplary method to express the proteins encoded by the extended cDNAs corresponding to the 5' ESTs or the nucleic acids described above. First, the methionine initiation codon for the gene and the polyA signal of the gene are identified. If the nucleic acid encoding the polypeptide to be expressed lacks a methionine to serve as the initiation site, an initiating methionine can be introduced next to the first codon of the nucleic acid using conventional techniques. Similarly, if the extended cDNA lacks a polyA signal, this sequence can be added to the construct by, for example, splicing out the polyA signal from pSG5 (Stratagene) using Bglll and Sail restriction endonuclease enzymes and incoφorating it into the mammalian expression vector pXTl (Stratagene). pXTl contains the LTRs and a portion of the gag gene from Moloney Murine Leukemia Virus. The position of the LTRs in the construct allow efficient stable transfection. The vector includes the Heφes Simplex thymidine kinase promoter and the selectable neomycin gene. The extended cDNA or portion thereof encoding the polypeptide to be expressed is obtained by PCR from the bacterial vector using oligonucleotide primers complementary to the extended cDNA or portion thereof and containing restriction endonuclease sequences for Pst I incoφorated into the 5'primer and Bglll at the 5' end of the corresponding cDNA 3' primer, taking care to ensure that the extended cDNA is positioned with the poly A signal. The purified fragment obtained from the resulting PCR reaction is digested with Pstl, blunt ended with an exonuclease, digested with Bgl II, purified and ligated to pXTl containing a poly A signal and prepared for this ligation (blunt/Bglll).
The ligated product is transfected into mouse NLH 3T3 cells using Lipofectin (Life Technologies, Inc., Grand Island, New York) under conditions outlined in the product specification. Positive transfectants are selected after growing the transfected cells in 600 μg/ml G418 (Sigma, St. Louis, Missouri). Preferably the expressed protein is released into the culture medium, thereby facilitating purification.
Alternatively, the extended cDNAs may be cloned into pED6dpc2 as described above. The resulting pED6dpc2 constructs may be transfected into a suitable host cell, such as COS 1 cells. Methotrexate resistant cells are selected and expanded. Preferably, the protein expressed from the extended cDNA is released into the culture medium thereby facilitating purification.
Proteins in the culture medium are separated by gel electrophoresis. If desired, the proteins may be ammonium sulfate precipitated or separated based on size or charge prior to electrophoresis.
As a control, the expression vector lacking a cDNA insert is introduced into host cells or organisms and the proteins in the medium are harvested. The secreted proteins present in the medium are detected using techniques familiar to those skilled in the art such as Coomassie blue or silver staining or using antibodies against the protein encoded by the extended cDNA. Antibodies capable of specifically recognizing the protein of interest may be generated using synthetic 15-mer peptides having a sequence encoded by the appropriate 5' EST, extended cDNA, or portion thereof. The synthetic peptides are injected into mice to generate antibody to the polypeptide encoded by the 5' EST, extended cDNA, or portion thereof. Secreted proteins from the host cells or organisms containing an expression vector which contains the extended cDNA derived from a 5' EST or a portion thereof are compared to those from the control cells or organism. The presence of a band in the medium from the cells containing the expression vector which is absent in the medium from the control cells indicates that the extended cDNA encodes a secreted protein. Generally, the band corresponding to the protein encoded by the extended cDNA will have a mobility near that expected based on the number of amino acids in the open reading frame of the extended cDNA. However, the band may have a mobility different than that expected as a result of modifications such as glycosylation, ubiquitination, or enzymatic cleavage.
Alternatively, if the protein expressed from the above expression vectors does not contain sequences directing its secretion, the proteins expressed from host cells containing an expression vector with an insert encoding a secreted protein or portion thereof can be compared to the proteins expressed in control host cells containing the expression vector without an insert. The presence of a band in samples from cells containing the expression vector with an insert which is absent in samples from cells containing the expression vector without an insert indicates that the desired protein or portion thereof is being expressed. Generally, the band will have the mobility expected for the secreted protein or portion thereof. However, the band may have a mobility different than that expected as a result of modifications such as glycosylation, ubiquitination, or enzymatic cleavage.
The protein encoded by the extended cDNA may be purified using standard immunochromatography techniques. In such procedures, a solution containing the secreted protein, such as the culture medium or a cell extract, is applied to a column having antibodies against the secreted protein attached to the chromatography matrix. The secreted protein is allowed to bind the immunochromatography column. Thereafter, the column is washed to remove non-specifically bound proteins. The specifically bound secreted protein is then released from the column and recovered using standard techniques. If antibody production is not possible, the extended cDNA sequence or portion thereof may be incoφorated into expression vectors designed for use in purification schemes employing chimeric polypeptides. In such strategies, the coding sequence of the extended cDNA or portion thereof is inserted in frame with the gene encoding the other half of the chimera. The other half of the chimera may be β-globin or a nickel binding polypeptide. A chromatography matrix having antibody to β-globin or nickel attached thereto is then used to purify the chimeric protein. Protease cleavage sites may be engineered between the β-globin gene or the nickel binding polypeptide and the extended cDNA or portion thereof Thus, the two polypeptides of the chimera may be separated from one another by protease digestion. One useful expression vector for generating β-globin chimerics is pSG5 (Stratagene), which encodes rabbit β-globin. Intron II of the rabbit β-globin gene facilitates splicing of the expressed transcript, and the polyadenylation signal incoφorated into the construct increases the level of expression. These techniques as described are well known to those skilled in the art of molecular biology. Standard methods are published in methods texts such as Davis et al.., (Basic Methods in Molecular Biology, Davis, Dibner, and Battey, ed., Elsevier Press, NY, 1986) and many of the methods are available from Stratagene, Life Technologies, Inc., or Promega. Polypeptide may additionally be produced from the construct using in vitro translation systems such as the In vitro Express™ Translation Kit (Stratagene).
Following expression and purification of the secreted proteins encoded by the 5'
ESTs, extended cDNAs, or fragments thereof, the purified proteins may be tested for the ability to bind to the surface of various cell types as described in Example 31 below. It will be appreciated that a plurality of proteins expressed from these cDNAs may be included in a panel of proteins to be simultaneously evaluated for the activities specifically described below, as well as other biological roles for which assays for determining activity are available.
EXAMPLE 31
Analysis of Secreted Proteins to Determine Whether they Bind to the Cell Surface The proteins encoded by the 5' ESTs, extended cDNAs, or fragments thereof are cloned into expression vectors such as those described in Example 30. The proteins are purified by size, charge, immunochromatography or other techniques familiar to those skilled in the art. Following purification, the proteins are labeled using techniques known to those skilled in the art. The labeled proteins are incubated with cells or cell lines derived from a variety of organs or tissues to allow the proteins to bind to any receptor present on the cell surface. Following the incubation, the cells are washed to remove non-specifically bound protein. The labeled proteins are detected by autoradiography. Alternatively, unlabeled proteins may be incubated with the cells and detected with antibodies having a detectable label, such as a fluorescent molecule, attached thereto.
Specificity of cell surface binding may be analyzed by conducting a competition analysis in which various amounts of unlabeled protein are incubated along with the labeled protein. The amount of labeled protein bound to the cell surface decreases as the amount of competitive unlabeled protein increases. As a control, various amounts of an unlabeled protein unrelated to the labeled protein is included in some binding reactions. The amount of labeled protein bound to the cell surface does not decrease in binding reactions containing increasing amounts of unrelated unlabeled protein, indicating that the protein encoded by the cDNA binds specifically to the cell surface.
As discussed above, secreted proteins have been shown to have a number of important physiological effects and, consequently, represent a valuable therapeutic resource. The secreted proteins encoded by the extended cDNAs or portions thereof made according to Examples 27-29 may be evaluated to determine their physiological activities as described below.
EXAMPLE 32
Assaying the Proteins Expressed from Extended cDNAs or Portions Thereof for Cytokine. Cell Proliferation or Cell Differentiation Activity
As discussed above, secreted proteins may act as cytokines or may affect cellular proliferation or differentiation. Many protein factors discovered to date, including all known cytokines, have exhibited activity in one or more factor dependent cell proliferation assays, and hence the assays serve as a convenient confirmation of cytokine activity. The activity of a protein encoded by the extended cDNAs is evidenced by any one of a number of routine factor dependent cell proliferation assays for cell lines including, without limitation, 32D, DA2, DA1G, T10, B9, B9/11, BaF3, MC9/G, M* (preB M*), 2E8, RB5, DAI, 123, T1165, HT2, CTLL2, TF-1, Mo7c and CMK. The proteins encoded by the above extended cDNAs or portions thereof may be evaluated for their ability to regulate T cell or thymocyte proliferation in assays such as those described above or in the following references, which are incoφorated herein by reference: Current Protocols in Immunology, Ed. by Coligan et al.., Greene Publishing Associates and Wiley-Interscience; Takai et al. J. Immunol. 137:3494- 3500, 1986., Bertagnolli et al, J. Immunol. 145:1706-1712, 1990., Bertagnolli et al, Cell. Immunol. 133:327-341, 1991; Bertagnolli, et al, J. Immunol 149:3778-3783, 1992; Bowman et al, J. Immunol 152:1756-1761, 1994. In addition, numerous assays for cytokine production and/or the proliferation of spleen cells, lymph node cells and thymocytes are known. These include the techniques disclosed in Current Protocols in Immunology, supra 1:3.12.1-3.12.14; and Schreiber In Current Protocols in Immunology, supra 1 : 6.8.1-6.8.8.
The proteins encoded by the cDNAs may also be assayed for the ability to regulate the proliferation and differentiation of hematopoietic or lymphopoietic cells. Many assays for such activity are familiar to those skilled in the art, including the assays in the following references, which are incoφorated herein by reference: Bottomly et al, In Current Protocols in Immunology., supra. 1 : 6.3.1-6.3.12,; deVries et al, J. Exp. Med. 173:1205-1211, 1991; Moreau et al, Nature 36:690-692, 1988; Greenberger et al, Proc. Natl. Acad Sci. U.S.A. 80:2931-2938, 1983; Nordan, R., In Current Protocols in Immunology., supra. 1 : 6.6.1- 6.6.5; Smith et al., Proc. Natl. Acad Sci. U.S.A. 83:1857-1861, 1986; Bennett et al, in Current Protocols in Immunology supra 1 : 6.15.1; Ciarletta et al, In Current Protocols in Immunology, supra 1 : 6.13.1.
The proteins encoded by the cDNAs may also be assayed for their ability to regulate T-cell responses to antigens. Many assays for such activity are familiar to those skilled in the art, including the assays described in the following references, which are incoφorated herein by reference: Chapter 3 (In Vitro Assays for Mouse Lymphocyte Function), Chapter 6 (Cytokines and Their Cellular Receptors) and Chapter 7, (Immunologic Studies in Humans) in Current Protocols in Immunology supra; Weinberger et al, Proc. Natl. Acad. Sci. USA 77:6091-6095, 1980; Weinberger et al, Eur. J. Immun. 11:405-411, 1981; Takai et al, J. Immunol. 137:3494-3500, 1986; Takai etal., J. Immunol. 140:508-512, 1988. Those proteins which exhibit cytokine, cell proliferation, or cell differentiation activity may then be formulated as pharmaceuticals and used to treat clinical conditions in which induction of cell proliferation or differentiation is beneficial. Alternatively, as described in more detail below, genes encoding these proteins or nucleic acids regulating the expression of these proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
EXAMPLE 33
Assaying the Proteins Expressed from Extended cDNAs or Portions Thereof for Activity as Immune System Regulators
The proteins encoded by the cDNAs may also be evaluated for their effects as immune regulators. For example, the proteins may be evaluated for their activity to influence thymocyte or splenocyte cytotoxicity. Numerous assays for such activity are familiar to those skilled in the art including the assays described in the following references, which are incoφorated herein by reference: Chapter 3 (In Vitro Assays for Mouse Lymphocyte
Function 3.1-3.19) and Chapter 7 (Immunologic studies in Humans) in Current Protocols in
Immunology , Coligan et al., Eds, Greene Publishing Associates and Wiley-Interscience;
Herrmann et al, Proc. Natl. Acad Sci. USA 78:2488-2492, 1981; Herrmann et al, J.
Immunol. 128:1968-1974, 1982; Handa et al., J. Immunol. 135:1564-1572, 1985; Takai et al, J. Immunol. 137:3494-3500, 1986; Takai et al, J. Immunol. 140:508-512, 1988;
Bowman et al, J. Virology 61:1992-1998; Bertagnolli et al, Cell. Immunol. 133:327-341,
1991; Brown etal, J. Immunol 153:3079-3092, 1994.
The proteins encoded by the cDNAs may also be evaluated for their effects on T-cell dependent immunoglobulin responses and isotype switching. Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are incoφorated herein by reference: Maliszewski, J. Immunol. 144:3028-
3033, 1990; Mond etal. in Current Protocols in Immunology, 1 : 3.8.1-3.8.16, supra.
The proteins encoded by the cDNAs may also be evaluated for their effect on immune effector cells, including their effect on Thl cells and cytotoxic lymphocytes. Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are incoφorated herein by reference: Chapter 3 (In Vitro Assays for Mouse1 Lymphocyte Function 3.1-3.19) and Chapter 7 (Immunologic Studies in Humans) in Current Protocols in Immunology, supra; Takai et al, J. Immunol. 137:3494-3500, 1986; Takai et α/., J. Immunol 140:508-512, 1988; Bertagnolli et l., J. Immunol. 149:3778-3783, 1992. The proteins encoded by the cDNAs may also be evaluated for their effect on dendritic cell mediated activation of naive T-cells. Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are incoφorated herein by reference: Guery et al, J. Immunol 134:536-544, 1995; Inaba et tf/., J. Exp. Med 173:549-559, 1991; Macatonia et α/., J. Immunol 154:5071-5079, 1995; Porgador et al.J. Exp. Med 182:255-260, 1995; Nair et al, J. Virol. 67:4062-4069, 1993; Huang et al, Science 264:961-965, 1994; Macatonia et al.J. Exp. Med 169: 1255- 1264, 1989; Bhardwaj et al, Journal of Clinical Investigation 94:797-807, 1994; and Inaba et al, J. Exp. Med 172:631-640, 1990.
The proteins encoded by the cDNAs may also be evaluated for their influence on the lifetime of lymphocytes. Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are incoφorated herein by reference: Darzynkiewicz et al, Cytometry 13:795-808, 1992; Gorczyca et al, Leukemia 7:659-670, 1993; Gorczyca etal, Cancer Res. 53:1945-1951, 1993; Ito etal, Cell 66:233- 243, 1991; Zacharchuk, J. Immunol. 145:4037-4045, 1990; Zamai et al, Cytometry 14:891- 897, 1993; Gorczyca et al, Int. J. Oncol. 1:639-648, 1992.
The proteins encoded by the cDNAs may also be evaluated for their influence on early steps of T-cell commitment and development. Numerous assays for such activity are familiar to those skilled in the art, including without limitation the assays disclosed in the following references, which are incoφorated herein by references: Antica et al, Blood 84:111-117, 1994; Fine et al, Cell. Immunol. 155:1 11-122, 1994; Galy et al, Blood 85:2770-2778, 1995; Toki etal, Proc. Nat. Acad Sci. USA 88:7548-7551, 1991.
Those proteins which exhibit activity as immune system regulators activity may then be formulated as pharmaceuticals and used to treat clinical conditions in which regulation of immune activity is beneficial. For example, the protein may be useful in the treatment of various immune deficiencies and disorders (including severe combined immunodeficiency), e.g., in regulating (up or down) growth and proliferation of T and/or B lymphocytes, as well as effecting the cytolytic activity of NK cells and other cell populations. These immune deficiencies may be genetic or be caused by viral (e.g., HIV) as well as bacterial or fungal infections, or may result from autoimmune disorders. More specifically, infectious diseases caused by viral, bacterial, fungal or other infection may be treatable using a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention, including infections by HIV, hepatitis viruses, heφesviruses, mycobacteria, Leishmania spp., plamodium and various fungal infections such as candidiasis. Of course, in this regard, a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also be useful where a boost to the immune system generally may be desirable, i.e., in the treatment of cancer. Alternatively, proteins encoded by extended cDNAs derived from the 5' ESTs of the present invention may be used in treatment of autoimmune disorders including, for example, connective tissue disease, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, autoimmune pulmonary inflammation, Guillain-Barre syndrome, autoimmune thyroiditis, insulin dependent diabetes mellitis, myasthenia gravis, graft-versus-host disease and autoimmune inflammatory eye disease. Such a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also to be useful in the treatment of allergic reactions and conditions, such as asthma (particularly allergic asthma) or other respiratory problems. Other conditions, in which immune suppression is desired (including, for example, organ transplantation), may also be treatable using a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention.
Using the proteins of the invention it may also be possible to regulate immune responses either up or down.
Down regulation may involve inhibiting or blocking an immune response already in progress or may involve preventing the induction of an immune response. The functions of activated T-cells may be inhibited by suppressing T cell responses or by inducing specific tolerance in T cells, or both. Immunosuppression of T cell responses is generally an active non-antigen-specific process which requires continuous exposure of the T cells to the suppressive agent. Tolerance, which involves inducing non-responsiveness or anergy in T cells, is distinguishable from immunosuppression in that it is generally antigen-specific and persists after the end of exposure to the tolerizing agent. Operationally, tolerance can be demonstrated by the lack of a T cell response upon reexposure to specific antigen in the absence of the tolerizing agent.
Down regulating or preventing one or more antigen functions (including without limitation B lymphocyte antigen functions, such as, for example, B7 costimulation), e.g., preventing high level lymphokine synthesis by activated T cells, will be useful in situations of tissue, skin and organ transplantation and in graft-versus-host disease (GVHD). For example, blockage of T cell function should result in reduced tissue destruction in tissue transplantation. Typically, in tissue transplants, rejection of the transplant is initiated through its recognition as foreign by T cells, followed by an immune reaction that destroys the transplant. The administration of a molecule which inhibits or blocks interaction of a B7 lymphocyte antigen with its natural ligand(s) on immune cells (such as a soluble, monomeric form of a peptide having B7-2 activity alone or in conjunction with a monomeric form of a peptide having an activity of another B lymphocyte antigen (e.g., B7-1, B7-3) or blocking antibody), prior to transplantation, can lead to the binding of the molecule to the natural ligand(s) on the immune cells without transmitting the corresponding costimulatory signal. Blocking B lymphocyte antigen function in this matter prevents cytokine synthesis by immune cells, such as T cells, and thus acts as an immunosuppressant. Moreover, the lack of costimulation may also be sufficient to anergize the T cells, thereby inducing tolerance in a subject. Induction of long-term tolerance by B lymphocyte antigen-blocking reagents may avoid the necessity of repeated administration of these blocking reagents. To achieve sufficient immunosuppression or tolerance in a subject, it may also be necessary to block the function of a combination of B lymphocyte antigens.
