US20130035260A1 - Genes involved in inflammatory bowel diseases and use thereof - Google Patents

Genes involved in inflammatory bowel diseases and use thereof Download PDF

Info

Publication number
US20130035260A1
US20130035260A1 US13/406,487 US201213406487A US2013035260A1 US 20130035260 A1 US20130035260 A1 US 20130035260A1 US 201213406487 A US201213406487 A US 201213406487A US 2013035260 A1 US2013035260 A1 US 2013035260A1
Authority
US
United States
Prior art keywords
nucleic acid
seq
sequence
gene
acid probe
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/406,487
Inventor
Jean Pierre Hugot
Gilles Thomas
Mohamed Zouali
Suzanne Lesage
Mathias Chamaillard
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.)
Fondation Jean Dausset-CEPH
Original Assignee
Fondation Jean Dausset-CEPH
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 Fondation Jean Dausset-CEPH filed Critical Fondation Jean Dausset-CEPH
Priority to US13/406,487 priority Critical patent/US20130035260A1/en
Publication of US20130035260A1 publication Critical patent/US20130035260A1/en
Assigned to FONDATION JEAN DAUSSET-CEPH reassignment FONDATION JEAN DAUSSET-CEPH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHAMAILLARD, MATHIAS, HUGOT, JEAN-PIERRE, LESAGE, SUZANNE, THOMAS, GILLES, ZOUALI, MOHAMED
Abandoned legal-status Critical Current