The efficacy of particular blocking reagents in preventing organ transplant rejection or GVHD can be assessed using animal models that are predictive of efficacy in humans. Examples of appropriate systems which can be used include allogeneic cardiac grafts in rats and xenogeneic pancreatic islet cell grafts in mice, both of which have been used to examine the immunosuppressive effects of CTLA4Ig fusion proteins in vivo as described in Lenschow et al, Science 257:789-792, 1992 and Turka et al, Proc. Natl Acad. Sci USA, 89:11102- 11105, 1992. In addition, murine models of GVHD (see Paul ed., Fundamental Immunology, Raven Press, New York, 1989, pp. 846-847) can be used to determine the effect of blocking B lymphocyte antigen function in vivo on the development of that disease. Blocking antigen function may also be therapeutically useful for treating autoimmune diseases. Many autoimmune disorders are the result of inappropriate activation of T cells that are reactive against self tissue and which promote the production of cytokines and autoantibodies involved in the pathology of the diseases. Preventing the activation of autoreactive T cells may reduce or eliminate disease symptoms. Administration of reagents which block costimulation of T cells by disrupting receptor/ligand interactions of B lymphocyte antigens can be used to inhibit T cell activation and prevent production of autoantibodies or T cell-derived cytokines which potentially involved in the disease process. Additionally, blocking reagents may induce antigen-specific tolerance of autoreactive T cells which could lead to long-term relief from the disease. The efficacy of blocking reagents in preventing or alleviating autoimmune disorders can be determined using a number of well- characterized animal models of human autoimmune diseases. Examples include murine experimental autoimmune encephalitis, systemic lupus erythmatosis in MRL/pr/pr mice or NZB hybrid mice, murine autoimmuno collagen arthritis, diabetes mellitus in OD mice and BB rats, and murine experimental myasthenia gravis (see Paul ed., supra, pp. 840-856).
Upregulation of an antigen function (preferably a B lymphocyte antigen function), as a means of up regulating immune responses, may also be useful in therapy. Upregulation of immune responses may involve either enhancing an existing immune response or eliciting an initial immune response as shown by the following examples. For instance, enhancing an immune response through stimulating B lymphocyte antigen function may be useful in cases of viral infection. In addition, systemic viral diseases such as influenza, the common cold, and encephalitis might be alleviated by the administration of stimulatory form of B lymphocyte antigens systemically.
Alternatively, antiviral immune responses may be enhanced in an infected patient by removing T cells from the patient, costimulating the T cells in vitro with viral antigen-pulsed APCs either expressing a peptide encoded by extended cDNAs derived from the 5' ESTs of the present invention or together with a stimulatory form of a soluble peptide encoded by extended cDNAs derived from the 5' ESTs of the present invention and reintroducing the in vitro primed T cells into the patient. The infected cells would now be capable of delivering a costimulatory signal to T cells in vivo, thereby activating the T cells. In'another application, upregulation or enhancement of antigen function (preferably B lymphocyte antigen function) may be useful in the induction of tumor immunity. Tumor cells (e.g., sarcoma, melanoma, lymphoma, leukemia, neuroblastoma, carcinoma) transfected with a nucleic acid encoding at least one peptide encoded by extended cDNAs derived from the 5' ESTs of the present invention can be administered to a subject to overcome tumor-specific tolerance in the subject. If desired, the tumor cell can be transfected to express a combination of peptides. For example, tumor cells obtained from a patient can be transfected ex vivo with an expression vector directing the expression of a peptide having B7-2-like activity alone, or in conjunction with a peptide having B7-l-like activity and or B7-3-like activity. The transfected tumor cells are returned to the patient to result in expression of the peptides on the surface of the transfected cell. Alternatively, gene therapy techniques can be used to target a tumor cell for transfection in vivo.
The presence of the peptide encoded by extended cDNAs derived from the 5' ESTs of the present invention having the activity of a B lymphocyte antigen(s) on the surface of the tumor cell provides the necessary costimulation signal to T cells to induce a T cell mediated immune response against the transfected tumor cells. In addition, tumor cells which lack or which fail to reexpress sufficient amounts of MHC class I or MHC class II molecules can be transfected with nucleic acids encoding all or a portion of (e.g., a cytoplasmic-domain truncated portion) of an MHC class I α chain and β microglobulin or an MHC class II α chain and an MHC class El β chain to thereby express MHC class I or MHC class II proteins on the cell surface, respectively. Expression of the appropriate MHC class I or class II molecules in conjunction with a peptide having the activity of a B lymphocyte antigen (e.g., B7-1, B7-2, B7-3) induces a T cell mediated immune response against the transfected tumor cell. Optionally, a gene encoding an antisense construct which blocks expression of an MHC class II associated protein, such as the invariant chain, can also be cotransfected with a DNA encoding a peptide having the activity of a B lymphocyte antigen to promote presentation of tumor associated antigens and induce tumor specific immunity. Thus, the induction of a T cell mediated immune response in a human subject may be sufficient to overcome tumor- specific tolerance in the subject. Alternatively, as described in more detail below, genes encoding these immune system regulator proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
EXAMPLE 34 Assaying the Proteins Expressed from Extended cDNAs or Portions Thereof for Hematopoiesis Regulating Activity The proteins encoded by the extended cDNAs or portions thereof may also be evaluated for their hematopoiesis regulating activity. For example, the effect of the proteins on embryonic stem cell differentiation may be evaluated. Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are incoφorated herein by reference: Johansson et al Cell Biol 15: 141- 151, 1995; Keller et al, Mol Cell. Biol 13:473-486, 1993; McClanahan et al, Blood 81:2903-2915, 1993.
The proteins encoded by the extended cDNAs or portions thereof may also be evaluated for their influence on the lifetime of stem cells and stem cell differentiation. Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are incoφorated herein by reference: Freshney, Methylcellulose Colony Forming Assays, in Culture of Hematopoietic Cells., Freshney, et al.. Eds. pp. 265-268, Wiley-Liss, Inc., New York, NY. 1994; Hirayama et al, Proc. Natl. Acad Sci. USA 89:5907-5911, 1992; McNiece and Briddell, in Culture of Hematopoietic Cells, supra; Neben et al, Exp. Hematol. 22:353-359, 1994; Ploemacher and Cobblestone In Culture of Hematopoietic Cells, supra\-2\, Spooncer et al, in Culture of Hematopoietic Cells, supra\63-\79 and Sutherland in Culture of Hematopoietic Cells, supra. 139-162.
Those proteins which exhibit hematopoiesis regulatory activity may then be formulated as pharmaceuticals and used to treat clinical conditions in which regulation of hematopoeisis is beneficial, such as in the treatment of myeloid or lymphoid cell deficiencies. Involvement in regulating hematopoiesis is indicated even by marginal biological activity in support of colony forming cells or of factor-dependent cell lines. For example, proteins supporting the growth and proliferation of erythroid progenitor cells alone or in combination with other cytokines, indicates utility, for example, in treating various anemias or for use in conjunction with irradiation/chemotherapy to stimulate the production of erythroid precursors and or erythroid cells. Proteins supporting the growth and proliferation of myeloid cells such as granulocytes and monocytes/macrophages (i.e., traditional CSF activity) may be useful, for example, in conjunction with chemotherapy to prevent or treat consequent myelo- suppression. Proteins supporting the growth and proliferation of megakaryocytes and consequently of platelets allows prevention or treatment of various platelet disorders such as thrombocytopenia, and generally may be used in place of or complementary to platelet transfusions. Proteins supporting the growth and proliferation of hematopoietic stem cells which are capable of maturing to any and all of the above-mentioned hematopoietic cells may therefore find therapeutic utility in various stem cell disorders (such as those usually treated with transplantion, including, without limitation, aplastic anemia and paroxysmal nocturnal hemoglobinuria), as well as in repopulating the stem cell compartment post irradiation/chemotherapy, either in vivo or ex vivo (i.e., in conjunction with bone marrow transplantation or with peripheral progenitor cell transplantation (homologous or heterologous)) as normal cells or genetically manipulated for gene therapy. Alternatively, as described in more detail below, genes encoding hematopoiesis regulating activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
EXAMPLE 35 Assaying the Proteins Expressed from Extended cDNAs or Portions Thereof for Regulation of Tissue Growth
The proteins encoded by the extended cDNAs or portions thereof may also be evaluated for their effect on tissue growth. Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in International Patent Publication No. WO95/16035, International Patent Publication No. WO95/05846 and International Patent
Publication No. W091/07491, which are incoφorated herein by reference.
Assays for wound healing activity include, without limitation, those described in: Winter, Epidermal Wound Healing, pps. 71-112, Maibach and Rovee, eds., Year Book Medical Publishers, Inc., Chicago, as modified by Eaglstein and Mertz, J. Invest. Dermatol 71:382-84, 1978, which are incoφorated herein by reference. Those proteins which are involved in the regulation of tissue growth may then be formulated as pharmaceuticals and used to treat clinical conditions in which regulation of tissue growth is beneficial. For example, a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention also may have utility in compositions used for bone, cartilage, tendon, ligament and or nerve tissue growth or regeneration, as well as for wound healing and tissue repair and replacement, and in the treatment of burns, incisions and ulcers.
A protein encoded by extended cDNAs derived from the 5' ESTs of the present invention, which induces cartilage and/or bone growth in circumstances where bone is not normally formed, has application in the healing of bone fractures and cartilage damage or defects in humans and other animals. Such a preparation employing a protein of the invention may have prophylactic use in closed as well as open fracture reduction and also in the improved fixation of artificial joints. De novo bone synthesis induced by an osteogenic agent contributes to the repair of congenital, trauma induced, or oncologic resection induced craniofacial defects, and also is useful in cosmetic plastic surgery. A protein of this invention may also be used in the treatment of periodontal disease, and in other tooth repair processes. Such agents may provide an environment to attract bone- forming cells, stimulate growth of bone-forming cells or induce differentiation of bone- forming cell progenitors. A protein of the invention may also be useful in the treatment of osteoporosis or osteoarthritis, such as through stimulation of bone and/or cartilage repair or by blocking inflammation or processes of tissue destruction (coUagenase activity, osteoclast activity, etc.) mediated by inflammatory processes.
Another category of tissue regeneration activity that may be attributable to the protein encoded by extended cDNAs derived from the 5' ESTs of the present invention is tendon/ligament formation. A protein encoded by extended cDNAs derived from the 5' ESTs of the present invention, which induces tendon/ligament-like tissue or other tissue formation in circumstances where such tissue is not normally formed, has application in the healing of tendon or ligament tears, deformities and other tendon or ligament defects in humans and other animals. Such a preparation employing a tendon/ligament-like tissue inducing protein may have prophylactic use in preventing damage to tendon or ligament tissue, as well as use in the improved fixation of tendon or ligament to bone or other tissues, and in repairing defects to tendon or ligament tissue. De novo tendon/ligament-like tissue formation induced by a composition encoded by extended cDNAs derived from the 5' ESTs of the present invention contributes to the repair of tendon or ligaments defects of congenital, traumatic or other origin and is also useful in cosmetic plastic surgery for attachment or repair of tendons or ligaments. The compositions encoded by extended cDNAs derived from the 5' ESTs of the present invention may provide an environment to attract tendon- or ligament- forming cells, stimulate growth of tendon- or ligament-forming cells, induce differentiation of progenitors of tendon- or ligament-forming cells, or induce growth of tendon/ligament cells or progenitors ex vivo for return in vivo to effect tissue repair. The compositions of the invention may also be useful in the treatment of tendinitis, caφal tunnel syndrome and other tendon or ligament defects. The compositions may also include an appropriate matrix and/or sequestering agent as a carrier as is well known in the art.
The protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also be useful for proliferation of neural cells and for regeneration of nerve and brain tissue, i.e., for the treatment of central and peripheral nervous system diseases and neuropathies, as well as mechanical and traumatic disorders, which involve degeneration, death or trauma to neural cells or nerve tissue. More specifically, a protein may be used in the treatment of diseases of the peripheral nervous system, such as peripheral nerve injuries, peripheral neuropathy and localized neuropathies, and central nervous system diseases, such as Alzheimer's, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome. Further conditions which may be treated in accordance with the present invention include mechanical and traumatic disorders, such as spinal cord disorders, head trauma and cerebrovascular diseases such as stroke. Peripheral neuropathies resulting from chemotherapy or other medical therapies may also be treatable using a protein of the invention. Proteins of the invention may also be useful to promote better or faster closure of non-healing wounds, including without limitation pressure ulcers, ulcers associated with vascular insufficiency, surgical and traumatic wounds, and the like.
It is expected that a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also exhibit activity for generation or regeneration of other tissues, such as organs (including, for example, pancreas, liver, intestine, kidney, skin, endothelium) muscle (smooth, skeletal or cardiac) and vascular (including vascular endothelium) tissue, or for promoting the growth of cells comprising such tissues. Part of the desired effects may be by inhibition or modulation of fibrotic scarring to allow normal tissue to generate. A protein of the invention may also exhibit angiogenic activity.
A protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also be useful for gut protection or regeneration and treatment of lung or liver fibrosis, reperfusion injury in various tissues, and conditions resulting from systemic cytokine damage.
A protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also be useful for promoting or inhibiting differentiation of tissues described above from precursor tissues or cells; or for inhibiting the growth of tissues described above. Alternatively, as described in more detail below, genes encoding tissue growth regulating activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
EXAMPLE 36 Assaying the Proteins Expressed from Extended cDNAs or Portions Thereof for Regulation of Reproductive Hormones The proteins encoded by the extended cDNAs or portions thereof may also be evaluated for their ability to regulate reproductive hormones, such as follicle stimulating hormone. Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are incoφorated herein by reference: Vale et al, Endocrinol. 91:562-572, 1972; Ling et al, Nature 321:779-782, 1986; Vale et al, Nature 321:776-779, 1986; Mason et al., Nature 318:659-663, 1985; Forage et al, Proc. Natl Acad. Sci. USA 83:3091-3095, 1986, Chapter 6.12 in Current Protocols in Immunology, Coligan et al. Eds. Greene Publishing Associates and Wiley-Intersciece ; Taub et al, J. Clin. Invest. 95:1370-1376, 1995; Lind et al, APMS 103:140-146, 1995; Muller et al, Eur. J. Immunol 25:1744-1748; Gruber et al, J. Immunol. 152:5860-5867, 1994; Johnston et al, J Immunol. 153:1762-1768, 1994. Those proteins which exhibit activity as reproductive hormones or regulators of cell movement may then be formulated as pharmaceuticals and used to treat clinical conditions in which regulation of reproductive hormones are beneficial. For example, a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may also exhibit activin- or inhibin-related activities. Inhibins are characterized by their ability to inhibit the release of follicle stimulating hormone (FSH), while activins are characterized by their ability to stimulate the release of FSH. Thus, a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention, alone or in heterodimers with a member of the inhibin α family, may be useful as a contraceptive based on the ability of inhibins to decrease fertility in female mammals and decrease spermatogenesis in male mammals. Administration of sufficient amounts of other inhibins can induce infertility in these mammals. Alternatively, the protein of the invention, as a homodimer or as a heterodimer with other protein subunits of the inhibin-B group, may be useful as a fertility inducing therapeutic, based upon the ability of activin molecules in stimulating FSH release from cells of the anterior pituitary. See, for example, United States Patent 4,798,885, the disclosure of which is incoφorated herein by reference. A protein of the invention may also be useful for advancement of the onset of fertility in sexually immature mammals, so as to increase the lifetime reproductive performance of domestic animals such as cows, sheep and pigs.
Alternatively, as described in more detail below, genes encoding reproductive hormone regulating activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
EXAMPLE 37
Assaying the Proteins Expressed from Extended cDNAs or
Portions Thereof for Chemotactic/Chemokinetic Activity The proteins encoded by the extended cDNAs or portions thereof may also be evaluated for chemotactic/chemokinetic activity. For example, a protein encoded by extended cDNAs derived from the 5' ESTs of the present invention may have chemotactic or chemokinetic activity (e.g., act as a chemokine) for mammalian cells, including, for example, monocytes, fibroblasts, neutrophils, T-cells, mast cells, eosinophils, epithelial and/or endothelial cells. Chemotactic and chemokinetic proteins can be used to mobilize or attract a desired cell population to a desired site of action. Chemotactic or chemokinetic proteins provide particular advantages in treatment of wounds and other trauma to tissues, as well as in treatment of localized infections. For example, attraction of lymphocytes, monocytes or neutrophils to tumors or sites of infection may result in improved immune responses against the tumor or infecting agent. A protein or peptide has chemotactic activity for a particular cell population if it can stimulate, directly or indirectly, the directed orientation or movement of such cell population. Preferably, the protein or peptide has the ability to directly stimulate directed movement of cells. Whether a particular protein has chemotactic activity for a population of cells can be readily determined by employing such protein or peptide in any known assay for cell chemotaxis.
The activity of a protein of the invention may, among other means, be measured by the following methods:
Assays for chemotactic activity (which will identify proteins that induce or prevent chemotaxis) consist of assays that measure the ability of a protein to induce the migration of cells across a membrane as well as the ability of a protein to induce the adhesion of one cell population to another cell population. Suitable assays for movement and adhesion include, without limitation, those described in: Current Protocols in Immunology, Ed by Coligan, Kruisbeek, Margulies, Shevach and Strober, Pub. Greene Publishing Associates and Wiley- Interscience, Chapter 6.12: 6.12.1-6.12.28; Taub et α/., J. Clin. Invest. 95:1370-1376, 1995; Lind et al, APMS 103:140-146, 1995; Mueller et al, Eur. J. Immunol. 25:1744-1748; Gruber et al, J. Immunol. 152:5860-5867, 1994; Johnston et al. J. Immunol, 153:1762- 1768, 1994.
EXAMPLE 38 Assaying the Proteins Expressed from Extended cDNAs or
Portions Thereof for Regulation of Blood Clotting
The proteins encoded by the extended cDNAs or portions thereof may also be evaluated for their effects on blood clotting. Numerous assays for such activity are familiar to those skilled in the art, including the assays disclosed in the following references, which are incoφorated herein by reference: Linet et al, J. Clin. Pharmacol. 26: 131-140, 1986; Burdick et al, Thrombosis Res. 45:413-419, 1987; Humphrey et al, Fibrinolysis 5:71-79, 1991; Schaub, Prostagl ndins 35:467-474, 1988.
Those proteins which are involved in the regulation of blood clotting may then be formulated as pharmaceuticals and used to treat clinical conditions in which regulation of blood clotting is beneficial. For example, a protein of the invention may also exhibit hemostatic or thrombolytic activity. As a result, such a protein is expected to be useful in treatment of various coagulations disorders (including hereditary disorders, such as hemophilias) or to enhance coagulation and other hemostatic events in treating wounds resulting from trauma, surgery or other causes. A protein of the invention may also be useful for dissolving or inhibiting formation of thromboses and for treatment and prevention of conditions resulting therefrom (such as infarction of cardiac and central nervous system vessels (e.g., stroke)). Alternatively, as described in more detail below, genes encoding blood clotting activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
EXAMPLE 39
Assaying the Proteins Expressed from Extended cDNAs or Portions Thereof for Involvement in Receptor/Ligand Interactions The proteins encoded by the extended cDNAs or a portion thereof may also be evaluated for their involvement in receptor/ligand interactions. Numerous assays for such involvement are familiar to those skilled in the art, including the assays disclosed in the following references, which are incoφorated herein by reference: Chapter 7. 7.28.1-7.28.22 in Current Protocols in Immunology, Coligan et al. Eds. Greene Publishing Associates and Wiley-Interscience; Takai et al, Proc. Natl Acad Sci. USA 84:6864-6868, 1987; Bierer et al, J. Exp. Med. 168:1145-1156, 1988; Rosenstein et l, J. Exp. Med. 169: 149-160, 1989; Stoltenborg et al, J. Immunol. Methods 175:59-68, 1994; Stitt et al, Cell 80:661-670, 1995; Gyuris et al, Cell 75:791-803, 1993.
For example, the proteins encoded by extended cDNAs derived from the 5' ESTs of the present invention may also demonstrate activity as receptors, receptor ligands or inhibitors or agonists of receptor/ligand interactions. Examples of such receptors and ligands include, without limitation, cytokine receptors and their ligands, receptor kinases and their ligands, receptor phosphatases and their ligands, receptors involved in cell-cell interactions and their ligands (including without limitation, cellular adhesion molecules (such as selectins, integrins and their ligands) and receptor/ligand pairs involved in antigen presentation, antigen recognition and development of cellular and humoral immune responses). Receptors and ligands are also useful for screening of potential peptide or small molecule inhibitors of the relevant receptor/ligand interaction. A protein encoded by extended cDNAs derived from the 5' ESTs of the present invention (including, without limitation, fragments of receptors and ligands) may themselves be useful as inhibitors of receptor/ligand interactions. Alternatively, as described in more detail below, genes encoding proteins involved in receptor/ligand interactions or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
EXAMPLE 40 Assaying the Proteins Expressed from Extended cDNAs or Portions Thereof for Anti-Inflammatory Activity The proteins encoded by the extended cDNAs or a portion thereof may also be evaluated for anti-inflammatory activity. The anti-inflammatory activity may be achieved by providing a stimulus to cells involved in the inflammatory response, by inhibiting or promoting cell-cell interactions (such as, for example, cell adhesion), by inhibiting or promoting chemotaxis of cells involved in the inflammatory process, inhibiting or promoting cell extravasation, or by stimulating or suppressing production of other factors which more directly inhibit or promote an inflammatory response. Proteins exhibiting such activities can be used to treat flammatory conditions including chronic or acute conditions, including without limitation inflammation associated with infection (such as septic shock, sepsis or systemic inflammatory response syndrome), ischemia-reperfusioninury, endotoxin lethality, arthritis, complement-mediated hyperacute rejection, nephritis, cytokine- or chemokine- induced lung injury, inflammatory bowel disease, Crohn's disease or resulting from over production of cytokines such as TNF or IL-1. Proteins of the invention may also be useful to treat anaphylaxis and hypersensitivity to an antigenic substance or material. Alternatively, as described in more detail below, genes encoding anti-inflammatory activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
EXAMPLE 41 Assaying the Proteins Expressed from Extended cDNAs or
Portions Thereof for Tumor Inhibition Activity The proteins encoded by the extended cDNAs or a portion thereof may also be evaluated for tumor inhibition activity. In addition to the activities described above for immunological treatment or prevention of tumors, a protein of the invention may exhibit other anti-tumor activities. A protein may inhibit tumor growth directly or indirectly (such as, for example, via ADCC). A protein may exhibit its tumor inhibitory activity by acting on tumor tissue or tumor precursor tissue, by inhibiting formation of tissues necessary to support tumor growth (such as, for example, by inhibiting angiogenesis), by causing production of other factors, agents or cell types which inhibit tumor growth, or by suppressing, eliminating or inhibiting factors, agents or cell types which promote tumor growth. Alternatively, as described in more detail below, genes tumor inhibition activity proteins or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
A protein of the invention may also exhibit one or more of the following additional activities or effects: inhibiting the growth, infection or function of, or killing, infectious agents, including, without limitation, bacteria, viruses, fungi and other parasites; effecting (suppressing or enhancing) bodily characteristics, including, without limitation, height, weight, hair color, eye color, skin, fat to lean ratio or other tissue pigmentation, or organ or body part size or shape (such as, for example, breast augmentation or diminution, change in bone form or shape); effecting biorhythms or circadian cycles or rhythms; effecting the fertility of male or female subjects; effecting the metabolism, catabolism, anabolism, processing, utilization, storage or elimination of dietary fat, lipid, protein, carbohydrate, vitamins, minerals, cofactors or other nutritional factors or component(s); effecting behavioral characteristics, including, without limitation, appetite, libido, stress, cognition (including cognitive disorders), depression (including depressive disorders) and violent behaviors; providing analgesic effects or other pain reducing effects; promoting differentiation and growth of embryonic stem cells in lineages other than hematopoietic lineages; hormonal or endocrine activity; in the case of enzymes, correcting deficiencies of the enzyme and treating deficiency-related diseases; treatment of hypeφroliferative disorders (such as, for example, psoriasis); immunoglobulin-like activity (such as, for example, the ability to bind antigens or complement); and the ability to act as an antigen in a vaccine composition to raise an immune response against such protein or another material or entity which is cross-reactive with such protein. Alternatively, as described in more detail below, genes encoding proteins involved in any of the above mentioned activities or nucleic acids regulating the expression of such proteins may be introduced into appropriate host cells to increase or decrease the expression of the proteins as desired.
EXAMPLE 42
Identification of Proteins which Interact with Polypeptides Encoded by Extended cDNAs
Proteins which interact with the polypeptides encoded by cDNAs derived from the 5' ESTs or fragments thereof, such as receptor proteins, may be identified using two hybrid systems such as the Matchmaker Two Hybrid System 2 (Catalog No. K 1604-1, Clontech). As described in the manual accompanying the kit which is incoφorated herein by reference, the the cDNAs derived from 5' ESTs, or fragments thereof, are inserted into an expression vector such that they are in frame with DNA encoding the DNA binding domain of the yeast transcriptional activator GAL4. cDNAs in a cDNA library which encode proteins which might interact with the polypeptides encoded by the extended cDNAs or portions thereof are inserted into a second expression vector such that they are in frame with DNA encoding the activation domain of GAL4. The two expression plasmids are transformed into yeast and the yeast are plated on selection medium which selects for expression of selectable markers on each of the expression vectors as well as GAL4 dependent expression of the HIS3 gene. Transformants capable of growing on medium lacking histidine are screened for GAL4 dependent lacZ expression. Those cells which are positive in both the histidine selection and the lacZ assay contain plasmids encoding proteins which interact with the polypeptide encoded by the extended cDNAs or portions thereof. Alternatively, the system described in Lustig et al, Methods in Enzymology 283: 83- 99, 1997, and in U.S. Patent No. 5,654,150, the disclosure of which is incoφorated herein by reference, may be used for identifying molecules which interact with the polypeptides encoded by extended cDNAs. In such systems, in vitro transcription reactions are performed on a pool of vectors containing extended cDNA inserts cloned downstream of a promoter which drives in vitro transcription. The resulting pools of mRNAs are introduced into Xenopus laevis oocytes. The oocytes are then assayed for a desired activity.
Alternatively, the pooled in vitro transcription products produced as described above may be translated in vitro. The pooled in vitro translation products can be assayed for a desired activity or for interaction with a known polypeptide.
Proteins or other molecules interacting with polypeptides encoded by extended cDNAs can be found by a variety of additional techniques. In one method, affinity columns containing the polypeptide encoded by the extended cDNA or a portion thereof can be constructed. In some versions, of this method the affinity column contains chimeric proteins in which the protein encoded by the extended cDNA or a portion thereof is fused to glutathione S-transferase. A mixture of cellular proteins or pool of expressed proteins as described above and is applied to the affinity column. Proteins interacting with the polypeptide attached to the column can then be isolated and analyzed on 2-D electrophoresis gel as described in Ramunsen et al, Electrophoresis 18:588-598, 1997, the disclosure of which is incoφorated herein by reference. Alternatively, the proteins retained on the affinity column can be purified by electrophoresis based methods and sequenced. The same method can be used to isolate antibodies, to screen phage display products, or to screen phage display human antibodies.
Proteins interacting with polypeptides encoded by extended cDNAs or portions thereof can also be screened by using an Optical Biosensor as described in Edwards and Leatherbarrow, Analytical Biochemistry 246:1-6, 1997, the disclosure of which is incorporated herein by reference. The main advantage of the method is that it allows the determination of the association rate between the protein and other interacting molecules. Thus, it is possible to specifically select interacting molecules with a high or low association rate. Typically a target molecule is linked to the sensor surface (through a carboxymethl dextran matrix) and a sample of test molecules is placed in contact with the target molecules. The binding of a test molecule to the target molecule causes a change in the refractive index and/ or thickness. This change is detected by the Biosensor provided it occurs in the evanescent field (which extend a few hundred nanometers from the sensor surface). In these screening assays, the target molecule can be one of the polypeptides encoded by extended cDNAs or a portion thereof and the test sample can be a collection of proteins extracted from tissues or cells, a pool of expressed proteins, combinatorial peptide and/ or chemical libraries, or phage displayed peptides. The tissues or cells from which the test proteins are extracted can originate from any species. In other methods, a target protein is immobilized and the test population is a collection of unique polypeptides encoded by the extended cDNAs or portions thereof.
To study the interaction of the proteins encoded by the extended cDNAs or portions thereof with drugs, the microdialysis coupled to HPLC method described by Wang et al, Chromatographia 44:205-208, 1997 or the affinity capillary electrophoresis method described by Busch et al, J. Chromatogr. 111:311-328, 1997, the disclosures of which are incoφorated herein by reference can be used.
It will be appreciated by those skilled in the art that the proteins expressed from the extended cDNAs or portions may be assayed for numerous activities in addition to those specifically enumerated above. For example, the expressed proteins may be evaluated for applications involving control and regulation of inflammation, tumor proliferation or metastasis, infection, or other clinical conditions. In addition, the proteins expressed from the extended cDNAs or portions thereof may be useful as nutritional agents or cosmetic agents.
The proteins expressed from the cDNAs or portions thereof may be used to generate antibodies capable of specifically binding to the expressed protein or fragments thereof as described in Example 40 below. The antibodies may capable of binding a full length protein encoded by a cDNA derived from a 5' EST, a mature protein (i.e. the protein generated by cleavage of the signal peptide) encoded by a cDNA derived from a 5' EST, or a signal peptide encoded by a cDNA derived from a 5' EST. Alternatively, the antibodies may be capable of binding fragments of at least 10 amino acids of the proteins encoded by the above cDNAs. In some embodiments, the antibodies may be capable of binding fragments of at least 15 amino acids of the proteins encoded by the above cDNAs. In other embodiments, the antibodies may be capable of binding fragments of at least 25 amino acids of the proteins expressed from the extended cDNAs which comprise at least 25 amino acids of the proteins encoded by the above cDNAs. In further embodiments, the antibodies may be capable of binding fragments of at least 40 amino acids of the proteins encoded by the above cDNAs.
EXAMPLE 43
Production of an Antibody to a Human Protein
Substantially pure protein or polypeptide is isolated from the transfected or transformed cells as described in Example 30. The concentration of protein in the final preparation is adjusted, for example, by concentration on an Amicon filter device, to the level of a few μg/ml. Monoclonal or polyclonal antibody to the protein can then be prepared as follows:
1. Monoclonal Antibody Production by Hvbridoma Fusion Monoclonal antibody to epitopes of any of the peptides identified and isolated as described can be prepared from murine hybridomas according to the classical method of Kohler, and Milstein, Nature 256:495, 1975 or derivative methods thereof. Briefly, a mouse is repetitively inoculated with a few micrograms of the selected protein or peptides derived therefrom over a period of a few weeks. The mouse is then sacrificed, and the antibody producing cells of the spleen isolated. The spleen cells are fused by means of polyethylene glycol with mouse myeloma cells, and the excess unfϊised cells destroyed by growth of the system on selective media comprising aminopterin (HAT media). The successfully fused cells are diluted and aliquots of the dilution placed in wells of a microtiter plate where growth of the culture is continued. Antibody-producing clones are identified by detection of antibody in the supernatant fluid of the wells by immunoassay procedures, such as ELISA, as originally described by Engvall, Meth. Enzymol. 70:419, 1980, the disclosure of which is incoφorated herein by reference and derivative methods thereof. Selected positive clones can be expanded and their monoclonal antibody product harvested for use. Detailed procedures for monoclonal antibody production are described in Davis et al. in Basic Methods in Molecular Biology Elsevier, New York. Section 21-2, the disclosure of which is incoφorated herein by reference.
2. Polyclonal Antibody Production by Immunization Polyclonal antiserum containing antibodies to heterogenous epitopes of a single protein can be prepared by immunizing suitable animals with the expressed protein or peptides derived therefrom, which can be unmodified or modified to enhance immunogenicity. Effective polyclonal antibody production is affected by many factors related both to the antigen and the host species. For example, small molecules tend to be less immunogenic than others and may require the use of carriers and adjuvant. Also, host animals response vary depending on site of inoculations and doses, with both inadequate or excessive doses of antigen resulting in low titer antisera. Small doses (ng level) of antigen administered at multiple intradermal sites appears to be most reliable. An effective immunization protocol for rabbits can be found in Vaitukaitis. et al, J. Clin. Endocrinol Metab. 33:988-991 (1971) , the disclosure of which is incoφorated herein by reference.
Booster injections can be given at regular intervals, and antiserum harvested when antibody titer thereof, as determined semi-quantitatively, for example, by double irnmunodifiiision in agar against known concentrations of the antigen, begins to fall. See, for example, Ouchterlony, et al, Chap. 19 in: Handbook of Experimental Immunology D. Wier (ed) Blackwell (1973) , the disclosure of which is incoφorated herein by reference. Plateau concentration of antibody is usually in the range of 0.1 to 0.2 mg/ml of serum (about 12 μM). Affinity of the antisera for the antigen is determined by preparing competitive binding curves, as described, for example, by Fisher, D., Chap. 42 in: Manual of Clinical Immunology, 2d Ed. (Rose and Friedman, Eds.) Amer. Soc. For Microbiol., Washington, D.C. (1980) , the disclosure of which is incoφorated herein by reference.
Antibody preparations prepared according to either protocol are useful in quantitative immunoassays which determine concentrations of antigen-bearing substances in biological samples; they are also used semi-quantitatively or qualitatively to identify the presence of antigen in a biological sample. The antibodies may also be used in therapeutic compositions for killing cells expressing the protein or reducing the levels of the protein in the body. V. Use of 5' ESTs or Sequences Obtainable Therefrom or Portions Thereof as Reagents
The 5' ESTs of the present invention (or cDNAs or genomic DNAs obtainable therefrom) may be used as reagents in isolation procedures, diagnostic assays, and forensic procedures. For example, sequences from the 5' ESTs (or cDNAs or genomic DNAs obtainable therefrom) may be detectably labeled and used as probes to isolate other sequences capable of hybridizing to them. In addition, sequences from 5' ESTs (or cDNAs or genomic DNAs obtainable therefrom) may be used to design PCR primers to be used in isolation, diagnostic, or forensic procedures.
1. Use of 5' ESTs or Sequences Obtainable Therefrom or Portions Thereof in Isolation, Diagnostic and Forensic Procedures
EXAMPLE 44 Preparation of PCR Primers and Amplification of DNA
The 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom) may be used to prepare PCR primers for a variety of applications, including isolation procedures for cloning nucleic acids capable of hybridizing to such sequences, diagnostic techniques and forensic techniques. The PCR primers are at least 10 bases, and preferably at least 12, 15, or 17 bases in length. More preferably, the PCR primers are at least 20-30 bases in length. In some embodiments, the PCR primers may be more than 30 bases in length. It is preferred that the primer pairs have approximately the same G/C ratio, so that melting temperatures are approximately the same. A variety of PCR techniques are familiar to those skilled in the art. For a review of PCR technology, see Molecular Cloning to Genetic Engineering, White Ed. in Methods in Moleailar Biology 67: Humana Press, Totowa 1997, the disclosure of which is incoφorated herein by reference. In each of these PCR procedures, PCR primers on either side of the nucleic acid sequences to be amplified are added to a suitably prepared nucleic acid sample along with dNTPs and a thermostable polymerase such as Taq polymerase, Pfu polymerase, or Vent polymerase. The nucleic acid in the sample is denatured and the PCR primers are specifically hybridized to complementary nucleic acid sequences in the sample. The hybridized primers are extended. Thereafter, another cycle of denaturation, hybridization, and extension is initiated. The cycles are repeated multiple times to produce an amplified fragment containing the nucleic acid sequence between the primer sites.
EXAMPLE 45 Use of 5 'ESTs as Probes
Probes derived from 5' ESTs (or cDNAs or genomic DNAs obtainable therefrom), including full length cDNAs or genomic sequences, may be labeled with detectable labels familiar to those skilled in the art, including radioisotopes and non-radioactive labels, to provide a detectable probe. The detectable probe may be single stranded or double stranded and may be made using techniques known in the art, including in vitro transcription, nick translation, or kinase reactions. A nucleic acid sample containing a sequence capable of hybridizing to the labeled probe is contacted with the labeled probe. If the nucleic acid in the sample is double stranded, it may be denatured prior to contacting the probe. In some applications, the nucleic acid sample may be immobilized on a surface such as a nitrocellulose or nylon membrane. The nucleic acid sample may comprise nucleic acids obtained from a variety of sources, including genomic DNA, cDNA libraries, RNA, or tissue samples.
Procedures used to detect the presence of nucleic acids capable of hybridizing to the detectable probe include well known techniques such as Southern blotting, Northern blotting, dot blotting, colony hybridization, and plaque hybridization. Ln some applications, the nucleic acid capable of hybridizing to the labeled probe may be cloned into vectors such as expression vectors, sequencing vectors, or in vitro transcription vectors to facilitate the characterization and expression of the hybridizing nucleic acids in the sample. For example, such techniques may be used to isolate and clone sequences in a genomic library or cDNA library which are capable of hybridizing to the detectable probe as described in Example 30 above. PCR primers made as described in Example 44 above may be used in forensic analyses, such as the DNA fingeφrinting techniques described in Examples 46-50 below. Such analyses may utilize detectable probes or primers based on the sequences of the the 5' ESTs or of cDNAs or genomic DNAs isolated using the 5' ESTs. EXAMPLE 46
Forensic Matching by DNA Sequencing In one exemplary method, DNA samples are isolated from forensic specimens of, for example, hair, semen, blood or skin cells by conventional methods. A panel of PCR primers based on a number of the 5' ESTs of Example 25, or cDNAs or genomic DNAs isolated therefrom as described above, is then utilized in accordance with Example 44 to amplify DNA of approximately 100-200 bases in length from the forensic specimen. Corresponding sequences are obtained from a test subject. Each of these identification DNAs is then sequenced using standard techniques, and a simple database comparison determines the differences, if any, between the sequences from the subject and those from the sample. Statistically significant differences between the suspect's DNA sequences and those from the sample conclusively prove a lack of identity. This lack of identity can be proven, for example, with only one sequence. Identity, on the other hand, should be demonstrated with a large number of sequences, all matching. Preferably, a minimum of 50 statistically identical sequences of 100 bases in length are used to prove identity between the suspect and the sample.
EXAMPLE 47