Links

Images

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4747Apoptosis related proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention relates to genes involved in inflammatory and/or immune diseases and certain cancers, in particularly cryptogenetic inflammatory bowel diseases, and also to the proteins encoded by these genes.
  • the present invention also relates to methods for diagnosing inflammatory diseases.
  • Cryptogenetic inflammatory bowel diseases are diseases characterized by an inflammation of the digestive tract, the cause of which is unknown. Depending on the location and the characteristics of the inflammation, two different nosological entities are distinguished: ulcerative colitis (UC) and Crohn's disease (CD). UC was described by S Wilkes in 1865, whereas the first case of regional ileitis was reported by Crohn in 1932. In reality, it is possible that these two diseases go back much further.
  • IBDs are chronic diseases which evolve throughout life and which affect approximately 1 to 2 individuals per 1000 inhabitants in western countries, which represents between 60000 and 100000 individuals suffering from these diseases in France. They are diseases which appear in young individuals (peak instance is in the third decade), progressing via attacks interspersed with remissions, with frequent complications such as undernutrition, retarded growth in children, bone demineralization and, in the end, malignant degeneration to colon cancer. No specific treatment exists. Conventional therapeutics make use of anti-inflammatories, of immunosuppressors and of surgery. All these therapeutic means are, themselves, a source of considerable iatrogenic morbidity. For all these reasons, IBDs appear to be a considerable public health problem.
  • the etiology of IBDs is currently unknown.
  • Environmental factors are involved in the occurrence of the disease, as witnessed by the secular increase in incidence of the disease and the incomplete concordance in monozygous twins.
  • the only environmental risk factors currently known are 1) tobacco, the role of which is harmful in CD and beneficial in UC, and 2) appendectomy which has a protective role for UC.
  • IBDs are more common in the Caucasian population, and in particular in the Jewish population of central Europe. Familial forms represent from 6 to 20% of IBD cases. They are particularly common when the disease begins early. However, it is studies in twins which have made it possible to confirm the genetic nature of these diseases. In fact, the concordance rate between twins for these diseases is greater in monozygous twins than in dizygous twins, which pleads strongly in favor of a hereditary component to IBDs, in particular to CD. In all probability, IBDs are complex genetic diseases involving several different genes, interacting with one another and with environmental factors. IBDs can therefore be classified within the context of multifactor diseases.
  • IBD-susceptibility genes Two major strategies have been developed in order to demonstrate the IBD-susceptibility genes. The first is based on the analysis of genes which are candidates for physiopathological reasons. Thus, many genes have been proposed as potentially important for IBDs. They are often genes which have a role in inflammation and the immune response. Mention may be made of the HLA, TAP, TNF and MICA genes, lymphocyte T receptor, ICAM1, interleukin 1, CCR5, etc. Other genes participate in diverse functions, such as GAI2, motilin, MRAMP, HMLH1, etc. In reality, none of the various candidate genes studied has currently definitively proved itself to have a role in the occurrence of IBDs.
  • the present invention relates to the demonstration of the nucleic acid sequence of genes involved in IBDs, and other inflammatory diseases, and also the use of these nucleic acid sequences.
  • CD-susceptibility gene is located in the pericentromeric region of chromosome 16 ( FIG. 1 ). It was the first gene for susceptibility to a complex genetic disease located by positional cloning and satisfying the strict criteria proposed in the literature (Lander and Kruglyak, 1995). This gene was named IBD1 (for inflammatory bowel disease 1). Since then, other locations have been proposed by other authors, in particular on chromosomes 12, 1, 3, 6 and 7 (Satsangi et al., 1996; Cho et al., 1998). Although they have been located, it has currently not been possible to identify any of these IBD-susceptibility genes.
  • chromosome 16 for several immune and inflammatory diseases, such as ankylosing spondylarthritis, Blau's syndrome, psoriasis, etc. (Becker et al., 1998; Tromp et al., 1996). All these diseases may then share the same gene (or the same group of genes) located on chromosome 16.
  • genes known to be in the pericentromeric region of chromosome 16 such as the interleukin 4 receptor, CD19, CD43 or CD11, appear to be good potential candidates for CD. Preliminary results do not however plead in favor of these genes being involved in CD.
  • the present invention provides not only the sequence of IBD1 gene, but also the partial sequence of another gene, called IBD1prox due to it being located in proximity to IBD, and demonstrated as reported in the examples below.
  • IBD1prox due to it being located in proximity to IBD, and demonstrated as reported in the examples below.
  • the peptide sequence expressed by the IBD1 and IBD1prox genes is represented by SEQ ID No. 2 and SEQ ID No. 5, respectively; the genomic sequence of these genes is represented by SEQ ID No. 3 and SEQ ID No. 6, respectively.
  • a subject of the present invention is a purified or isolated nucleic acid, characterized in that it Comprises a nucleic acid sequence chosen from the following group of sequences:
  • nucleic acid sequence according to the invention defined in c) has a percentage identity of at least 80%, after optimal alignment, with a sequence as defined in a) or b) above, preferably 90%, most preferably 98%.
  • nucleic acid refers to any suitable nucleic acid sequence
  • polynucleotide refers to any suitable nucleic acid sequence
  • oligonucleotide refers to any suitable nucleotide sequence