Positive Identification by DNA Sequencing The technique outlined in the previous example may also be used on a larger scale to provide a unique fingeφrint-type identification of any individual. In this technique, primers are prepared from a large number of 5'EST sequences from Example 25, or cDNA or genomic DNA sequences obtainable therefrom. Preferably, 20 to 50 different primers are used. These primers are used to obtain a corresponding number of PCR-generated DNA segments from the individual in question in accordance with Example 44. Each of these DNA segments is sequenced, using the methods set forth in Example 46. The database of sequences generated through this procedure uniquely identifies the individual from whom the sequences were obtained. The same panel of primers may then be used at any later time to absolutely correlate tissue or other biological specimen with that individual. EXAMPLE 48
Southern Blot Forensic Identification The procedure of Example 47 is repeated to obtain a panel of at least 10 amplified sequences from an individual and a specimen. Preferably, the panel contains at least 50 amplified sequences. More preferably, the panel contains 100 amplified sequences. In some embodiments, the panel contains 200 amplified sequences. This PCR-generated DNA is then digested with one or a combination of, preferably, four base specific restriction enzymes. Such enzymes are commercially available and known to those of skill in the art. After digestion, the resultant gene fragments are size separated in multiple duplicate wells on an agarose gel and transferred to nitrocellulose using Southern blotting techniques well known to those with skill in the art. For a review of Southern blotting see Davis et al. (Basic Methods in Molecular Biology, 1986, Elsevier Press, pp 62-65) , the disclosure of which is incoφorated herein by reference..
A panel of probes based on the sequences of 5' ESTs (or cDNAs or genomic DNAs obtainable therefrom), or fragments thereof of at least 10 bases, are radioactively or colorimetrically labeled using methods known in the art, such as nick translation or end labeling, and hybridized to the Southern blot using techniques known in the art (Davis et al, supra). Preferably, the probe comprises at least 12, 15, or 17 consecutive nucleotides from the 5' EST (or cDNAs or genomic DNAs obtainable therefrom). More preferably, the probe comprises at least 20-30 consecutive nucleotides from the 5' EST (or cDNAs or genomic DNAs obtainable therefrom). In some embodiments, the probe comprises more than 30 nucleotides from the 5' EST (or cDNAs or genomic DNAs obtainable therefrom).
Preferably, at least 5 to 10 of these labeled probes are used, and more preferably at least about 20 or 30 are used to provide a unique pattern. The resultant bands appearing from the hybridization of a large sample of 5' EST (or cDNAs or genomic DNAs obtainable therefrom) will be a unique identifier. Since the restriction enzyme cleavage will be different for every individual, the band pattern on the Southern blot will also be unique. Increasing the number of 5' EST (or cDNAs or genomic DNAs obtainable therefrom) probes will provide a statistically higher level of confidence in the identification since there will be an increased number of sets of bands used for identification. EXAMPLE 49
Dot Blot Identification Procedure Another technique for identifying individuals using the 5' EST sequences disclosed herein utilizes a dot blot hybridization technique. Genomic DNA is isolated from nuclei of subject to be identified. Oligonucleotide probes of approximately 30 bp in length are synthesized that correspond to at least 10, preferably 50 sequences from the 5' ESTs or cDNAs or genomic DNAs obtainable therefrom. The probes are used to hybridize to the genomic DNA through conditions known to those in the art. The oligonucleotides are end labeled with P32 using polynucleotide kinase (Pharmacia). Dot Blots are created by spotting the genomic DNA onto nitrocellulose or the like using a vacuum dot blot manifold (BioRad, Richmond California). The nitrocellulose filter containing the genomic sequences is baked or UV linked to the filter, prehybridized and hybridized with labeled probe using techniques known in the art (Davis et al, supra). The 32P labeled DNA fragments are sequentially hybridized with successively stringent conditions to detect minimal differences between the 30 bp sequence and the DNA.
Tetramethylammonium chloride is useful for identifying clones containing small numbers of nucleotide mismatches (Wood et al, Proc. Natl. Acad. Sci. USA 82(6): 1585-1588, 1985) which is hereby incoφorated by reference. A unique pattern of dots distinguishes one individual from another individual. 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom) or oligonucleotides containing at least 10 consecutive bases from these sequences can be used as probes in the following alternative fingeφrinting technique. Preferably, the probe comprises at least 12, 15, or 17 consecutive nucleotides from the 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom). More preferably, the probe comprises at least 20-30 consecutive nucleotides from the 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom). In some embodiments, the probe comprises more than 30 nucleotides from the 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom).
Preferably, a plurality of probes having sequences from different genes are used in the alternative fingeφrinting technique. Example 50 below provides a representative alternative fingeφrinting procedure in which the probes are derived from 5 'EST. EXAMPLE 50
Alternative "Fingeφrint" Identification Technique 20-mer oligonucleotides are prepared from a large number, e.g. 50, 100, or 200, of 5'EST using commercially available oligonucleotide services such as Genset, Paris, France. Cell samples from the test subject are processed for DNA using techniques well known to those with skill in the art. The nucleic acid is digested with restriction enzymes such as EcoRI and Xbal. Following digestion, samples are applied to wells for electrophoresis. The procedure, as known in the art, may be modified to accommodate polyacrylamide electrophoresis, however in this example, samples containing 5 ug of DNA are loaded into wells and separated on 0.8% agarose gels. The gels are transferred onto nitrocellulose using standard Southern blotting techniques.
10 ng of each of the oligonucleotides are pooled and end-labeled with j2P. The nitrocellulose is prehybridized with blocking solution and hybridized with the labeled probes. Following hybridization and washing, the nitrocellulose filter is exposed to X-Omat AR X-ray film. The resulting hybridization pattern will be unique for each individual.
It is additionally contemplated within this example that the number of probe sequences used can be varied for additional accuracy or clarity.
The proteins encoded by the extended cDNAs may also be used to generate antibodies as explained in Examples 30 and 43 in order to identify the tissue type or cell species from which a sample is derived as described in example 51.
EXAMPLE 51
Identification of Tissue Types or Cell Species by Means of Labeled Tissue Specific Antibodies
Identification of specific tissues is accomplished by the visualization of tissue specific antigens by means of antibody preparations according to Examples 30 and 43 which are conjugated, directly or indirectly to a detectable marker. Selected labeled antibody species bind to their specific antigen binding partner in tissue sections, cell suspensions, or in extracts of soluble proteins from a tissue sample to provide a pattern for qualitative or semi-qualitative inteφretation. Antisera for these procedures must have a potency exceeding that of the native preparation, and for that reason, antibodies are concentrated to a mg/ml level by isolation of the gamma globulin fraction, for example, by ion-exchange chromatography or by ammonium sulfate fractionation. Also, to provide the most specific antisera, unwanted antibodies, for example to common proteins, must be removed from the gamma globulin fraction, for example by means of insoluble immunoabsorbents, before the antibodies are labeled with the marker. Either monoclonal or heterologous antisera is suitable for either procedure. A. Immunohistochemical techniques Purified, high-titer antibodies, prepared as described above, are conjugated to a detectable marker, as described, for example, by Fudenberg, Chap. 26 in: Basic and Clinical Immunology, 3rd Ed. Lange, Los Altos, California, 1980, or Rose,, et al, Chap. 12 in: Methods in Immunodiagnosis, 2d Ed. John Wiley and Sons, New York (1980), the disclosures of which are incoφorated herein by reference. A fluorescent marker, either fluorescein or rhodamine, is preferred, but antibodies can also be labeled with an enzyme that supports a color producing reaction with a substrate, such as horseradish peroxidase. Markers can be added to tissue-bound antibody in a second step, as described below. Alternatively, the specific antitissue antibodies can be labeled with feπitin or other electron dense particles, and localization of the feπitin coupled antigen-antibody complexes achieved by means of an electron microscope. In yet another approach, the antibodies are radiolabeled, with, for example 125I, and detected by overlaying the antibody treated preparation with photographic emulsion.
Preparations to carry out the procedures can comprise monoclonal or polyclonal antibodies to a single protein or peptide identified as specific to a tissue type, for example, brain tissue, or antibody preparations to several antigenically distinct tissue specific antigens can be used in panels, independently or in mixtures, as required.
Tissue sections and cell suspensions are prepared for immunohistochemical examination according to common histological techniques. Multiple cryostat sections (about 4 μm, unfixed) of the unknown tissue and known control, are mounted and each slide covered with different dilutions of the antibody preparation. Sections of known and unknown tissues should also be treated with preparations to provide a positive control, a negative control, for example, pre-immune sera, and a control for non-specific staining, for example, buffer.
Treated sections are incubated in a humid chamber for 30 min at room temperature, rinsed, then washed in buffer for 30-45 min. Excess fluid is blotted away, and the marker developed.
If the tissue specific antibody was not labeled in the first incubation, it can be labeled at this time in a second antibody-antibody reaction, for example, by adding fluorescein- or enzyme-conjugated antibody against the immunoglobulin class of the antiserum-producing species, for example, fluorescein labeled antibody to mouse IgG. Such labeled sera are commercially available.
The antigen found in the tissues by the above procedure can be quantified by measuring the intensity of color or fluorescence on the tissue section, and calibrating that signal using appropriate standards. B. Identification of tissue specific soluble proteins The visualization of tissue specific proteins and identification of unknown tissues from that procedure is carried out using the labeled antibody reagents and detection strategy as described for immunohistochemistry; however the sample is prepared according to an electrophoretic technique to distribute the proteins extracted from the tissue in an orderly array on the basis of molecular weight for detection. A tissue sample is homogenized using a Virtis apparatus; cell suspensions are disrupted by Dounce homogenization or osmotic lysis, using detergents in either case as required to disrupt cell membranes, as is the practice in the art. Insoluble cell components such as nuclei, microsomes, and membrane fragments are removed by ultracentrifugation, and the soluble protein-containing fraction concentrated if necessary and reserved for analysis. A sample of the soluble protein solution is resolved into individual protein species by conventional SDS polyacrylamide electrophoresis as described, for example, by Davis, et al, Section 19-2 in: Basic Methods in Molecular Biology, Leder ed., Elsevier, New York, 1986, the disclosure of which is incoφorated herein by reference, using a range of amounts of polyacrylamide in a set of gels to resolve the entire molecular weight range of proteins to be detected in the sample. A size marker is run in parallel for puφoses of estimating molecular weights of the constituent proteins. Sample size for analysis is a convenient volume of from 5 to 55 μl, and containing from about 1 to 100 μg protein. An aliquot of each of the resolved proteins is transferred by blotting to a nitrocellulose filter paper, a process that maintains the pattern of resolution. Multiple copies are prepared. The procedure, known as Western Blot Analysis, is well described in Davis, L. et al, supra Section 19-3. One set of nitrocellulose blots is stained with Coomassie blue dye to visualize the entire set of proteins for comparison with the antibody bound proteins. The remaining nitrocellulose filters are then incubated with a solution of one or more specific antisera to tissue specific proteins prepared as described in Examples 30 and 43. In this procedure, as in procedure A above, appropriate positive and negative sample and reagent controls are run. In either procedure A or B, a detectable label can be attached to the primary tissue antigen-primary antibody complex according to various strategies and permutations thereof. In a straightforward approach, the primary specific antibody can be labeled; alternatively, the unlabeled complex can be bound by a labeled secondary anti-IgG antibody. In other approaches, either the primary or secondary antibody is conjugated to a biotin molecule, which can, in a subsequent step, bind an avidin conjugated marker. According to yet another strategy, enzyme labeled or radioactive protein A, which has the property of binding to any IgG, is bound in a final step to either the primary or secondary antibody.
The visualization of tissue specific antigen binding at levels above those seen in control tissues to one or more tissue specific antibodies, prepared from the gene sequences identified from extended cDNA sequences, can identify tissues of unknown origin, for example, forensic samples, or differentiated tumor tissue that has metastasized to foreign bodily sites.
In addition to their applications in forensics and identification, 5' ESTs (or cDNAs or genomic DNAs obtainable therefrom) may be mapped to their chromosomal locations. Example 52 below describes radiation hybrid (RH) mapping of human chromosomal regions using 5 'ESTs. Example 53 below describes a representative procedure for mapping an 5' EST to its location on a human chromosome. Example 54 below describes mapping of 5' ESTs on metaphase chromosomes by Fluorescence In Situ Hybridization (FISH). Those skilled in the art will appreciate that the method of Examples 52-54 may also be used to map cDNAs or genomic DNAs obtainable from the 5' ESTs to their chromosomal locations. 2. Use of 5' ESTs or Sequences Obtainable Therefrom or Portions Thereof in Chromosome Mapping
EXAMPLE 52 Radiation hybrid mapping of 5'ESTs to the human genome Radiation hybrid (RH) mapping is a somatic cell genetic approach that can be used for high resolution mapping of the human genome. In this approach, cell lines containing one or more human chromosomes are lethally irradiated, breaking each chromosome into fragments whose size depends on the radiation dose. These fragments are rescued by fusion with cultured rodent cells, yielding subclones containing different portions of the human genome. This technique is described by Benham et al, Genomics 4:509-517, 1989; and Cox et al, Science 250:245-250, 1990, the entire contents of which are hereby incoφorated by reference. The random and independent nature of the subclones permits efficient mapping of any human genome marker. Human DNA isolated from a panel of 80-100 cell lines provides a mapping reagent for ordering 5'EST. In this approach, the frequency of breakage between markers is used to measure distance, allowing construction of fine resolution maps as has been done using conventional ESTs (Schuler et al, Science 274:540-546, 1996, hereby incoφorated by reference).
RH mapping has been used to generate a high-resolution whole genome radiation hybrid map of human chromosome 17q22-q25.3 across the genes for growth hormone (GH) and thymidine kinase (TK) (Foster et al, Genomics 33:185-192, 1996), the region surrounding the Gorlin syndrome gene (Obermayr et al, Eur. J. Hum. Genet. 4:242-245, 1996), 60 loci covering the entire short arm of chromosome 12 (Raeymaekers et al, Genomics 29:170-178, 1995), the region of human chromosome 22 containing the neurofibromatosis type 2 locus (Frazer et al, Genomics 14:574-584, 1992) and 13 loci on the long arm of chromosome 5 (Warrington etal, Genomics 11:701-708, 1991).
EXAMPLE 53
Mapping of 5'ESTs to HumanChromosomes using PCR techniques 5' ESTs (or cDNAs or genomic DNAs obtainable therefrom) may be assigned to human chromosomes using PCR based methodologies. In such approaches, oligonucleotide primer pairs are designed from the 5' ESTs (or cDNAs or genomic DNAs obtainable therefrom) to minimize the chance of amplifying through an intron. Preferably, the oligonucleotide primers are 18-23 bp in length and are designed for PCR amplification. The creation of PCR primers from known sequences is well known to those with skill in the art. For a review of PCR technology see Erlich in PCR Technology; Principles and Applications or DNA Amplification, Freeman and Co., New York, 1992, the disclosure of which is incoφorated herein by reference.
The primers are used in polymerase chain reactions (PCR) to amplify templates from total human genomic DNA. PCR conditions are as follows: 60 ng of genomic DNA is used as a template for PCR with 80 ng of each oligonucleotide primer, 0.6 unit of Taq polymerase, and 1 μCu of a 32P-labeled deoxycytidine triphosphate. The PCR is performed in a microplate thermocycler (Techne) under the following conditions: 30 cycles of 94°C, 1.4 min; 55°C, 2 min; and 72°C, 2 min; with a final extension at 72°C for 10 min. The amplified products are analyzed on a 6%> polyacrylamide sequencing gel and visualized by autoradiography. If the length of the resulting PCR product is identical to the distance between the ends of the primer sequences in the extended cDNA from which the primers are derived, then the PCR reaction is repeated with DNA templates from two panels of human- rodent somatic cell hybrids, BIOS PCRable DNA (BIOS Coφoration) and NIGMS Human- Rodent Somatic Cell Hybrid Mapping Panel Number 1 (NIGMS, Camden, NJ).
PCR is used to screen a series of somatic cell hybrid cell lines containing defined sets of human chromosomes for the presence of a given 5' EST (or cDNA or genomic DNA obtainable therefrom). DNA is isolated from the somatic hybrids and used as starting templates for PCR_reactions using the primer pairs from the 5' EST (or cDNA or genomic DNA obtainable therefrom). Only those somatic cell hybrids with chromosomes containing the human gene corresponding to the 5' EST (or cDNA or genomic DNA obtainable therefrom) will yield an amplified fragment. The 5' EST (or cDNA or genomic DNA obtainable therefrom) are assigned to a chromosome by analysis of the segregation pattern of PCR products from the somatic hybrid DNA templates. The single human chromosome present in all cell hybrids that give rise to an amplified fragment is the chromosome containing that 5'EST (or cDNA or genomic DNA obtainable therefrom). For a review of techniques and analysis of results from somatic cell gene mapping experiments, see Ledbetter et al, Genomics 6:475-481, 1990, the disclosure of which is incoφorated herein by reference. EXAMPLE 54 Mapping of Extended 5' ESTs to Chromosomes Using Fluorescence In Situ
Hybridization Fluorescence in situ hybridization allows the 5'EST (or cDNA or genomic DNA obtainable therefrom) to be mapped to a particular location on a given chromosome. The chromosomes to be used for fluorescence in situ hybridization techniques may be obtained from a variety of sources including cell cultures, tissues, or whole blood.
In a preferred embodiment, chromosomal localization of an 5'EST (or cDNA or genomic DNA obtainable therefrom) is obtained by FISH as described by Cherif et al. (Proc. Natl. Acad Sci. U.S.A., 87:6639-6643, 1990) , the disclosure of which is incoφorated herein by reference.. Metaphase chromosomes are prepared from phytohemagglutinin (PHA)- stimulated blood cell donors. PHA-stimulated lymphocytes from healthy males are cultured for 72 h in RPMI-1640 medium. For synchronization, methotrexate (10 μM) is added for 17 h, followed by addition of 5-bromodeoxyuridine (5-BrdU, 0.1 mM) for 6 h. Colcemid (1 μg/ml) is added for the last 15 min before harvesting the cells. Cells are collected, washed in RPMI, incubated with a hypotonic solution of KCl (75 mM) at 37°C for 15 min and fixed in three changes of methanol: acetic acid (3:1). The cell suspension is dropped onto a glass slide and air dried. The 5'EST (or cDNA or genomic DNA obtainable therefrom) is labeled with biotin- 16 dUTP by nick translation according to the manufacturer's instructions (Bethesda Research Laboratories, Bethesda, MD), purified using a Sephadex G-50 column (Pharmacia, Upsala, Sweden) and precipitated. Just prior to hybridization, the DNA pellet is dissolved in hybridization buffer (50% formamide, 2 X SSC, 10% dextran sulfate, 1 mg/ml sonicated salmon sperm DNA, pH 7) and the probe is denatured at 70°C for 5-10 min.
Slides kept at -20°C are treated for 1 h at 37°C with RNase A (100 μg/ml), rinsed three times in 2 X SSC and dehydrated in an ethanol series. Chromosome preparations are denatured in 70% formamide, 2 X SSC for 2 min at 70°C, then dehydrated at 4°C. The slides are treated with proteinase K (10 μg/100 ml in 20 mM Tris-HCl, 2 mM CaCl2) at 37°C for 8 min and dehydrated. The hybridization mixture containing the probe is placed on the slide, covered with a coverslip, sealed with rubber cement and incubated overnight in a humid chamber at 37°C. After hybridization and post-hybridization washes, the biotinylated probe is detected by avidin-FITC and amplified with additional layers of biotinylated goat anti-avidin and avidin-FITC. For chromosomal localization, fluorescent R-bands are obtained as previously described (Cherif et al, supra.). The slides are observed under a LEICA fluorescence microscope (DMRXA). Chromosomes are counterstained with propidium iodide and the fluorescent signal of the probe appears as two symmetrical yellow-green spots on both chromatids of the fluorescent R-band chromosome (red). Thus, a particular 5'EST (or cDNA or genomic DNA obtainable therefrom) may be localized to a particular cytogenetic R-band on a given chromosome.
Once the 5'EST (or cDNA or genomic DNA obtainable therefrom) have been assigned to particular chromosomes using the techniques described in Examples 52-54 above, they may be utilized to construct a high resolution map of the chromosomes on which they are located or to identify the chromosomes in a sample.
EXAMPLE 55 Use of 5'EST to Construct or Expand Chromosome Maps
Chromosome mapping involves assigning a given unique sequence to a particular chromosome as described above. Once the unique sequence has been mapped to a given chromosome, it is ordered relative to other unique sequences located on the same chromosome. One approach to chromosome mapping utilizes a series of yeast artificial chromosomes (YACs) bearing several thousand long inserts derived from the chromosomes of the organism from which the extended cDNAs (or genomic DNAs obtainable therefrom) are obtained. This approach is described in Nagaraja et al, Genome Research 7:210-222, 1997, the disclosure of which is incoφorated herein by reference. Briefly, in this approach each chromosome is broken into overlapping pieces which are inserted into the YAC vector. The YAC inserts are screened using PCR or other methods to determine whether they include the 5'EST (or cDNA or genomic DNA obtainable therefrom) whose position is to be determined. Once an insert has been found which includes the 5'EST (or cDNA or genomic DNA obtainable therefrom), the insert can be analyzed by PCR or other methods to determine whether the insert also contains other sequences known to be on the chromosome or in the region from which the 5'EST (or cDNA or genomic DNA obtainable therefrom) was derived. This process can be repeated for each insert in the YAC library to determine the location of each of the extended cDNAs (or genomic DNAs obtainable therefrom) relative to one another and to other known chromosomal markers. In this way, a high resolution map of the distribution of numerous unique markers along each of the organisms chromosomes may be obtained.
As described in Example 56 below extended cDNAs (or genomic DNAs obtainable therefrom) may also be used to identify genes associated with a particular phenotype, such as hereditary disease or drug response.
3. Use of 5ΕSTs or Sequences Obtained Therefrom or Fragments Thereof in Gene Identification
EXAMPLE 56 Identification of genes associated with hereditary diseases or drug response This example illustrates an approach useful for the association of 5'ESTs (or cDNA or genomic DNA obtainable therefrom) with particular phenotypic characteristics. In this example, a particular 5'EST (or cDNA or genomic DNA obtainable therefrom) is used as a test probe to associate that 5'EST (or cDNA or genomic DNA obtainable therefrom) with a particular phenotypic characteristic.
5'ESTs (or cDNA or genomic DNA obtainable therefrom) are mapped to a particular location on a human chromosome using techniques such as those described in Examples 52 and 53 or other techniques known in the art. A search of Mendelian Inheritance in Man (McKusick in Mendelian Inheritance in Man (available on line through Johns Hopkins University Welch Medical Library) reveals the region of the human chromosome which contains the 5'EST (or cDNA or genomic DNA obtainable therefrom) to be a very gene rich region containing several known genes and several diseases or phenotypes for which genes have not been identified. The gene corresponding to this 5'EST (or cDNA or genomic DNA obtainable therefrom) thus becomes an immediate candidate for each of these genetic diseases.
Cells from patients with these diseases or phenotypes are isolated and expanded in culture. PCR primers from the 5'EST (or cDNA or genomic DNA obtainable therefrom) are used to screen genomic DNA, mRNA or cDNA obtained from the patients. 5'ESTs (or cDNA or genomic DNA obtainable therefrom) that are not amplified in the patients can be positively associated with a particular disease by further analysis. Alternatively, the PCR analysis may yield fragments of different lengths when the samples are derived from an individual having the phenotype associated with the disease than when the sample is derived from a healthy individual, indicating that the gene containing the 5'EST may be responsible for the genetic disease.
VL Use of 5'EST (or cDNA or Genomic DNA Obtainable Therefrom) to Construct Vectors The present 5'ESTs (or cDNA or genomic DNA obtainable therefrom) may also be used to construct secretion vectors capable of directing the secretion of the proteins encoded by genes therein. Such secretion vectors may facilitate the purification or enrichment of the proteins encoded by genes inserted therein by reducing the number of background proteins from which the desired protein must be purified or enriched. Exemplary secretion vectors are described in Example 57 below.
1. Construction of Secretion Vectors
EXAMPLE 57
Construction of Secretion Vectors The secretion vectors include a promoter capable of directing gene expression in the host cell, tissue, or organism of interest. Such promoters include the Rous Sarcoma Virus promoter, the SV40 promoter, the human cytomegalovirus promoter, and other promoters familiar to those skilled in the art.
A signal sequence from a 5' EST (or cDNAs or genomic DNAs obtainable therefrom) is operably linked to the promoter such that the mRNA transcribed from the promoter will direct the translation of the signal peptide. The host cell, tissue, or organism may be any cell, tissue, or organism which recognizes the signal peptide encoded by the signal sequence in the 5' EST (or cDNA or genomic DNA obtainable therefrom). Suitable hosts include mammalian cells, tissues or organisms, avian cells, tissues, or organisms, insect cells, tissues or organisms, or yeast. In addition, the secretion vector contains cloning sites for inserting genes encoding the proteins which are to be secreted. The cloning sites facilitate the cloning of the insert gene in frame with the signal sequence such that a fusion protein in which the signal peptide is fused to the protein encoded by the inserted gene is expressed from the mRNA transcribed from the promoter. The signal peptide directs the extracellular secretion of the fusion protein.
The secretion vector may be DNA or RNA and may integrate into the chromosome of the host, be stably maintained as an extrachromosomal replicon in the host, be an artificial chromosome, or be transiently present in the host. Many nucleic acid backbones suitable for use as secretion vectors are known to those skilled in the art, including retroviral vectors, SV40 vectors, Bovine Papilloma Virus vectors, yeast integrating plasmids, yeast episomal plasmids, yeast artificial chromosomes, human artificial chromosomes, P element vectors, baculovirus vectors, or bacterial plasmids capable of being transiently introduced into the host.
The secretion vector may also contain a polyA signal such that the polyA signal is located downstream of the gene inserted into the secretion vector.
After the gene encoding the protein for which secretion is desired is inserted into the secretion vector, the secretion vector is introduced into the host cell, tissue, or organism using calcium phosphate precipitation, DEAE-Dextran, electroporation, liposome-mediated transfection, viral particles or as naked DNA. The protein encoded by the inserted gene is then purified or enriched from the supernatant using conventional techniques such as ammonium sulfate precipitation, immunoprecipitation, immunochromatography, size exclusion chromatography, ion exchange chromatography, and HPLC. Alternatively, the secreted protein may be in a sufficiently enriched or pure state in the supernatant or growth media of the host to permit it to be used for its intended puφose without further enrichment. The signal sequences may also be inserted into vectors designed for gene therapy. In such vectors, the signal sequence is operably linked to a promoter such that mRNA transcribed from the promoter encodes the signal peptide. A cloning site is located downstream of the signal sequence such that a gene encoding a protein whose secretion is desired may readily be inserted into the vector and fused to the signal sequence. The vector is introduced into an appropriate host cell. The protein expressed from the promoter is secreted extracellularly, thereby producing a therapeutic effect. The 5' ESTs may also be used to clone sequences located upstream of the 5' ESTs which are capable of regulating gene expression, including promoter sequences, enhancer sequences, and other upstream sequences which influence transcription or translation levels. Once identified and cloned, these upstream regulatory sequences may be used in expression vectors designed to direct the expression of an inserted gene in a desired spatial, temporal, developmental, or quantitative fashion. Example 58 describes a method for cloning sequences upstream of the extended cDNAs or 5' ESTs.
2. Identification of Upstream Sequences With Promoting or Regulatory Activities EXAMPLE 58
Use of Extended cDNAs or 5' ESTs to Clone Upstream Sequences from Genomic DNA
Sequences derived from extended cDNAs or 5' ESTs may be used to isolate the promoters of the corresponding genes using chromosome walking techniques. In one chromosome walking technique, which utilizes the GenomeWalker™ kit available from Clontech, five complete genomic DNA samples are each digested with a different restriction enzyme which has a 6 base recognition site and leaves a blunt end. Following digestion, oligonucleotide adapters are ligated to each end of the resulting genomic DNA fragments.
For each of the five genomic DNA libraries, a first PCR reaction is performed according to the manufacturer's instructions (which are incoφorated herein by reference) using an outer adaptor primer provided in the kit and an outer gene specific primer. The gene specific primer should be selected to be specific for the extended cDNA or 5' EST of interest and should have a melting temperature, length, and location in the extended cDNA or 5'EST which is consistent with its use in PCR reactions. Each first PCR reaction contains 5 ng of genomic DNA, 5 μl of 10X Tth reaction buffer, 0.2 mM of each dNTP, 0.2 μM each of outer adaptor primer and outer gene specific primer, 1.1 mM of Mg(OAc)2, and 1 μl of the Tth polymerase 50X mix in a total volume of 50 μl. The reaction cycle for the first PCR reaction is as follows: 1 min - 94°C / 2 sec - 94°C, 3 min - 72°C (7 cycles) / 2 sec - 94°C, 3 min - 67°C (32 cycles) / 5 min - 67°C.
The product of the first PCR reaction is diluted and used as a template for a second PCR reaction according to the manufacturer's instructions using a pair of nested primers which are located internally on the amplicon resulting from the first PCR reaction. «For example, 5 μl of the reaction product of the first PCR reaction mixture may be diluted 180 times. Reactions are made in a 50 μl volume having a composition identical to that of the first PCR reaction except the nested primers are used. The first nested primer is specific for the adaptor, and is provided with the GenomeWalker™ kit. The second nested primer is specific for the particular extended cDNA or 5' EST for which the promoter is to be cloned and should have a melting temperature, length, and location in the extended cDNA or 5' EST which is consistent with its use in PCR reactions. The reaction parameters of the second PCR reaction are as follows: 1 min - 94°C / 2 sec - 94°C, 3 min - 72°C (6 cycles) / 2 sec - 94°C, 3 min - 67°C (25 cycles) / 5 min - 67°C. The product of the second PCR reaction is purified, cloned, and sequenced using standard techniques.
Alternatively, two or more human genomic DNA libraries can be constructed by using two or more restriction enzymes. The digested genomic DNA is cloned into vectors which can be converted into single stranded, circular, or linear DNA. A biotinylated oligonucleotide comprising at least 15 nucleotides from the extended cDNA or 5' EST sequence is hybridized to the single stranded DNA. Hybrids between the biotinylated oligonucleotide and the single stranded DNA containing the extended cDNA or EST sequence are isolated as described in Example 29 above. Thereafter, the single stranded DNA containing the extended cDNA or EST sequence is released from the beads and converted into double stranded DNA using a primer specific for the extended cDNA or 5' EST sequence or a primer corresponding to a sequence included in the cloning vector. The resulting double stranded DNA is transformed into bacteria. DNAs containing the 5' EST or extended cDNA sequences are identified by colony PCR or colony hybridization.
Once the upstream genomic sequences have been cloned and sequenced as described above, prospective promoters and transcription start sites within the upstream sequences may be identified by comparing the sequences upstream of the extended cDNAs or 5' ESTs with databases containing known transcription start sites, transcription factor binding sites, or promoter sequences. In addition, promoters in the upstream sequences may be identified using promoter reporter vectors as described in Example . EXAMPLE 59
Identification of Promoters in Cloned Upstream Sequences The genomic sequences upstream of the extended cDNAs or 5' ESTs are cloned into a suitable promoter reporter vector, such as the pSEAP-Basic, pSEAP-Enhancer, pβgal- Basic, pβgal-Enhancer, or pEGFP-1 Promoter Reporter vectors available from Clontech. Briefly, each of these promoter reporter vectors include multiple cloning sites positioned upstream of a reporter gene encoding a readily assayable protein such as secreted alkaline phosphatase, β galactosidase, or green fluorescent protein. The sequences upstream of the extended cDNAs or 5' ESTs are inserted into the cloning sites upstream of the reporter gene in both orientations and introduced into an appropriate host cell. The level of reporter protein is assayed and compared to the level obtained from a vector which lacks an insert in the cloning site. The presence of an elevated expression level in the vector containing the insert with respect to the control vector indicates the presence of a promoter in the insert. If necessary, the upstream sequences can be cloned into vectors which contain an enhancer for augmenting transcription levels from weak promoter sequences. A significant level of expression above that observed with the vector lacking an insert indicates that a promoter sequence is present in the inserted upstream sequence.
Appropriate host cells for the promoter reporter vectors may be chosen based on the results of the above described determination of expression patterns of the extended cDNAs and ESTs. For example, if the expression pattern analysis indicates that the mRNA corresponding to a particular extended cDNA or 5' EST is expressed in fibroblasts, the promoter reporter vector may be introduced into a human fibroblast cell line.
Promoter sequences within the upstream genomic DNA may be further defined by constructing nested deletions in the upstream DNA using conventional techniques such as Exonuclease HI digestion. The resulting deletion fragments can be inserted into the promoter reporter vector to determine whether the deletion has reduced or obliterated promoter activity. In this way, the boundaries of the promoters may be defined. If desired, potential individual regulatory sites within the promoter may be identified using site directed mutagenesis or linker scanning to obliterate potential transcription factor binding sites within the promoter individually or in combination. The effects of these mutations on transcription levels may be determined by inserting the mutations into the cloning sites in the promoter reporter vectors.
EXAMPLE 60 Cloning and Identification of Promoters
Using the method described in Example 58 above with 5' ESTs, sequences upstream of several genes were obtained. Using the primer pairs GGG AAG ATG GAG ATA GTA
TTG CCT G (SEQ ID NO:29) and CTG CCA TGT ACA TGA TAG AGA GAT TC (SEQ
ID NO:30), the promoter having the internal designation P13H2 (SEQ ID NO:31) was obtained.
Using the primer pairs GTA CCA GGGG ACT GTG ACC ATT GC (SEQ ID NO:32) and CTG TGA CCA TTG CTC CCA AGA GAG (SEQ ID NO:33), the promoter having the internal designation P15B4 (SEQ ID NO:34) was obtained.
Using the primer pairs CTG GGA TGG AAG GCA CGG TA (SEQ ID NO:35) and GAG ACC ACA CAG CTA GAC AA (SEQ ID NO:36), the promoter having the internal designation P29B6 (SEQ ID NO:37) was obtained.
Figure 4 provides a schematic description of the promoters isolated and the way they are assembled with the corresponding 5' tags. The upstream sequences were screened for the presence of motifs resembling transcription factor binding sites or known transcription start sites using the computer program Matlnspector release 2.0, August 1996.
Table VII describes the transcription factor binding sites present in each of these promoters. The columns labeled matrice provides the name of the Matlnspector matrix used. The column labeled position provides the 5' position of the promoter site. Numeration of the sequence starts from the transcription site as determined by matching the genomic sequence with the 5' EST sequence. The column labeled "orientation" indicates the DNA strand on which the site is found, with the + strand being the coding strand as determined by matching the genomic sequence with the sequence of the 5' EST. The column labeled "score" provides the Matlnspector score found for this site. The column labeled "length" provides the length of the site in nucleotides. The column labeled "sequence" provides the sequence of the site found. Bacterial clones containing plasmids containing the promoter sequences described above described above are presently stored in the inventor's laboratories under the internal identification numbers provided above. The inserts may be recovered from the deposited materials by growing an aliquot of the appropriate bacterial clone in the appropriate medium. The plasmid DNA can then be isolated using plasmid isolation procedures familiar to those skilled in the art such as alkaline lysis minipreps or large scale alkaline lysis plasmid isolation procedures. If desired the plasmid DNA may be further enriched by centrUugation on a cesium chloride gradient, size exclusion chromatography, or anion exchange chromatography. The plasmid DNA obtained using these procedures may then be manipulated using standard cloning techniques familiar to those skilled in the art. Alternatively, a PCR can be done with primers designed at both ends of the EST insertion. The PCR product which corresponds to the 5' EST can then be manipulated using standard cloning techniques familiar to those skilled in the art.
The promoters and other regulatory sequences located upstream of the extended cDNAs or 5' ESTs may be used to design expression vectors capable of directing the expression of an inserted gene in a desired spatial, temporal, developmental, or quantitative manner. A promoter capable of directing the desired spatial, temporal, developmental, and quantitative patterns may be selected using the results of the expression analysis described in Example 26 above. For example, if a promoter which confers a high level of expression in muscle is desired, the promoter sequence upstream of an extended cDNA or 5' EST derived from an mRNA which is expressed at a high level in muscle, as determined by the method of Example 26, may be used in the expression vector.
Preferably, the desired promoter is placed near multiple restriction sites to facilitate the cloning of the desired insert downstream of the promoter, such that the promoter is able to drive expression of the inserted gene. The promoter may be inserted in conventional nucleic acid backbones designed for extrachromosomal replication, integration into the host chromosomes or transient expression. Suitable backbones for the present expression vectors include retroviral backbones, backbones from eukaryotic episomes such as SV40 or Bovine Papilloma Virus, backbones from bacterial episomes, or artificial chromosomes. Preferably, the expression vectors also include a polyA signal downstream of the multiple restriction sites for directing the polyadenylation of mRNA transcribed from the gene inserted into the expression vector.
Following the identification of promoter sequences using the procedures of Examples 58-60, proteins which interact with the promoter may be identified as described in Example 61 below.
EXAMPLE 61
Identification of Proteins Which Interact with Promoter Sequences, Upstream Regulatory Sequences, or mRNA
Sequences within the promoter region which are likely to bind transcription factors may be identified by homology to known transcription factor binding sites or through conventional mutagenesis or deletion analyses of reporter plasmids containing the promoter sequence. For example, deletions may be made in a reporter plasmid containing the promoter sequence of interest operably linked to an assayable reporter gene. The reporter plasmids carrying various deletions within the promoter region are transfected into an appropriate host cell and the effects of the deletions on expression levels is assessed. Transcription factor binding sites within the regions in which deletions reduce expression levels may be further localized using site directed mutagenesis, linker scanning analysis, or other techniques familiar to those skilled in the art.
Nucleic acids encoding proteins which interact with sequences in the promoter may be identified using one-hybrid systems such as those described in the manual accompanying the Matchmaker One-Hybrid System kit available from Clontech (Catalog No. K1603-1), the disclosure of which is incoφorated herein by reference. Briefly, the Matchmaker One-hybrid system is used as follows. The target sequence for which it is desired to identify binding proteins is cloned upstream of a selectable reporter gene and integrated into the yeast genome. Preferably, multiple copies of the target sequences are inserted into the reporter plasmid in tandem. A library comprised of fusions between cDNAs to be evaluated for the ability to bind to the promoter and the activation domain of a yeast transcription factor, such as GAL4, is transformed into the yeast strain containing the integrated reporter sequence. The yeast are plated on selective media to select cells expressing the selectable marker linked to the promoter sequence. The colonies which grow on the selective media contain genes encoding proteins which bind the target sequence. The inserts in the genes encoding the fusion proteins are further characterized by sequencing. In addition, the inserts may be inserted into expression vectors or in vitro transcription vectors. Binding of the polypeptides encoded by the inserts to the promoter DNA may be confirmed by techniques familiar to those skilled in the art, such as gel shift analysis or DNAse protection analysis.
YTL Use of 5' ESTs (or cDNAs or Genomic DNAs Obtainable Therefrom) in Gene Therapy
The present invention also comprises the use of 5'ESTs (or cDNA or genomic DNA obtainable therefrom) in gene therapy strategies, including antisense and triple helix strategies as described in Examples 62 and 63 below. In antisense approaches, nucleic acid sequences complementary to an mRNA are hybridized to the mRNA intracellularly, thereby blocking the expression of the protein encoded by the mRNA. The antisense sequences may prevent gene expression through a variety of mechanisms. For example, the antisense sequences may inhibit the ability of ribosomes to translate the mRNA. Alternatively, the antisense sequences may block transport of the mRNA from the nucleus to the cytoplasm, thereby limiting the amount of mRNA available for translation. Another mechanism through which antisense sequences may inhibit gene expression is by interfering with mRNA splicing. In yet another strategy, the antisense nucleic acid may be incoφorated in a ribozyme capable of specifically cleaving the target mRNA.
EXAMPLE 62 Preparation and Use of Antisense Oligonucleotides
The antisense nucleic acid molecules to be used in gene therapy may be either DNA or RNA sequences. They may comprise a sequence complementary to the sequence of the 5'EST (or cDNA or genomic DNA obtainable therefrom). The antisense nucleic acids should have a length and melting temperature sufficient to permit formation of an intracellular duplex with sufficient stability to inhibit the expression of the mRNA in the duplex. Strategies for designing antisense nucleic acids suitable for use in gene therapy are disclosed in Green et al, Ann. Rev. Biochem. 55:569-597, 1986; and Izant and Weintraub, Cell 36:1007-1015, 1984, which are hereby incoφorated by reference.
In some strategies, antisense molecules are obtained from a nucleotide sequence encoding a protein by reversing the orientation of the coding region with respect to a promoter so as to transcribe the opposite strand from that which is normally transcribed in the cell. The antisense molecules may be transcribed using in vitro transcription systems such as those which employ T7 or SP6 polymerase to generate the transcript. Another approach involves transcription of the antisense nucleic acids in vivo by operably linking DNA containing the antisense sequence to a promoter in an expression vector. Alternatively, oligonucleotides which are complementary to the strand normally transcribed in the cell may be synthesized in vitro. Thus, the antisense nucleic acids are complementary to the corresponding mRNA and are capable of hybridizing to the mRNA to create a duplex. In some embodiments, the antisense sequences may contain modified sugar phosphate backbones to increase stability and make them less sensitive to RNase activity. Examples of modifications suitable for use in antisense strategies are described by Rossi et al, Pharmacol. Ther. 50(2):245-254, 1991, which is hereby incoφorated by reference.
Various types of antisense oligonucleotides complementary to the sequence of the 5'EST (or cDNA or genomic DNA obtainable therefrom) may be used. In one preferred embodiment, stable and semi-stable antisense oligonucleotides described in International Application No. PCT WO94/23026, hereby incoφorated by reference, are used. In these molecules, the 3' end or both the 3' and 5' ends are engaged in intramolecular hydrogen bonding between complementary base pairs. These molecules are better able to withstand exonuclease attacks and exhibit increased stability compared to conventional antisense oligonucleotides. In another preferred embodiment, the antisense oligodeoxynucleotides against heφes simplex virus types 1 and 2 described in International Application No. WO 95/04141, hereby incoφorated by reference, are used.
Ln yet another preferred embodiment, the covalently cross-linked antisense oligonucleotides described in International Application No. WO 96/31523, hereby incoφorated by reference, are used. These double- or single-stranded oligonucleotides comprise one or more, respectively, inter- or intra-oligonucleotide covalent cross-linkages, wherein the linkage consists of an amide bond between a primary amine group of one strand and a carboxyl group of the other strand or of the same strand, respectively, the primary amine group being directly substituted in the 2' position of the strand nucleotide monosaccharide ring, and the carboxyl group being carried by an aliphatic spacer group substituted on a nucleotide or nucleotide analog of the other strand or the same strand, respectively.
The antisense oligodeoxynucleotides and oligonucleotides disclosed in International Application No. WO 92/18522, incoφorated by reference, may also be used. These molecules are stable to degradation and contain at least one transcription control recognition sequence which binds to control proteins and are effective as decoys therefore. These molecules may contain "hairpin" structures, "dumbbell" structures, "modified dumbbell" structures, "cross-linked" decoy structures and "loop" structures.
In another preferred embodiment, the cyclic double-stranded oligonucleotides described in European Patent Application No. 0 572 287 A2, hereby incoφorated by reference are used. These ligated oligonucleotide "dumbbells" contain the binding site for a transcription factor and inhibit expression of the gene under control of the transcription factor by sequestering the factor.
Use of the closed antisense oligonucleotides disclosed in International Application No. WO 92/19732, hereby incoφorated by reference, is also contemplated. Because these molecules have no free ends, they are more resistant to degradation by exonucleases than are conventional oligonucleotides. These oligonucleotides may be multifunctional, interacting with several regions which are not adjacent to the target mRNA.
The appropriate level of antisense nucleic acids required to inhibit gene expression may be determined using in vitro expression analysis. The antisense molecule may be introduced into the cells by diffusion, injection, infection, transfection or h-region-mediated import using procedures known in the art. For example, the antisense nucleic acids can be introduced into the body as a bare or naked oligonucleotide, oligonucleotide encapsulated in lipid, oligonucleotide sequence encapsidated by viral protein, or as an oligonucleotide operably linked to a promoter contained in an expression vector. The expression vector may be any of a variety of expression vectors known in the art, including retroviral or viral vectors, vectors capable of extrachromosomal replication, or integrating vectors The vectors may be DNA or RNA.
The antisense molecules are introduced onto cell samples at a number of different concentrations preferably between lxlO"10M to lxlO^M. Once the minimum concentration that can adequately control gene expression is identified, the optimized dose is translated into a dosage suitable for use in vivo. For example, an inhibiting concentration in culture of lxlO"7 translates into a dose of approximately 0 6 mg/kg bodyweight. Levels of oligonucleotide approaching 100 mg/kg bodyweight or higher may be possible after testing the toxicity of the oligonucleotide in laboratory animals It is additionally contemplated that cells from the vertebrate are removed, treated with the antisense oligonucleotide, and reintroduced into the vertebrate.
It is further contemplated that the antisense oligonucleotide sequence is incoφorated into a ribozyme sequence to enable the antisense to specifically bind and cleave its target mRNA. For technical applications of ribozyme and antisense oligonucleotides see Rossi et al, supra.
Ln a preferred application of this invention, the polypeptide encoded by the gene is first identified, so that the effectiveness of antisense inhibition on translation can be monitored using techniques that include but are not limited to antibody-mediated tests such as RIAs and ELISA, functional assays, or radiolabeling The 5' ESTs of the present invention (or cDNAs or genomic DNAs obtainable therefrom) may also be used in gene therapy approaches based on intracellular triple helix formation. Triple helix oligonucleotides are used to inhibit transcription from a genome. They are particularly useful for studying alterations in cell activity as it is associated with a particular gene. The 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom) of the present invention or, more preferably, a portion of those sequences, can be used to inhibit gene expression in individuals having diseases associated with expression of a particular gene. Similarly, a portion of 5' EST sequences (or cDNAs or genomic DNAs obtainable therefrom) can be used to study the effect of inhibiting transcription of a particular gene within a cell. Traditionally, homopurine sequences were considered the most useful for triple helix strategies. However, homopyrimidine sequences can also inhibit gene expression Such homopyrimidine oligonucleotides bind to the major groove at homopurine:homopyrimidine sequences. Thus, both types of sequences from the 5'EST or from the gene corresponding to the 5'EST are contemplated within the scope of this invention.
EXAMPLE 63
Preparation and Use of Triple Helix Probes The sequences of the 5' ESTs (or cDNAs or genomic DNAs obtainable therefrom) are scanned to identify 10-mer to 20-mer homopyrimidine or homopurine stretches which could be used in triple-helix based strategies for inhibiting gene expression. Following identification of candidate homopyrimidine or homopurine stretches, their efficiency in inhibiting gene expression is assessed by introducing varying amounts of oligonucleotides containing the candidate sequences into tissue culture cells which normally express the target gene. The oligonucleotides may be prepared on an oligonucleotide synthesizer or they may be purchased commercially from a company specializing in custom oligonucleotide synthesis, such as GENSET, Paris, France.
The oligonucleotides may be introduced into the cells using a variety of methods known to those skilled in the art, including but not limited to calcium phosphate precipitation, DEAE-Dextran, electroporation, liposome-mediated transfection or native uptake.
Treated cells are monitored for altered cell function or reduced gene expression using techniques such as Northern blotting, RNase protection assays, or PCR based strategies to monitor the transcription levels of the target gene in cells which have been treated with the oligonucleotide. The cell functions to be monitored are predicted based upon the homologies of the target gene corresponding to the extended cDNA from which the oligonucleotide was derived with known gene sequences that have been associated with a particular function. The cell functions can also be predicted based on the presence of abnormal physiologies within cells derived from individuals with a particular inherited disease, particularly when the extended cDNA is associated with the disease using techniques described in Example 56.
The oligonucleotides which are effective in inhibiting gene expression in tissue culture cells may then be introduced in vivo using the techniques described above and in Example 62 at a dosage calculated based on the in vitro results, as described in Example 62. In some embodiments, the natural (beta) anomers of the oligonucleotide units can be replaced with alpha anomers to render the oligonucleotide more resistant to nucleases. Further, an intercalating agent such as ethidium bromide, or the like, can be attached to the 3' end of the alpha oligonucleotide to stabilize the triple helix. For information on the generation of oligonucleotides suitable for triple helix formation see Griffin et al, Science 245:967-971, 1989, which is hereby incoφorated by this reference.
EXAMPLE 64
Use of cDNAs Obtained Using the 5' ESTs to Express an Encoded Protein in a Host Organism
The cDNAs obtained as described above using the 5' ESTs of the present invention may also be used to express an encoded protein in a host organism to produce a beneficial effect. In such procedures, the encoded protein may be transiently expressed in the host organism or stably expressed in the host organism. The encoded protein may have any of the activities described above. The encoded protein may be a protein which the host organism lacks or, alternatively, the encoded protein may augment the existing levels of the protein in the host organism.
A full length extended cDNA encoding the signal peptide and the mature protein, or an extended cDNA encoding only the mature protein is introduced into the host organism. The extended cDNA may be introduced into the host organism using a variety of techniques known to those of skill in the art. For example, the extended cDNA may be injected into the host organism as naked DNA such that the encoded protein is expressed in the host organism, thereby producing a beneficial effect.
Alternatively, the extended cDNA may be cloned into an expression vector downstream of a promoter which is active in the host organism. The expression vector may be any of the expression vectors designed for use in gene therapy, including viral or retroviral vectors. The expression vector may be directly introduced into the host organism such that the encoded protein is expressed in the host organism to produce a beneficial effect. In another approach, the expression vector may be introduced into cells in vitro. Cells containing the expression vector are thereafter selected and introduced into the host organism, where they express the encoded protein to produce a beneficial effect. EXAMPLE 65
Use of Signal Peptides Encoded by 5' ESTs or Sequences obtained Therefrom to Import Proteins Into Cells
The short core hydrophobic region (h) of signal peptides encoded by the 5'ESTS or extended cDNAs derived from SEQ ID NOs: 38-305 may also be used as a carrier to import a peptide or a protein of interest, so-called cargo, into tissue culture cells (Lin et al, J. Biol.
Chem., 270: 14225-14258, 1995; Du et al, J. Peptide Res, 51: 235-243, 1998; Rojas et al,
Nature Biotech., 16: 370-375, 1998).
When cell permeable peptides of limited size (approximately up to 25 amino acids) are to be translocated across cell membrane, chemical synthesis may be used in order to add the h region to either the C-terminus or the N-terminus to the cargo peptide of interest. Alternatively, when longer peptides or proteins are to be imported into cells, nucleic acids can be genetically engineered, using techniques familiar to those skilled in the art, in order to link the extended cDNA sequence encoding the h region to the 5' or the 3' end of a DNA sequence coding for a cargo polypeptide. Such genetically engineered nucleic acids are then translated either in vitro or in vivo after transfection into appropriate cells, using conventional techniques to produce the resulting cell permeable polypeptide. Suitable hosts cells are then simply incubated with the cell permeable polypeptide which is then translocated across the membrane. This method may be applied to study diverse intracellular functions and cellular processes. For instance, it has been used to probe functionally relevant domains of intracellular proteins and to examine protein-protein interactions involved in signal transduction pathways (Lin et al, supra; Lin et al, J. Biol. Chem., 271: 5305-5308, 1996; Rojas et al, Biol Chem., 271: 27456-27461, 1996; Liu et al, Proc. Natl. Acad Sci. USA, 93: 11819-11824, 1996; Rojas et al, Bioch. Biophys. Res. Commun., 234: 675-680, 1997). Such techniques may be used in cellular therapy to import proteins producing therapeutic effects. For instance, cells isolated from a patient may be treated with imported therapeutic proteins and then re-introduced into the host organism.
Alternatively, the h region of signal peptides of the present invention could be used in combination with a nuclear localization signal to deliver nucleic acids into cell nucleus. Such oligonucleotides may be antisense oligonucleotides or oligonucleotides designed to form triple helixes, as described in examples 62 and 63 respectively, in order to inhibit processing and/or maturation of a target cellular RNA.
As discussed above, the cDNAs or portions thereof obtained using the 5' ESTs of the present invention can be used for various puφoses. The polynucleotides can be used to express recombinant protein for analysis, characterization or therapeutic use; as markers for tissues in which the corresponding protein is preferentially expressed (either constitutively or at a particular stage of tissue differentiation or development or in disease states); as molecular weight markers on Southern gels; as chromosome markers or tags (when labeled) to identify chromosomes or to map related gene positions; to compare with endogenous DNA sequences in patients to identify potential genetic disorders; as probes to hybridize and thus discover novel, related DNA sequences; as a source of information to derive PCR primers for genetic fingeφrinting; for selecting and making oligomers for attachment to a "gene chip" or other support, including for examination for expression patterns; to raise anti-protein antibodies using DNA immunization techniques; and as an antigen to raise anti-DNA antibodies or elicit another immune response. Where the polynucleotide encodes a protein which binds or potentially binds to another protein (such as, for example, in a receptor-ligand interaction), the polynucleotide can also be used in interaction trap assays (such as, for example, that described in Gyuris et al, Cell 75:791-803, 1993, the disclosure of which is hereby incoφorated by reference) to identify polynucleotides encoding the other protein with which binding occurs or to identify inhibitors of the binding interaction.
The proteins or polypeptides provided by the present invention can similarly be used in assays to determine biological activity, including in a panel of multiple proteins for high- throughput screening; to raise antibodies or to elicit another immune response; as a reagent (including the labeled reagent) in assays designed to quantitatively determine levels of the protein (or its receptor) in biological fluids; as markers for tissues in which the corresponding protein is preferentially expressed (either constitutively or at a particular stage of tissue differentiation or development or in a disease state); and, of course, to isolate correlative receptors or ligands. Where the protein binds or potentially binds to another protein (such as, for example, in a receptor-ligand interaction), the protein can be used to identify the other protein with which binding occurs or to identify inhibitors of the binding interaction. Proteins involved in these binding interactions can also be used to screen for peptide or small molecule inhibitors or agonists of the binding interaction.
Any or all of these research utilities are capable of being developed into reagent grade or kit format for commercialization as research products. Methods for performing the uses listed above are well known to those skilled in the art. References disclosing such methods include without limitation Molecular Cloning; A
Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, Sambrook,. Fritsch and
Maniatis eds., 1989, and Methods in Enzymology; Guide to Molecular Cloning Techniques,
Academic Press, Berger and Kimmel eds., 1987. Polynucleotides and proteins of the present invention can also be used as nutritional sources or supplements. Such uses include without limitation use as a protein or amino acid supplement, use as a carbon source, use as a nitrogen source and use as a source of carbohydrate. In such cases the protein or polynucleotide of the invention can be added to the feed of a particular organism or can be administered as a separate solid or liquid preparation, such as in the form of powder, pills, solutions, suspensions or capsules. In the case of microorganisms, the protein or polynucleotide of the invention can be added to the medium in or on which the microorganism is cultured.
Although this invention has been described in terms of certain preferred embodiments, other embodiments which will be apparent to those of ordinary skill in the art in view of the disclosure herein are also Vvithin the scope of this invention. Accordingly, the scope of the invention is intended to be defined only by reference to the appended claims. All documents cited herein are incoφorated herein by reference in their entirety.
Table 1 : Parameters used for each step of EST analysis
* use "Quick Fast" Database scanner f alignement further constrained to begin closer than 10bp to EST 5' end π using BLOSUM62 substitution matrix
TABLE D
SEQ. ID VON HEIJNE TISSUE INTERNAL
NO. CATEGORY SCORE SOURCE DESIGNATION
ID38 new 15.8 Heart 25-13-1-HlO-PU
ID39 new 14 Fetal kidney 58-47-2-B11-PU
ID40 new 12.3 Dystrophic muscle 29-3-3-H8-PU
ID41 new 12.2 Fetal kidney 58-4-2-A3-PU
ID42 new 11.9 Kidney 21-10-4-G1-PU
ID43 new 11.3 Fetal kidney 58-27-3-B10-PU
ID44 new 10.7 Fetal kidney 58-35-2-F10-PU
ID45 new 10.7 Fetal kidney 58-37-2-G10-PU
ID46 new 10.6 Dystrophic muscle 29-11-1-Cl l-PU
BD47 new 10 Fetal kidney 58-20-4-G7-PU
ID48 new 10 Fetal kidney 58-2-4-E9-PU
ID49 new 9.6 Fetal kidney 58-37-3-D8-PU
ID50 new 9.5 Fetal kidney 58-46- 1-Fl-PU
ID51 new 9.2 Dystrophic muscle 29-9-4-D8-PU
ID52 new 9.2 Muscle 27-10-4-C6-PU
ID53 new 8.3 Heart 67-5-4-H9-PU
ID54 new 8.1 Fetal kidney 58-4-3-H4-PU
ID55 new 8 Muscle 27-16-3-D12-PU
ID56 new 7.9 Fetal kidney 58-54-2-C2-PU
ID57 new 7.9 Heart 25-9-3-A3-PU
ED58 new 7.9 Dystrophic muscle 29-1 1-3-F1-PU
ID59 new 7.9 Fetal kidney 58-32-3-G6-PU
ID60 new 7.8 Fetal kidney 58-22-2-H8-PU
ID61 new 7.8 Fetal kidney 58-2-4-H4-PU
ID62 new 7.8 Heart 67-4-3-G3-PU
ID63 new 7.8 Fetal kidney 58-24- 1-Gl l-PU
ID64 new 1.1 Fetal kidney 58-19-3-H1-PU
ED65 new 7.5 Fetal kidney 58-45-4-B11-PU
ED66 new 7.3 Fetal kidney 58-44-2-D3-PU
ID67 new 7.2 Dystrophic muscle 29-3-3-E7-PU
ID68 new 7.1 Dystrophic muscle 29-12-3-A3-PU
ID69 new 7.1 Fetal kidney 58-14-2-B3-PU
ID70 new 7.1 Fetal kidney 58-10-3-D12-PU
ID71 new 7 Fetal kidney 58-6-2-E5-PU
DD72 new 7 Dystrophic muscle 29-7-1-Cl-PU
ID73 new 6.9 Fetal kidney 58-26-4-A12-PU
ID74 new 6.9 Fetal kidney 58-7-2-H9-PU
ED75 new 6.9 Fetal kidney 58-14-2-D5-PU
ID76 new 6.1 Fetal kidney 58-3-4-E1-PU
ED77 new 6.1 Fetal kidney 58-43 -4-G3-PU
ID78 new 6.1 Fetal kidney 58-11-1-GlO-PU
ID79 new 6.6 Fetal kidney 58-4-4-G2-PU
ED80 new 6.6 Fetal kidney 58-41-3-D6-PU
ID81 new 6.6 Heart 25-8-2-H10-PU
ED82 new 6.5 Muscle 27-18-4-E5-PU
ID83 new 6.4 Dystrophic muscle 29-4-1-G6-PU
ID84 new 6.4 Muscle 27-10-2-B1-PU
ID85 new 6.4 Fetal kidney 58-38-1-E5-PU
ID86 new 6.3 Muscle 27-4-3-D9-PU SEQ. ID VON HEIJNE TISSUE INTERNAL
NO. CATEGORY SCORE SOURCE DESIGNATION
ID87 new 6.3 Fetal kidney 58-53-1-Gl-PU
ID88 new 6.3 Fetal kidney 58-7-3-F6-PU
ID89 new 6.3 Heart 25-7-2-B12-PU
ID90 new 6.1 Fetal kidney 58-16-3-E11-PU
ID91 new 6 Fetal kidney 58-15-4-C2-PU
ED92 new 6 Fetal kidney 58-34-3-A9-PU
ID93 new 5.9 Fetal kidney 58-16-1-El-PU
ID94 new 5.9 Fetal kidney 58-4-3-E6-PU
ID95 new 5.9 Fetal kidney 58-37-3-B11-PU
ID96 new 5.9 Fetal kidney 58-35-3-C6-PU
ID97 new 5.8 Fetal kidney 58-35-1-D9-PU
ID98 new 5.8 Fetal kidney 58-26-3-B2-PU
ID99 new 5.7 Fetal kidney 58-48-1-F8-PU
IDIOO new 5.7 Fetal kidney 58-27-4-A6-PU
IDIOI new 5.7 Fetal kidney 58-26-3-D1-PU
ID 102 new 5.7 Muscle 27-19-4-B4-PU
ID103 new 5.6 Fetal kidney 58-23-3-B2-PU
ID 104 new 5.5 Heart 25-1-2-C1-PU
ID 105 new 5.5 Fetal kidney 58-14-3-F10-PU
ID106 new 5.5 Fetal kidney 58-25-1-El l-PU
ID107 new 5.5 Muscle 27-9-4-A10-PU
ID108 new 5.5 Heart 25-4-2-D8-PU
ID 109 new 5.4 Fetal kidney 58-29-3-G8-PU
ID110 new 5.4 Fetal kidney 58-4-4-E5-PU
LD111 new 5.4 Fetal kidney 58-24-2-H2-PU
ID112 new 5.4 Muscle 27-11-2-C8-PU
ID113 new 5.4 Fetal kidney 58-41-2-E3-PU
ID114 new 5.3 Muscle 27-22-1-G8-PU
ID115 new 5.3 Dystrophic muscle 29-1-1-C9-PU
ID116 new 5.3 Fetal kidney 58-22-2-A3-PU
ID117 new 5.2 Fetal kidney 58-42-2-G1-PU
ED118 new 5.2 Fetal kidney 58-52-2-E5-PU
ID119 new 5.2 Fetal kidney 58-24-2-G2-PU
ID120 new 5.2 Fetal kidney 58-29- 1-A3-PU
ED121 new 5.1 Fetal kidney 58-26-1-G8-PU
ID122 new 5.1 Fetal kidney 58-29-4-G12-PU
ID123 new 5.1 Dystrophic muscle 29-8-3-E8-PU
ED124 new 5.1 Dystrophic muscle 29-3-4-C1-PU
ID125 new 5 Fetal kidney 58-17-2-H1-PU
ED126 new 5 Fetal kidney 58-9-3-E3-PU
ID127 new 5 Muscle 27-19-3-G7-PU
ID128 new 5 Fetal kidney 58-41-3-B4-PU
ID129 new 5 Dystrophic muscle 29-7-4-G7-PU
ID130 new 5 Muscle 27-9-3 -D4-PU
ID131 new 4.9 Kidney 21-3-4-C5-PU
ID132 new 4.9 Heart 25-11-2-D6-PU
ID133 new 4.9 Heart 67-7-2-F3-PU
ID134 new 4.8 Fetal kidney 58-4-3-D3-PU
ID135 new 4.8 Fetal kidney 58-49-3-B5-PU
JD136 new 4.8 Fetal kidney 58-28-3-G12-PU
ID137 new 4.7 Fetal kidney 58-53-1-A5-PU SEQ. ID VON HEIJNE TISSUE INTERNAL
NO. CATEGORY SCORE SOURCE DESIGNATION
ID138 new 4.7 Fetal kidney 58-3-3-E10-PU
ID139 new 4.7 Fetal kidney- 58-8-1-G7-PU
ID 140 new 4.6 Fetal kidney 58-23-1-G9-PU
ID141 new 4.6 Fetal kidney 58-21-1-H8-PU
ID142 new 4.6 Fetal kidney 58-54-2-E10-PU
ED143 new 4.6 Fetal kidney 58-46-3-E4-PU
ID 144 new 4.6 Fetal kidney 58-6-3-G3-PU
ID 145 new 4.6 Fetal kidney 58-41-2-B5-PU
ID146 new 4.6 Dystrophic muscle 29-7-3 -F2-PU
ID147 new 4.5 Fetal kidney 58-2-4-G12-PU
ID148 new 4.5 Fetal kidney 58-11-2-G8-PU
ID149 new 4.4 Fetal kidney 58-17-1-C4-PU
ID150 new 4.4 Fetal kidney 58-46-1-G7-PU
ID151 new 4.4 Heart 67-3-2-F4-PU
ID152 new 4.4 Fetal kidney 58-8-4-E12-PU
ID153 new 4.4 Fetal kidney 58-4-2-D9-PU
ID154 new 4.4 Fetal kidney 58-25-1-B5-PU
ID155 new 4.4 Fetal kidney 58-15-1-ClO-PU
ID156 new 4.3 Dystrophic muscle 29-4-4-A10-PU
ED157 new 4.3 Fetal kidney 58-32-3-H7-PU
ID158 new 4.3 Kidney 21-4-4-D12-PU
ID159 new 4.3 Fetal kidney- 58-45-4-G9-PU
ID 160 new 4.3 Fetal kidney 58-1-2-E2-PU
ID161 new 4.2 Fetal kidney 58-25-4-E6-PU
ID 162 new 4.2 Fetal kidney 58-36-4-C6-PU
ED163 new 4.2 Dystrophic muscle 29-9-3-D5-PU
ID 164 new 4.2 Fetal kidney 58-3-3-B8-PU
ID165 new 4.2 Heart 25-4-4-B4-PU
ID 166 new 4.2 Kidney 21-10-3-A3-PU
ID167 new 4.2 Muscle 27-19-4-B5-PU
ID168 new 4.2 Fetal kidney 58-23-3-D10-PU
ID169 new 4.1 Fetal kidney 58-41-1-F8-PU
BD170 new 4.1 Heart 25-7-2-B1-PU
ID171 new 4.1 Fetal kidney- 58-53-3-G4-PU
ID 172 new 4.1 Fetal kidney 58-52-2-C2-PU
ID173 new 4 Muscle 27-21-4-E12-PU
ID 174 new 4 Fetal kidney 58-22-2-B8-PU
ID175 new 4 Fetal kidney 58-9-3-A8-PU
ID176 new 4 Muscle 27-5-4-C10-PU
ID 177 new 4 Fetal kidney 58-38-1-G5-PU
ID178 new 4 Fetal kidney 58-34-4-F6-PU
ID179 new 4 Heart 25-1-4-D2-PU
ID180 new 4 Fetal kidney 58-48-2-D6-PU
ID181 new 3.9 Fetal kidney 58-9-3-C10-PU
ID182 new 3.9 Fetal kidney 58-9-4-F2-PU
LD183 new 3.9 Fetal kidney 58-32-3-G3-PU
ID184 new 3.9 Fetal kidney 58-52-1-F6-PU
ID185 new 3.9 Fetal kidney- 58-29-1-El-PU
ID186 new 3.9 Muscle 27-3-4-A3-PU
ID187 new 3.9 Muscle 27-16-3 -H2-PU
ID188 new 3.9 Fetal kidney 58-1-3-E1-PU SEQ. ID VON HEIJNE TISSUE INTERNAL
NO. CATEGORY SCORE SOURCE DESIGNATION
ID189 new 3.8 Kidney 21-5-4-F10-PU
ED190 new 3.8 Kidney 21-1-3-C9-PU
ID191 new 3.8 Fetal kidney 58-1-2-C7-PU
ID192 new 3.8 Fetal kidney 58-10-3-B6-PU
ID193 new 3.8 Fetal kidney 58-11-4-C8-PU
ID 194 new 3.8 Heart 67-6-4-B12-PU
ID195 new 3.8 Fetal kidney 58-7-3-B5-PU
ID196 new 3.8 Fetal kidney 58-46-3-C6-PU
ID197 new 3.7 Dystrophic muscle 29-2-4-D8-PU
ID198 new 3.1 Fetal kidney 58-7-1-DlO-PU
ID199 new 2.1 Kidney 21-2-4-A11-PU
ID200 new 3.6 Fetal kidney 58-45-3-B7-PU
ID201 new 3.6 Fetal kidney 58-29-1-D7-PU
ID202 new 3.6 Fetal kidney 58-16-3-B3-PU
ED203 new 3.6 Dystrophic muscle 29-7-3-C3-PU
ID204 new 3.6 Fetal kidney 58-42-3-C2-PU
ID205 new 3.5 Fetal kidney 58-38-3-G8-PU
ID206 new 3.5 Dystrophic muscle 29-6-2-B12-PU
ID207 new 3.5 Fetal kidney 58-8-1-Dl-PU
ID208 new 3.5 Fetal kidney 58-24-1-H2-PU
ID209 new 3.5 Fetal kidney 58-41-4-G9-PU
ID210 ext-est-not-vrt 12.7 Muscle 27-22-3-H1-PU
ID211 ext-est-not-vrt 10.5 Fetal kidney 58-29-1-Fl l-PU
ID212 ext-est-not-vrt 8 Fetal kidney 58-14-2-B12-PU
ID213 ext-est-not-vrt 1.1 Fetal kidney 58-5-1-C4-PU
ID214 ext-est-not-vrt 7.1 Fetal kidney 58-37-4-C7-PU
JD215 ext-est-not-vrt 6.1 Muscle 27-21-2-C8-PU
ID216 ext-est-not-vrt 6.1 Heart 67-1-1-C8-PU
ED217 ext-est-not-vrt 6.3 Fetal kidney 58-26-3-G6-PU
ID218 ext-est-not-vrt 6.2 Fetal kidney 58-15-3-B12-PU
ID219 ext-est-not-vrt 6 Muscle 27-5-2-Gl l-PU
ID220 ext-est-not-vrt 6 Fetal kidney 58-8-1-HlO-PU
ID221 ext-est-not-vrt 5.8 Fetal kidney 58-38-4-D2-PU
ID222 ext-est-not-vrt 5.6 Fetal kidney 58-53-2-E6-PU
ED223 ext-est-not-vrt 5.6 Fetal kidney 58-52-2-C7-PU
ID224 ext-est-not-vrt 5.5 Fetal kidney 58-34-2-E7-PU
ID225 ext-est-not-vrt 5.4 Fetal kidney 58-4-1-A2-PU
ID226 ext-est-not-vrt 5.2 Fetal kidney 58-11-1-D3-PU
ID227 ext-est-not-vrt 5.2 Fetal kidney 58-34-3-C9-PU
ID228 ext-est-not-vrt 5.2 Fetal kidney 58-35-4-H11-PU
ID229 ext-est-not-vrt 4.6 Fetal kidney- 58-3-4-H7-PU
ID230 ext-est-not-vrt 4.5 Fetal kidney 58-25- 1-F3-PU
ID231 ext-est-not-vrt 4.5 Fetal kidney 58-4-4-A8-PU
ID232 ext-est-not-vrt 4.4 Fetal kidney 58-11-1-Cl-PU
ID233 ext-est-not-vrt 3.9 Muscle 27-19-2-F5-PU
ID234 ext-est-not-vrt 3.5 Dystrophic muscle 29-2-2-A2-PU
ID235 est-not-ext 14.1 Fetal kidney 58-29-2-B9-PU
ED236 est-not-ext 11.4 Dystrophic muscle 29-11-2-E4-PU
ID237 est-not-ext 11.2 Fetal kidney 58-7-2-A7-PU
ID238 est-not-ext 10.8 Muscle 27-22-3-G4-PU
ID239 est-not-ext 9.9 Fetal kidney 58-9-1-Gl-PU SEQ. ID VON HEIJNE TISSUE INTERNAL
NO. CATEGORY SCORE SOURCE DESIGNATION
ID240 est-not-ext 9.1 Dystrophic muscle 29-8-1-H5-PU
ID241 est-not-ext 9.6 Fetal kidney 58-40-1-F5-PU
ID242 est-not-ext 9.5 Fetal kidney 58-6-4-G2-PU
ID243 est-not-ext 9.2 Fetal kidney 58-25-2-E7-PU
ID244 est-not-ext 8.9 Fetal kidney 58-48-1-Al l-PU
ID245 est-not-ext 8.8 Fetal kidney 58-35-2-B6-PU
ID246 est-not-ext 8.5 Kidney 21-7-4-C7-PU
ID247 est-not-ext 8.4 Fetal kidney 58-45-1-E6-PU
ID248 est-not-ext 8.1 Fetal kidney 58-39-1-A12-PU
ID249 est-not-ext 8 Fetal kidney 58-46- 1-C7-PU
ID250 est-not-ext 7.9 Dystrophic muscle 29-12-3-E10-PU
ID251 est-not-ext 7.9 Fetal kidney 58-17-2-D9-PU
ID252 est-not-ext 7.9 Fetal kidney 58-52-3-B7-PU
DD253 est-not-ext 7.6 Fetal kidney 58-24-3-E7-PU
ID254 est-not-ext 7.6 Heart 25-8-4-B 12-PU
ID255 est-not-ext 7.6 Dystrophic muscle 29-4-4-D 12-PU
ID256 est-not-ext 7.4 Muscle 27-1-2-B3-PU
ID257 est-not-ext 7.3 Fetal kidney 58-48-1-G3-PU
ED258 est-not-ext 7.3 Dystrophic muscle 29-2-3-F8-PU
ID259 est-not-ext 7.2 Fetal kidney 58-19-3-B3-PU
ID260 est-not-ext 7 Fetal kidney 58-14-2-C4-PU
ID261 est-not-ext 6.1 Fetal kidney 58-16-3-B6-PU
ID262 est-not-ext 6.6 Fetal kidney 58-9-4-F6-PU
ID263 est-not-ext 6.4 Fetal kidney 58-1-1-E3-PU
ID264 est-not-ext 6.4 Fetal kidney 58-33-3-B4-PU
JD265 est-not-ext 6.3 Dystrophic muscle 29-12-1-Hl-PU
ID266 est-not-ext 6.3 Muscle 27-9-3-A5-PU
ID267 est-not-ext 6.2 Muscle 27- 17-4-C 12-PU
ID268 est-not-ext 6.2 Fetal kidney 58-33-1-Fl-PU
ID269 est-not-ext 5.9 Fetal kidney 58-48-4-H2-PU
ID270 est-not-ext 5.9 Fetal kidney 58-42-1-A6-PU
ID271 est-not-ext 5.7 Fetal kidney 58-33-4-E1-PU
ID272 est-not-ext 5.7 Fetal kidney 58-26-2-E 12-PU
ID273 est-not-ext 5.6 Fetal kidney 58-26-1 -El 2-PU
ID274 est-not-ext 5.5 Fetal kidney 58-54-1-D 11-PU
ID275 est-not-ext 5.5 Muscle 27-9-2-F9-PU
ID276 est-not-ext 5.4 Fetal kidney 58-30-2-H 10-PU
ID277 est-not-ext 5.3 Fetal kidney 58-29-1-Hl-PU
ID278 est-not-ext 5.3 Kidney 21-1-4-F2-PU
ID279 est-not-ext 5.1 Fetal kidney 58-42-4-H7-PU
ID280 est-not-ext 5 Fetal kidney 58-34-3-H10-PU
ID281 est-not-ext 5 Kidney 21-7-3-B4-PU
ID282 est-not-ext 4.9 Fetal kidney 58-4-2-D 12-PU
ED283 est-not-ext 4.8 Fetal kidney- 58-31-2-C10-PU
ID284 est-not-ext 4.7 Fetal kidney 58-37-3-C10-PU
ID285 est-not-ext 4.1 Fetal kidney 58-1-1-Dl l-PU
ID286 est-not-ext 4.6 Fetal kidney 58-52-1-Al l-PU
ID287 est-not-ext 4.3 Fetal kidney 58-4-3-E 10-PU
ID288 est-not-ext 4.3 Heart 67-6-4-F2-PU
ID289 est-not-ext 4.2 Fetal kidney 58-49-3-G10-PU
ID290 est-not-ext 4.1 Dystrophic muscle 29-10-3-B11-PU SEQ. ID * VON HEIJNE TISSUE INTERNAL
NO. CATEGORY SCORE SOURCE DESIGNATION
ID291 est-not-ext 4.1 Heart 25-5-4-A7-PU
ID292 est-not-ext 4.1 Fetal kidney 58-33-2-C6-PU
ID293 est-not-ext 4 Heart 25-7-3-D4-PU
ID294 est-not-ext 3.9 Heart 67-1-3-B11-PU
ID295 est-not-ext 3.9 Fetal kidney 58-23-1-G5-PU
ID296 est-not-ext 3.7 Fetal kidney 58-6-1-B6-PU
ID297 est-not-ext 3.7 Dystrophic muscle 29-6-2-H8-PU
ID298 est-not-ext 3.1 Fetal kidney 58-43-4-B8-PU
ID299 est-not-ext 3.6 Muscle 27-3-4-G9-PU
ID300 est-not-ext 3.6 Fetal kidney- 58-38- 1-F 10-PU
ID301 est-not-ext 3.5 Heart 67-6-4-E7-PU
ID302 est-not-ext 3.5 Fetal kidney 58-54-1-E6-PU
ID303 est-not-ext 3.5 Heart 67-4-4-G7-PU
ID304 est-not-ext 3.5 Fetal kidney- 58-23-4-F4-PU
DD305 ext-vrt-not-genomic 10.5 Fetal kidney 58-42-3-A12-PU
TABLE HI
SEQ. ID
NO. SIGNAL PEPTIDE
ID38 MMWRPSVLLLLLLLRHGAQG
ID39 MERPLCSHLCSCLAMLALLSPLSLA
ID40 MIHLGHILFLLLLPVAAA
ID41 MAVKLGTLLLALALGLAQPASA
ID42 METLGALLVLEFLLLSPVEA
ID43 MLLPLLLSSLLGGSQA
ID44 MLWLLFFLVTAIHA
ID45 MAGSPSRAAGRRLQLPLLCLFLQGATA
ID46 MKWPWTCLAILCPGPVLSPPCSGPXLALALLLVLPLLWP
ID47 MPSWIGAVILPLLGLLLSLPAGA
ID48 MLLHWVRSQXXSDXKLWLSLLVPSCLCA
ID49 MKYLRHRRPNATLJXAIGAFTLLLFSLLVSPPTC
ID50 MPGPRVWGKYLWRSPHSKGCPGAMWWLLLWGVLQA
ID51 MCGPAMFPAGPPWPRVRWQVLWALLAVLLASWRLWA
ID52 MHRRKLPLTNKRQLQJTQOJSKFJTSDELFRNILFSLRTLW
ED53 MKLWVSALLMAWFGVLS
ID54 MQLPLALCLVCLLVHTAFR
ID55 MLCIHXXRIIQDSFIALKILLCSVAVXLSPS
ID56 MGGFFPPTEVREVCANQGAAHNRDRLPFLSLFWPWAPG
ID57 MKLFYNQLVSETKHDFAHLWILLLFSFCWM
ID58 MPSESPPLLFFHJ FHSCFS
ID59 MSSM SEYΗGGVKTiT^YKAYPSQLAMMNSILRGLNSKQHCLLESPTGSGKSLALLCSA
LAWQQSLS
ID60 MALFLELFLNSYSLLFVRFLGFVSCLQS
ID61 MNEDEKEMKEILMAGSSLSAGVSG
ID62 MGSFLLGGIIPLLXXLSLCLC
ID63 NI QVATTNYLELAREVKPVCLLCSGCSCAWS
ID64 MFCLAPFFLALCFPKSTS
ID65 MSESRFQPQNQGGSLQLPLQCLLCCISPPVFC
ID66 MPKHCHSFITSSCLLGLLHLSSQ
ID67 MCLLFXFDCFPFLFPFSFS
ID68 MASERXPNRPXCLLVASGXAEGVSA
ID69 MFPDYKLGGSYLLAFQLVFLRATSG
JD70 MRRISLTSSPVRLLLXLXLLLIALE
ID71 MTFLLLLFXNAGRS
ID72 MRT\^VLTMKASVIEMFLVLLVTGVHS
ID73 MSSPLLVEQSSTKSPKSWSWSFLAFSCISLLFIFFSIANS
ID74 MYLFCLFSVSKTIPLLLLFFHLSFL
ID75 MIVCLLILKFLSPAET
ID76 MDKSIKSSIIWSLILCFLFILHTHT
ID77 MF IFπ^GFTLLLMTLAMKPRHPIFDLLLLLXXSNQ
ID78 MCPSLEEAPSVKGTLPCSGQQQPFPFGASNIPLLLGRSRKVARGAPVLWPFLTWINPALS
ID79 MLQDLLSALWFCHPCCL
ID80 MMDLRPLLSLAAYLSGPHQ
ID81 MEMPPCLLPGLPLVRTSFS
ID82 MTVELWLRLRGKGLAMLHVTRGVXG
ID83 MSIEDFVNRSILLILLCSSPPDRV
ID84 MRIHYLLFALLFLFLVPVPG
ID85 MCLLTALVTQVIS SEQ. ED NO. SIGNAL PEPTIDE
ID86 MMGNPGLxΛLVAGTPPSRS ED87 IMNHLMPLTVLHS\TJEMLRTPRTPPWPCVSLLWAPRXFA ID88 MGHWFGDDCNSLLXLRASQLSEG ED89 MAGGRRDYSQLFGRGPGPJ.SRARASVWWSPPvATACPAPPSLPDLKRQELVSRJJΞCGCRG
PVGATADFFLSLLXSVSETPG
ED90 MFWXGSLWCFHSFISFSLS ID91 MAWPNVFQXGSLLSQFXXHHVWFLLTFFSYSLLHA ID92 MILRNLWILAVGLSLPSSS ID93 MLTVNDVRFYRNVRSNHFPFVRLCGLLHLWLKVFS ID94 MNLKPGLPCNLFLNLCILAXPFS ID95 MMQGEAHPSASLroRTIKMRKETEARKVVLAWGLLNVSMA ID96 MMNQTHPXXLLILAHITQS ID97 MGLPERRGLVLLLSLAEILF ID98 MWGLEEDRSYQGLRPLCWALLYNCFSSS ID99 MLCRDGSACWRSRRLPLPAAVRAHGPMADXXDSARGCVVFEDVFVYFSREEWELL
DDAQRLLYHDVMLENFALLASLGIAFSRS
ID 100 MLITRLQSGroFAIQLDESTDIGSCTTLLVYVRYAWQDDFLEDFLCFLNLTSHLSG ID101 MESPQLHCILNSNSVACSFAVGAGFLAFLSCLAFLVLD ID102 MSNKYIKPSMSPGNTDHLFLLFPRSCSS ID103 MVELKQLGPRSFFFFLFLLPPXPP ID 104 MPYWJPYΠWSLIJIPALFFFPLHC ID105 MPPLAAVMGSLPLLLCMDLPHSVLS ID106 MLQIPERREFLFLGFPSNSWP ED107 MFFVHFLITLFCCC VG ED108 MACFGEKRHAKSCLLHLRCLQLY A ID109 MVDRDENILLKQIYSPLSLALQSSCCLC ID110 MKVKPPFVSVSLCVCDCVRG ID111 MISSCGVKYLFSHASLFFMVGSTGSLILLTSCFYTLVSS ID112 MGGGIAESFLCNFLVSLSLS ID113 MDALERGSLRNEQALVIYAGLAYFLCCQGVIFG LD114 MEYLFQQPGHSRGEARAAAASLETLSSLWFLPLPTHVYT ID115 MVSSMLITELSFIFA ID116 MPLFTMNLVSALASSAXG ID117 MICK)r CIKKN ILDYL JRMVYSAQLKLILLLHCSIRVFF ID118 MKJPVWHKTCFLKSESFSPDNLSVSLPCRPSQWSQGQGKSFLLLQLIHEDKA ID119 MGAAVFFGCTFVAFXPAFA ID120 MVGGLDPPGRRRFQKGFDWRNLWSSCWLAPLADG ID121 MSKMPVFASLLWSCFYQISG ID122 MXVTQLLPFSSPDSA ED123 MGKAWQEMR\πΞWGADKG VRSSFHFLPWALGAMA ID 124 MKVMMRKIIKKKDQCLPGICRSLKRRKSPRSPGMKVIRLSQFLLKCWP ID 125 MTFSFFCFT>TGFKPLLFIiYFLFXSFSIXTLLWGLNC ID 126 MAGGMKVAVSPAVGPGPWGSGVGGGGTVRLLLILSGCLVYG ID127 MVEMTGVWQCQAEAVKGLPPLLSCSCPPPLLG ID 128 MQITPGSAAGLLPLLLGNAPG JJD129 MJ STWLLLTLQNSVFT ID130 MATΗSYWGKSLQSFK'TFMRVCIVLALCHTSRP JD131 MKLRFTLLPLVLHSQS ID132 M ILGFAFCPGFffRFNFIPFLVIYSFVLS ED133 MNRVPADSPNMCLICLLSYIALGAIHA ID 134 MDLFLNLPLVIGTIP SEQ. ID
NO. SIGNAL PEPTIDE
ID 135 MXKNHRNKKSIHFPLCTIPSXMXKSCTLPLQRT WDXXPSFVH WXQ ARLQSPPXSHL VXLS
VIRSTLVLSQCLC
ID 136 MSFIALVYSSLSFQ
ID137 MVFDTLKSRIVLFLNSXFPIIC
ID 138 MLEMEMTWLRLCDECSRWGMAS AWGRGGKLLGAQ VALHPRNCSKAKIFLFSILLMSLRT
CD 139 MDDLMLFFLGALCRESG
ID 140 MVLGALNLPSQELPTLLLLPVGAPG
ID 141 MLVSKIQTFVSFLSIPVLG
ID 142 MCWVAHTFRGVHEHHAMLLSTGLNILGTQA
ID 143 MQCWD LWEACTGRCQA
ID 144 MTGYPWANSITTVLCILGCHGNLCC
ID 145 MVSCDVXSYVIIFTALFLXLHSVA
ID 146 MKSFDKKLFJAIFLMCLKSIG
ID 147 MFGAGDEDDTDFLSPSGGARLASLFGLDQXAXG
ID 148 MVLTLGESWPVLVGRRFLSLSAADGXDX
ID 150 MESTLGAGIVI AEALQNQLA WLENVWL WXXLXXXIPXILFLFYFP AAYY A
ID 151 Mil VSELGTPTG VL VG VFLSTFLYC
ID 152 MNWNVRGTRGFLLCPLVCGLRR
ID153 MLRCGGRGLLLGLAVAAAA
ID 154 MELLMIVFSIFLLL
ID 155 MSLLFIFRSILISC
ID157 MDTSSVGGLELTDQTPVLLGSTAMATSLT
ID 158 MDTGESFSPFTTSCRGHWRILLLTHVPPWILE
ID 159 MPYLDPYITQPIIQIERKLVLLSVLKEPVSR
ID 160 MDTSSVGGLELTDQTPVLLGSTAMATSLT
ED161 MHVLFNIVTTNXXNHFGLLDFVVQCCDS
ID 162 MPPQSCCSKTAYWLSFMSWAQS
ID 163 MSC\TTHFLQGGLG
ID 164 MSISLSSLIXLPIWΓNMAQI
ID 165 MTALNLVAPFSDGDSGSVSLASFCNAWLSPVFQ
ID 166 MWSRPVQVLGLLATCQH
ID 167 M YRLRIQITTSLNQILLFLLISC
ID 168 MPFFSNQPTQVSVLLFFCCSPLYSP
ID169 MRVKDPTKALPEKAJKRSKRPTWHDEDSSDDIAVGLTCQHVSHA
ID170 MVSLGYYLIFVLYLWLCFMQISEEKLIEEHTGTYLTSSSPLCQL
ID171 MSLTSRXXI XTIKIQNISITKVLCCLLIATPTFF
ID 172 MXAEAAGWSTSVAAAVA
ID 173 MWIMSSCLALTYTNS
ID 174 MPRGVYNSNALVLVTRGSSS
ID 175 MffiPCEKMKHYDMNWFLCMYECFFTΗLLETEFLLPC VHPFS VI A
ID176 MAMWNRPCQXLPQQPLVAEPTAEGEPHLPTGRELTEANRFAYAALCGISLSQXFP
ID 177 MEQVCLLVSYAVDSAAG
ID 178 MRKISHCLHCWPESGATLRCWASTPVSG
ED 179 MCINDHJIKLLHPCGSITLTSS
ID 180 MRCRVALQCGLTIP ALX
ID 181 MTVRYGKJ?LSLLKDGA£NDLT\V .KHCERFT,KQQQTSIKSSLLCLQGNYAGHDWFVSSLF
MIMLGDKΕKTFQFLHQFSRLLTSAFLWLPRLHI
BD182 MAFDVSCFFWWLFSAGCKV
ID 183 MLTRLVLSAHLSSTTSPPWTHA SEQ. ID NO. SIGNAL PEPTIDE
ID184 MRYFQGPSPYSEIEIELCDHVYSFQGLCVNLLLGFEPVIS
ID 185 MXXKRTHXXXSVFNGLVYAAGGRNAEGSLASLECYVPSTNQ
ED 186 M)?TJKVQSQSFYXPYRDCLNFHKSTYLLFFHLLLNDFFT D 187 MQPLKIIFYLS VSI WIILIIYTFQCNS
ID 188 MMRTTARVAACTAAAPLQ A
ID 189 MEAATTLHPGPRPALPLGARARWASSCLHPSARS
ID 190 MQGVRGPVSFSWSTTMLCPVIFFPSNCWK
ID 191 MXXFSFXLLFXXFXFFRQ
ID 192 MLLLSEALSESVRLLFRFSVIMA
ED 193 MALISLPCTTAFPLLSS
ED 194 MSEEE AAQEPRSS VWEQDQQNWQR WALPL VRATCT
ED 195 MAAAAAAGAASGLPGPVAQGLKE ALVDTLTGILSP VQE VRAAAEEQEKVLE VTEEFGVHL
AELTVDPQGALA
ID 196 MNSGGGFGLGLGFGLTPTS\αQVTNLSSAVTSEQMRTLFSFLGEEEELRLYPPDNAPLAF
SSXVCYVKFRDPSSVGVAQHLTm'VFIDRXLXSCSLCRRLVSRFXXXYLNFCPVCYC
ID 197 MIEMLIFLDCVLS
ED 198 MHP FLAAHGPAFHKGYKHSTINI VDIYPMMCHILGLKPHPNNGTFGHTKCLL VDQ WCINL
PEAIAIVIGSLLVLTMLTC
ID 199 MIWPMSASVATLWS
ID200 MGroiFV SHjPDFHPIHLFIYLVFVECLLC
ED201 MKELNQKLTNKNNKJΞDLEQEIKIQKQKQETLQEEITSLQSS VQEYEEKNXKEKQLLVKT
KKELADSKQAETDHLILQASLKGELEA
ID202 MGNTLKEMQDVQGALQCYTPVAIQINPAFADAHSM.AS1ΗKDSGNIPEAIASYRTALKLKP DFT3DAYC π.AHCLQIVCDWTDYDERMKJXLVSIVADQLEKNRLLLCILIIVCY[
ED203 MLILADTRRVQGGTLGLIPAVLNRV rv'AYAIPSIPSLFC
ED204 MLVGEYFCVFLFPLISNTSS
ID205 MT^APSLLITKLLTGSESPDGNPPALGRPLLLQGACPCLIFL
ED206 MDPSASKSCLFYLQKVSG
ID207 MSLTASGPRAAWEERVGGLHTWGANIPTAPDSQRWLCLQAYLASFS
ID208 MKYQMVSGSAQLASPLLPGATP
ID209 MNGTFPGTYVYLVAYGDLRIFGCFWGLMYX\VLLLG
ID210 MGPSTPLLILFLLSWSGPLQG
ED211 MKFISTSLLLMLLVSSLSPVQG
ED212 MNYQYGFNMVMSHPHAVNEI ALSLNNKNPRTK AL VLELL AA VCL VRG
E>213 MAQSIHMYAARVQWGL VMCFLS YFGTF A
ED214 MGSGYSHSLHLFHLLIRPXQG
ID215 MARCFSLVLLLTSIWT
ID216 MAMRYNRLTVLAGAMLALGLMTCLSVLFGYATS
ID217 MPQQPVEQGSPLLRQLLLPLPPFSFP
ED218 MPSRSPFTWSHLCWRAGRCPRWRACLSSSSVRMCSPAAPSRFGALGXSARRWPRRDA DTWCAPQGVMRASLLPMLLGSWA
ID219 MSHTEVKLKEPFGNKLLDAVCLVPNKSLTYGIILTHGASG
ID220 MELGSCLEGGREAAEEEGEPEVKKRRLLCXEFXSVASCDA
ID221 MGRTYIVEETVGQYLSNINLQGKAFVSGLLIGQCSS
ED222 MGSRKCGGCLSCLLEPLALWS
ID223 MGSRKCGGCLSCLLIPLALWS
ED224 MWWFQQGLSFLPSALVIWTSA
ED225 MFNASTFTOWSSSIFFVFTFKSKKSAGLPLIFSLWCSGVLL
ID226 MKMASSLAFLLLNFHVSLLLVQLLTPCSA
ED227 MfflLQLLTTVDDGIQAIVHCPDTGKDIWNLLFDLVCHEFCQS
ID228 MSDQIKFIMDSLNKEPFRKNYNLITFXSLEPMQLLQVLSDVLA SEQ. ID NO. SIGNAL PEPTIDE
ID229 MATSSQXRQLLSDYGPPSLGYTQGTGNSQXPQSKYAELLAIIXELGKEIRPMYAGSKSAM
ERLI RGIIHAXGLVRECLA ID230 MRLLGAAAVAALGRG
ID231 MAQRLLLRRFLASVIS
ID232 MFRLNSLSALAELAVG
ID233 MSGSNGSKENSH IKARTSPYPGSKVERSQW ΕKVGWLVEWQDYKPVEYTAVSVLA
GPRWA ID234 MRTTLMFSLTAQWXTS
ID235 MSDLLLLGLIGGLTLLLLLTLLAFA
ID236 MEGTEMGARPGGHPXKWSFLWSLALWLPLALS
ED237 MXTT.RKVXSILSLQVLLTTVTSTVFLYFESVRTFVXESPALILLFALGSLGLIFA
ID238 MAATLGPLGS WQQ WRRCLS ARDGSRMLLLLLLLGSGQG
ED239 MSS WMYLGYPIVTSNTTCLKLISSSFPQELPFLLFPFP VN A
ID240 MAPGVIIIQLCLLLLPSCSLS
ED241 MRHGFIQQQFSLTAFSXXXXEFTLXXLSQLLSSAAPKHTAAPTALPCLQGQQLNSLSLGT
SELSCVLASSCLSTKTDPSGLSLSLGASAPVQC ED242 IFQMQKCL TFVRGYHVFYINLNAVILIIFLSFLPFINS
ED243 MSLSQRGFPVLALFLSGSLA
ID244 MAARWRFWCVSVTMWALLIVCDVPSASA
ID245 MFAPAVMP^FEKNKTLGYGVPMLLLIVGGSFG
ED246 MELPSGPGPERLFDSHRLPGDCFLLLVLLLYAPVGFC
ID247 iMAQSQG VXRYXKAFCKGFTVA VAVTFLDRVACVARVEGASMQPSLNPGGSXSS
DVVXXNΪTWKVRNFEVHRGDIVSLVLLTVTPSXRQ ID248 MSSAAADHWAWLLVLSFVFGCNV
ID249 MNLFKTNHVFFLLLLAHIIA
ID250 MPALLPVASRLLLLPRVLLTMASG
ID251 MIGSGLAGSGGAGGPSSTVTWCALFSNHVAATQASLLLSFVWMPALLPVASRLLLLPRVL
LTMASG ID252 MPALLPVASRLLLLPRVLLTMASG
ID253 MEASWGSFNAERGWYVSVQQPEEAEAEELSPLLSNELHRQRSPGVSFGLSVFNLMNAIMG
SGJXGLAYVMANTGVFGFSFLLLTVALLASYS ED254 MPSSFFLLLRFFLRIDG
ID255 MKRTHLFIVGIYFLSSCRA
ID256 MGDKIWLPFPVLLLAALPPVLLP
ID257 MPHSSLHPSIPCPRGHGAQKAALVLLSACLVTLWGLG
ID258 MGAWGRGWPWEERQGHHLLLLLLPAPTLK
ID259 MGQCGITSSKTVL\^ ILffWGAAGILCYVGAY ITYDDYDHFTΕDV^
RALLFIIGLIGCCAT ID260 MPXAFSVSSFPVSffAVLTQTDWTEPWLMGLATFHALCVLLTCLSSRSYRLQIGHFLCLV
E VYC ID261 MLLLSLFFPLRISL
ID262 METGERARLIL VLQLLLRERR
ED263 MCGXXFSLPCLRLFLVVTCYXLLLLHKEILGCSSVCQLCTG
ID264 MNPVTESPSCLFSPPSESALASQLALSASCDQRAPFSLAGVXSXXPRLASRQVAPPFGSR
ACCFLSAFSPTLT
ID265 MSRSSKWLGLSVLLTAATVA
ID266 MGIQTSPVLLASLGVGLVTLLGLAVG
ED267 MYPSYLLIXPPIPSQFLKQCXPPTLSDPFLPLALRSLDVLLLSSAXLVXXS
ED268 MEQKHRXELEQLKLXTKΞNKILLLXTFQTWCLR
ID269 MMTAPVLAAQTLKFLTLLQKSNA
ID270 MDSAACAAAATPVPALALAXAPDLAQA SEQ. ID NO. SIGNAL PEPTIDE
ID271 MASLGLQLVGYILGLLGLLGTLVA ID272 MASLGLQLVGYELGLLGLLGTLVA BD273 MLCSLLLCECLLLVAGYA ID274 MASRLCGGALWYVCPCPSGAWM ED275 MTSALTQGLERIPDQLGYLVLSEGAVLA ED276 MASPSPJ .QTKPVITCFKSVLLIXTXIXWITGVILLAVGIWG ED277 MADAASQVLLGSGLTELSQP ED278 MSRNLRTALIFGGFISLIGA ED279 MPHGLWCFΉLWLSLYS ED280 MSLVAVFLSCGLIS ED281 MMKRAAAAAVGGALAVGAVPVVLS ED282 MAVI\TJKPWFYDiMKKVλVEGYPIQSTjTSQYWYYMffiLSFYWSLLFSIASD\ j7f^ fflHVATIILISFSWFANYIRA
DD283 MΠSLFΓYLFLTCSNT ID284 MAAELVEAKNMVMSFRVSDLQMLLGFVGRSKS ED285 MΓGLSMXGGGSXXGDVXPXYYGKXGPLRXLPEPSGPLPPSSGLSQPQVHALCPLSPLVTT ID286 MQMYSRQLASXEWLTIQGGLLGXGLXXXSLT ED287 MASLEVSRSPPVRSRRELEVRSPRQ IKYSVLLPTYNEPVENLPLIVWLLVKSFSES ID288 MDKDSQGLLDSSLMASGTAS ID289 MGLLTFGYEEXXXKTEHNPDHHSCLAVSWEAAGCHG ID290 MGLYAAVAGVLAGVES ID291 MGLYAAAAGVLAGVESRQGSΠ GLVYSSNFQNVKQLYALVCETQRYSAVLDAVIASA
GLLRA
ID292 MGAQHTALLL>ΓΓEVRWLSRGKVLVRLFELRRELLVFMDSAFTILSDCLTNSSWLLRLAYLA
DEFT
ED293 MSLRNLWRDYKVLWMVPLVGLIHL ED294 MVLRSLVEYSQDVLAHPVSEEHLPDVSLIGEFSDPAELGKLLQLVLGCAIS ED295 MIHGFCLAPTTSA ED296 MXCPRTWCLACVEASPG ED297 MADVEDGEETCALASHSGSSG ED298 MFKVAAPPMLΓXXIIMFLLΠVCGSP ED299 MDFWDPAVFXMCLWSLRNLFS ED300 MSPAGKHNSESKFTFFVALDGSVPLLSLSHSIGI ED301 MH ALVCVGLHTEGPWG ED302 MFGAAARSADLVLLEKM.QAAHGYAQEDRERMHRXIVSLXQNLLNFMIGS)1DLWQCF
LWFYIGSSLNGTRG
ED303 MAARWRFWCVSVTMVVALLIVCDVPSA ED304 MWLLLQPSMIQEVWT ED305 MLHLHXSCLCFRSWLPAMLAVLLSLAPSASS 129/1
TABLE IV
129/2
TABLE V
129/3
TABLE VI 129/4
Description of Transcription Factor Binding Sites present on promoters isolated from SignalTag sequences
Promoter sequence P13H2 (646 bp):
Matrix Position Orientation Score Length Sequence
CMYB_01 -502 + 0.983 9 TGTCAGTTG
MYOD_Q6 -501 0.961 10 CCCAACTGAC
Sθ_01 -444 - 0.960 11 AATAGAATTAG
S8_01 -425 + 0.966 11 AACTAAATTAG
DELTAEF1_01 -390 - 0.960 11 GCACACCTCAG
GATA_C -364 - 0.964 11 AGATAAATCCA
CMYB_01 -349 + 0.958 9 CTTCAGTTG
GATAΪ"_02 -343 + 0.959 14 TTGTAGATAGGACA
GATA_C -339 + 0.953 11 AGATAGGACAT
TAL1ALPHAE47_01 -235 + 0.973 16 CATAACAGATGGTAAG
TAL1 BETAE47_01 -235 + 0.983 16 CATAACAGATGGTAAG
TAL1 BETAITF2~_01 -235 + 0.978 16 CATAACAGATGGTAAG
MYOD_Q6 -232 - 0.954 10 ACCATCTGTT
GATA1_04 -217 - 0.953 13 TCAAGATAAAGTA
I 1_01 -126 + 0.963 13 AGTTGGGAATTCC
IK2_01 -126 + 0.985 12 AGTTGGGAATTC
CREL_01 -123 + 0.962 10 TGGGAATTCC
GATA"Ϊ_02 -96 + 0.950 14 TCAGTGATATGGCA
SRY_02 -41 - 0.951 12 TAAAACAAAACA
E2F_02 -33 + 0.957 8 TTTAGCGC 2F1 01 -5 - 0.975 8 TGAGGGGA
Promoter sequence P16B4 (8β1bp) :
Matrix Position Orientation Score Length Sequence
NFY_Qβ -748 - 0.956 11 GGACCAATCAT
MZF1_01 -738 + 0.962 8 CCTGGGGA
CMYB_01 -684 + 0.994 9 TGACCGTTG
VMYB_02 -682 - 0.985 9 TCCAACGGT
STAT_01 -673 + 0.968 9 TTCCTGGAA
STAT_01 -673 - 0.951 9 TTCCAGGAA
M2F1_01 -556 - 0.956 8 TTGGGGGA
IK2_01 -451 + 0.965 12 GAATGGGATTTC
MZF1_01 -424 + 0.986 8 AGAGGGGA
SRY_02 -398 - 0.955 12 GAAAACAAAACA
MZF1_01 -216 + 0.960 8 GAAGGGGA
MYOD_Q6 -190 + 0.981 10 AGCATCTGCC
DELTAEF1_01 -176 + 0.958 11 TCCCACCTTCC
S8_01 5 - 0.992 11 GAGGCAATTAT
MZF1 01 16 - 0.986 8 AGAGGGGA
Promoter sequence P29B6 (556 bp) :
Matrix Position Orientation Score Length Sequence
ARNT_01 -311 + 0.964 16 GGACTCACGTGCTGCT
NMYC 01 -309 + 0.965 12 ACTCACGTGCTG
USF_01 -309 + 0.985 12 ACTCACGTGCTG
USF_01 -309 - 0.985 12 CAGCACGTGAGT
NMYC_01 -309 - 0.956 12 CAGCACGTGAGT
MYCMAX_02 -309 - 0.972 12 CAGCACGTGAGT
USF_C -307 + 0.997 8 TCACGTGC
USF_C -307 - 0.991 8 GCACGTGA
MZF1 01 -292 - 0.968 8 CATGGGGA
ELK1_02 -105 + 0.963 14 CTCTCCGGAAGCCT
CETS~1 P54_01 -102 + 0.974 10 TCCGGAAGCC
AP1_Q4 -42 - 0.963 11 AGTGACTGAAC
AP1 FJ_Q2 -42 - 0.961 11 AGTGACTGAAC
PADS C 45 + 1.000 9 TGTGGTCTC
TABLE VII

Claims

1. A purified or isolated nucleic acid comprising the sequence of one of SEQ ED
NOs: 38-305 or comprising a sequence complementary thereto.
2. The nucleic acid of Claim 1, wherein said nucleic acid is recombinant.
3. A purified or isolated nucleic acid comprising at least 10 consecutive bases of the sequence of one of SEQ ID NOs: 38-305 or one of the sequences complementary thereto.
4. A purified or isolated nucleic acid comprising at least 15 consecutive bases of one of the sequences of SEQ ID NOs: 38-305 or one of the sequences complementary thereto.
5. The nucleic acid of Claim 4, wherein said nucleic acid is recombinant.
6. A purified or isolated nucleic acid of at least 15 bases capable of hybridizing under stringent conditions to the sequence of one of SEQ ID NOs: 38-305 or one of the sequences complementary to the sequences of SEQ ID NOs: 38-305.
7. The nucleic acid of Claim 6, wherein said nucleic acid is recombinant.
8. A purified or isolated nucleic acid encoding a human gene product, said human gene product having a sequence partially encoded by one of the sequences of SEQ ID NO: 38-305.
9. A purified or isolated nucleic acid having the sequence of one of SEQ ED
NOs: 38-305 or having a sequence complementary thereto.
10. A purified or isolated nucleic acid comprising the nucleotides of one of SEQ D NOs: 38-305 which encode a signal peptide.
11. A purified or isolated polypeptides comprising a signal peptide encoded by one of the sequences of SEQ ID NOs: 38-305.
12. A vector encoding a fusion protein comprising a polypeptide and a signal peptide, said vector comprising a first nucleic acid encoding a signal peptide encoded by one of the sequences of SEQ ID NOs: 38-305 operably linked to a second nucleic acid encoding a polypeptide.
13. A method of directing the extracellular secretion of a polypeptide or the insertion of a polypetide into the membrane comprising the steps of: obtaining a vector according to Claim 12; and introducing said vector into a host cell such that said fusion protein is secreted into the extracellular environment of said host cell or inserted into the membrane of said host cell.
14. A method of importing a polypeptide into a cell comprising contacting said cell with a fusion protein comprising a signal peptide encoded by one of the sequences of
SEQ ED NOs: 38-305 operably linked to said polypeptide.
15. A method of making a cDNA encoding a human secretory protein that is partially encoded by one of SEQ ED NOs 38-305, comprising the steps of: obtaining a cDNA comprising one of the sequences of SEQ ID NOs: 38-305; contacting said cDNA with a detectable probe comprising at least 15 consecutive nucleotides of said sequence of SEQ ED NO: 38-305 or a sequence complementary thereto under conditions which permit said probe to hybridize to said cDNA; identifying a cDNA which hybridizes to said detectable probe; and isolating said cDNA which hybridizes to said probe.
16. An isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein encoded by one of SEQ ED NOs 38-305 or a fi'agment thereof of at least 10 amino acids, said cDNA being obtainable by the method of Claim 15.
17. The cDNA of Claim 16 wherein said cDNA comprises the full protein coding sequence partially included in one of the sequences of SEQ ED NOs: 38-305.
18. A method of making a cDNA comprising one of the sequences of SEQ ED NOs: 38-305, comprising the steps of: contacting a collection of mRNA molecules from human cells with a first primer capable of hybridizing to the polyA tail of said mRNA; hybridizing said first primer to said polyA tail; reverse transcribing said mRNA to make a first cDNA strand; making a second cDNA strand complementary to said first cDNA strand using at least one primer comprising at least 15 nucleotides of one of the sequences of SEQ ED NOs 38-305; and isolating the resulting cDNA comprising said first cDNA strand and said second cDNA strand.
19. An isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein encoded by one of SEQ ID NOs 38-305 or a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method of Claim 18.
20. The cDNA of Claim 19 wherein said cDNA comprises the full protein coding sequence partially included in one of the sequences of SEQ ED NOs: 38-305.
21. The method of Claim 18, wherein the second cDNA strand is made by: contacting said first cDNA strand with a first pair of primers, said first pair of primers comprising a second primer comprising at least 15 consecutive nucleotides of one of the sequences of SEQ ED NOs 38-305 and a third primer having a sequence therein which is included within the sequence of said first primer; performing a first polymerase chain reaction with said first pair of nested primers to generate a first PCR product; contacting said first PCR product with a second pair of primers, said second pair of primers comprising a fourth primer, said fourth primer comprising at least 15 consecutive nucleotides of said sequence of one of SEQ ED NO:s 38-305 , and a fifth primer, said fourth and fifth primers being capable of hybridizing to sequences within said first PCR product; and performing a second polymerase chain reaction, thereby generating a second PCR product.
22. An isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein encoded by one of SEQ ID NOs 38-305, or a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method of Claim 21.
23. The cDNA of Claim 22 wherein said cDNA comprises the full protein coding sequence partially included in one of the sequences of SEQ ID NOs: 38-305.
24. The method of Claim 18 wherein the second cDNA strand is made by: contacting said first cDNA strand with a second primer comprising at least 15 consecutive nucleotides of the sequences of SEQ ED NOs: 38-305; hybridizing said second primer to said first strand cDNA; and extending said hybridized second primer to generate said second cDNA strand.
25. An isolated or purified cDNA encoding a human secretory protein, said human secretory protein comprising the protein partially encoded by one of SEQ ED NOs 38- 305 or comprising a fragment thereof of at least 10 amino acids, said cDNA being obtainable by the method of Claim 24.
26. The cDNA of Claim 25, wherein said cDNA comprises the full protein coding sequence partially included in of one of the sequences of SEQ ED NOs: 38-305.
27. A method of making a protein comprising one of the sequences of SEQ ID NO: 306-573, comprising the steps of: obtaining a cDNA encoding the full protein sequence partially included in one of the sequences of sequence of SEQ ED NO : 38-305 ; inserting said cDNA in an expression vector such that said cDNA is operably linked to a promoter; introducing said expression vector into a host cell whereby said host cell produces the protein encoded by said cDNA; and isolating said protein.
28. An isolated protein obtainable by the method of Claim 27.
29. A method of obtaining a promoter DNA comprising the steps of: obtaining DNAs located upstream of the nucleic acids of SEQ D NO: 38-305 or the sequences complementary thereto; screening said upstream DNAs to identify a promoter capable of directing transcription initiation; and isolating said DNA comprising said identified promoter.
30. The method of Claim 29, wherein said obtaining step comprises chromosome walking from said nucleic acids of SEQ ED NO: 38-305 or sequences complementary thereto.
31. The method of Claim 30, wherein said screening step comprises inserting said upstream sequences into a promoter reporter vector.
32. The method of Claim 30, wherein said screening step comprises identifying motifs in said upstream DNAs which are transcription factor binding sites or transcription start sites.
33. An isolated promoter obtainable by the method of Claim 32.
34. An isolated or purified protein comprising one of the sequences of SEQ ED NO: 306-573.
35. In an array of discrete ESTs or fragments thereof of at least 15 nucleotides in length, the improvement comprising inclusion in said array of at least one of the sequences of SEQ ED NOs: 38-305, or one of the sequences complementary to the sequences of SEQ ED NOs: 38-305, or a fragment thereof of at least 15 consecutive nucleotides.
36. The array of Claim 35 including therein at least two of the sequences of SEQ ED NOs: 38-305, the sequences complementary to the sequences of SEQ ED NOs: 38-305, or fragments thereof of at least 15 consecutive nucleotides.
37. The array of Claim 35 including therein at least five of the sequences of SEQ
ID NOs: 38-305, the sequences complementary to the sequences of SEQ ID NOs: 38-305, or fragments thereof of at least 15 consecutive nucleotides.
EP98936596A 1997-08-01 1998-07-31 5' ESTs FOR SECRETED PROTEINS EXPRESSED IN MUSCLE AND OTHER MESODERMAL TISSUES Withdrawn EP1000152A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP03016881A EP1367124A1 (en) 1997-08-01 1998-07-31 5' ests for secreted proteins expressed in muscle and other mesodermal tissues

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US90513497A 1997-08-01 1997-08-01
US905134 1997-08-01
PCT/IB1998/001238 WO1999006554A2 (en) 1997-08-01 1998-07-31 5' ESTs FOR SECRETED PROTEINS EXPRESSED IN MUSCLE AND OTHER MESODERMAL TISSUES

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP03016881A Division EP1367124A1 (en) 1997-08-01 1998-07-31 5' ests for secreted proteins expressed in muscle and other mesodermal tissues

Publications (1)

Publication Number Publication Date
EP1000152A2 true EP1000152A2 (en) 2000-05-17

Family

ID=25420335

Family Applications (2)

Application Number Title Priority Date Filing Date
EP98936596A Withdrawn EP1000152A2 (en) 1997-08-01 1998-07-31 5' ESTs FOR SECRETED PROTEINS EXPRESSED IN MUSCLE AND OTHER MESODERMAL TISSUES
EP03016881A Withdrawn EP1367124A1 (en) 1997-08-01 1998-07-31 5' ests for secreted proteins expressed in muscle and other mesodermal tissues

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP03016881A Withdrawn EP1367124A1 (en) 1997-08-01 1998-07-31 5' ests for secreted proteins expressed in muscle and other mesodermal tissues

Country Status (5)

Country Link
EP (2) EP1000152A2 (en)
JP (1) JP2001512016A (en)
AU (1) AU8555798A (en)
CA (1) CA2297109A1 (en)
WO (1) WO1999006554A2 (en)

Families Citing this family (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6416973B1 (en) 1997-08-01 2002-07-09 Schering Corporation Nucleic acids encoding mammalian cell membrane protein MDL-1
DE69836082T2 (en) * 1997-08-01 2007-05-10 Schering Corp. MEMBRANE PROTEINS FROM MAMMALIAN CELLS; RELATED REAGENTS
EP1012285B1 (en) * 1997-09-10 2005-08-03 ZymoGenetics, Inc. Beta-defensins
US6576755B1 (en) 1997-09-10 2003-06-10 Zymogenetics, Inc. Beta-defensins
EP1627886A1 (en) * 1997-10-07 2006-02-22 Ono Pharmaceutical Co., Ltd. Polypeptide, cDNA encoding the same, and use of them
DE69942782D1 (en) 1998-11-27 2010-10-28 Ucb Sa Compositions and methods for increasing bone mineralization
US20040009535A1 (en) 1998-11-27 2004-01-15 Celltech R&D, Inc. Compositions and methods for increasing bone mineralization
EP1135494A2 (en) * 1998-12-11 2001-09-26 Incyte Pharmaceuticals, Inc. Neuron-associated proteins
US7078203B1 (en) 1999-03-31 2006-07-18 The University Of North Carolina At Chapel Hill Isolated DNA encoding cullin regulators ROC1 and ROC2, isolated proteins encoded by the same, and methods utilizing the same
WO2000058472A2 (en) * 1999-03-31 2000-10-05 The University Of North Carolina At Chapel Hill Isolated dna encoding cullin regulators roc1 and roc2, isolated proteins encoded by the same, and methods utilizing the same
US20030082586A1 (en) 1999-06-29 2003-05-01 Millennium Pharmaceuticals, Inc. Antibodies having diagnostic, preventive, therapeutic, and other uses
US20020037513A1 (en) * 2000-03-24 2002-03-28 Rosana Kapeller-Libermann 33338, a novel human ubiquitin hydrolase-like molecule and uses thereof
US6979551B2 (en) 2000-04-03 2005-12-27 Rigel Pharmaceuticals, Inc. Assays for identifying ubiquitin agents and for identifying agents that modify the activity of ubiquitin agents
US6740495B1 (en) 2000-04-03 2004-05-25 Rigel Pharmaceuticals, Inc. Ubiquitin ligase assay
EP1285060A2 (en) 2000-04-26 2003-02-26 Millennium Pharmaceuticals, Inc. 26335, a novel human serine/threonine dehydratase and uses thereof
AU2001252621A1 (en) * 2000-04-27 2001-11-12 Takeda Chemical Industries Ltd. Novel proetin and use thereof
JP2002335964A (en) * 2000-04-27 2002-11-26 Takeda Chem Ind Ltd New protein and application thereof
US7927597B2 (en) 2001-04-10 2011-04-19 Agensys, Inc. Methods to inhibit cell growth
WO2002083921A2 (en) 2001-04-10 2002-10-24 Agensys, Inc. Nuleic acids and corresponding proteins useful in the detection and treatment of various cancers
WO2003020897A2 (en) * 2001-08-31 2003-03-13 Diadexus, Inc. Compositions and methods relating to ovary specific genes and proteins
WO2003087763A2 (en) 2002-04-03 2003-10-23 Celltech R & D, Inc. Association of polymorphisms in the sost gene region with bone mineral density
EA015166B1 (en) 2003-06-16 2011-06-30 Ю-Си-Би Мэньюфэкчуринг, Инк. Immunogen sclerostin peptides (sost), inducing formation of specific antibodies
US7592429B2 (en) 2005-05-03 2009-09-22 Ucb Sa Sclerostin-binding antibody
US8003108B2 (en) 2005-05-03 2011-08-23 Amgen Inc. Sclerostin epitopes
CL2008002775A1 (en) 2007-09-17 2008-11-07 Amgen Inc Use of a sclerostin binding agent to inhibit bone resorption.
HUE033315T2 (en) 2010-05-14 2017-11-28 Amgen Inc High concentration antibody formulations
CN103649118A (en) 2011-03-01 2014-03-19 安进公司 Bispecific binding agents
AU2012236962B2 (en) 2011-03-25 2017-05-11 Amgen Inc. Anti-sclerostin antibody crystals and formulations thereof
WO2013019954A1 (en) 2011-08-04 2013-02-07 Amgen Inc. Method for treating bone gap defects
CA2858974A1 (en) 2011-12-28 2013-07-04 Amgen Inc. Method of treating alveolar bone loss through the use of anti-sclerostin antibodies
EP3626267A1 (en) 2012-07-05 2020-03-25 UCB Pharma, S.A. Treatment for bone diseases
UY35148A (en) 2012-11-21 2014-05-30 Amgen Inc HETERODIMERIC IMMUNOGLOBULINS
MA41142A (en) 2014-12-12 2017-10-17 Amgen Inc ANTI-SCLEROSTINE ANTIBODIES AND THE USE OF THEM TO TREAT BONE CONDITIONS AS PART OF THE TREATMENT PROTOCOL
GB201604124D0 (en) 2016-03-10 2016-04-27 Ucb Biopharma Sprl Pharmaceutical formulation
KR20200138254A (en) 2018-03-30 2020-12-09 암젠 인크 C-terminal antibody variant
CN110845572B (en) * 2019-11-28 2023-03-21 中国药科大学 Tumor-targeted GRP (glass-fiber reinforced Polypropylene) analogue and application thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3337748B2 (en) * 1992-09-25 2002-10-21 財団法人神奈川科学技術アカデミー Method for synthesizing full-length cDNA, method for producing intermediate thereof, and method for producing recombinant vector containing full-length cDNA
FR2733762B1 (en) * 1995-05-02 1997-08-01 Genset Sa METHOD FOR THE SPECIFIC COUPLING OF THE HAIR OF THE 5 'END OF A RNAM FRAGMENT AND PREPARATION OF RNAM AND COMPLETE DNA
US5707829A (en) * 1995-08-11 1998-01-13 Genetics Institute, Inc. DNA sequences and secreted proteins encoded thereby
WO1998042738A1 (en) * 1997-03-21 1998-10-01 Human Genome Sciences, Inc. 87 human secreted proteins
US5871970A (en) * 1997-03-31 1999-02-16 Incyte Pharmaceuticals, Inc. Calcium-binding protein

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9906554A3 *

Also Published As

Publication number Publication date
WO1999006554A2 (en) 1999-02-11
EP1367124A1 (en) 2003-12-03
AU8555798A (en) 1999-02-22
JP2001512016A (en) 2001-08-21
CA2297109A1 (en) 1999-02-11
WO1999006554B1 (en) 2001-05-31
WO1999006554A3 (en) 1999-05-27

Similar Documents

Publication Publication Date Title
EP1000146B1 (en) 5'ESTs FOR NON TISSUE SPECIFIC SECRETED PROTEINS
EP1000150B1 (en) 5' ESTs FOR SECRETED PROTEINS EXPRESSED IN BRAIN
EP1037977B1 (en) EXTENDED cDNAs FOR SECRETED PROTEINS
EP1378571B1 (en) 5' ESTs for secreted proteins expressed in various tissues
US6936692B2 (en) Complementary DNAs
EP1367124A1 (en) 5' ests for secreted proteins expressed in muscle and other mesodermal tissues
WO1999006550A2 (en) 5' ESTs FOR SECRETED PROTEINS EXPRESSED IN PROSTATE
EP1000149B1 (en) 5' ESTs FOR SECRETED PROTEINS IDENTIFIED FROM BRAIN TISSUES
EP1000151B1 (en) 5' ESTs FOR SECRETED PROTEINS EXPRESSED IN VARIOUS TISSUES
EP0994899A2 (en) 5' ESTs FOR SECRETED PROTEINS EXPRESSED IN ENDODERM
AU2006201603B2 (en) Extended cDNAs for secreted proteins
AU2003262114B8 (en) cDNAs encoding secreted proteins
EP1757699A1 (en) cDNAs encoding secreted proteins
EP1903111A2 (en) Extended cDNAs for secreted proteins
AU2006225329A1 (en) cDNAs encoding secreted proteins

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20000224

AK Designated contracting states

Kind code of ref document: A2

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

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: GENSET

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: SERONO GENETICS INSTITUTE S.A.

17Q First examination report despatched

Effective date: 20080213

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

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

18D Application deemed to be withdrawn

Effective date: 20080826