  • polynucleotide sequence refers to any suitable sequence of nucleic acids, which may or may not be modified, making it possible to define a fragment or a region of a nucleic acid, which may or may not comprise unnatural nucleotides, and which may correspond equally to a double-stranded DNA, a single-stranded DNA and transcription products of said DNAs.
  • the nucleic acid sequences according to the invention also encompass PNAs (Peptide Nucleic Acids), or the like.
  • nucleotide sequences in their natural chromosomal environment that is to say in the natural state. They are sequences which have been isolated and/or purified, that is to say they have been taken directly or indirectly, for example by copying, their environment having been at least partially modified.
  • nucleic acids obtained by chemical synthesis are also intended to be denoted.
  • the term “percentage identity” between two nucleic acid or amino acid sequences is intended to denote a percentage of nucleotides or of amino acid residues which are identical between the two sequences to be compared, obtained after the best alignment, this percentage being purely statistical and the differences between the two sequences being distributed randomly and over their entire length.
  • the term “best alignment” or “optimal alignment” is intended to denote the alignment for which the percentage identity determined as below is highest. Sequence comparisons between two nucleic acid or amino acid sequences are conventionally carried out by comparing these sequences after having aligned them optimally, said comparison being carried out by segment or by “window of comparison” so as to identify and compare local regions of sequence similarity.
  • the optimal alignment of the sequences for the comparison may be carried out, besides manually, by means of the local homology algorithm of Smith and Waterman (1981), by means of the local homology algorithm of Neddleman and Wunsch (1970), by means of the similarity search method of Pearson and Lipman (1988), by means of computer programs using these algorithms (GAP, BESTFIT, BLAST P, BLAST N, FASTA and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.).
  • the BLAST program is preferably used, with the BLOSUM 62 matrix.
  • the PAM or PAM250 matrices may also be used.
  • the percentage identity between two nucleic acid or amino acid sequences is determined by comparing these two sequences aligned optimally, the nucleic acid or amino acid sequence to be compared possibly comprising additions or deletions with respect to the reference sequence for optimal alignment between these two sequences.
  • the percentage identity is calculated by determining the number of identical positions for which the nucleotide or the amino acid residue is identical between the two sequences, dividing this number of identical positions by the total number of positions compared and multiplying the resultant number by 100 so as to obtain the percentage identity between these two sequences.
  • nucleic acid sequences having a percentage identity of at least 80%, preferably 90%, more preferably 98%, after optimal alignment with a reference sequence is intended to denote the nucleic acid sequences which, compared to the reference nucleic acid sequence, have certain modifications, such as in particular a deletion, a truncation, an extension, a chimeric fusion and/or a substitution, in particular of the point type, and the nucleic acid sequence of which exhibits at least 80%, preferably 90%, more preferably 98%, identity, after optimal alignment, with the reference nucleic acid sequence.
  • They are preferably sequences whose complementary sequences are capable of hybridizing specifically with the sequence SEQ ID No. 1 or SEQ ID No. 4 of the invention.
  • the specific or high stringency hybridization conditions will be such that they ensure at least 80%, preferably 90%, more preferably 98%, identity, after optimal alignment, between one of the two sequences and the sequence complementary to the other.
  • Hybridization under high stringency conditions means that the conditions of temperature and of ionic strength are chosen such that they allow the hybridization between two complementary DNA fragments to be maintained.
  • high stringency conditions for the hybridization step for the purposes of defining the polynucleotide fragments described above are advantageously as follows.
  • the DNA-DNA or DNA-RNA hybridization is carried out in two steps: (1) prehybridization at 42° C. for 3 hours in phosphate buffer (20 mM, pH 7.5) containing 5 ⁇ SSC (1 ⁇ SSC corresponds to a solution of 0.15 M NaCl+0.015 M sodium citrate), 50% of formamide, 7% of sodium dodecyl sulfate (SDS), 10 ⁇ Denhardt's, 5% of dextran sulfate and 1% of salmon sperm DNA; (2) hybridization per se for 20 hours at a temperature which depends on the length of the probe (i.e.: 42° C. for a probe >100 nucleotides in length), followed by 2 washes of 20 minutes at 20° C.
  • nucleic acid sequences having a percentage identity of at least 80%, preferably 90%, more preferably 98%, after optimal alignment, with the sequence according to the invention preference is also given to the variant nucleic acid sequences of SEQ ID No. 1 or of SEQ ID No. 4, or of fragments thereof, that is to say all the nucleic acid sequences corresponding to allelic variants, that is to say individual variations of the sequence SEQ ID No. 1 or SEQ ID No. 4.
  • These natural mutated sequences correspond to polymorphisms present in mammals, in particular in humans and, in particular, to polymorphisms which may lead to the occurrence of a pathological condition.
  • the present invention relates to the variant nucleic acid sequences in which the mutations lead to a modification of the amino acid sequence of the polypeptide, or of fragments thereof, encoded by the normal sequence of SEQ ID No. 1 or SEQ ID No. 4.
  • variant nucleic acid sequence is also intended to denote any RNA or cDNA resulting from a mutation and/or variation of a splice site of the genomic nucleic acid sequence the cDNA of which has the sequence SEQ ID No. 1 or SEQ ID No. 4.
  • the invention preferably relates to a purified or isolated nucleic acid according to the present invention, characterized in that it comprises or consists of one of the sequences SEQ ID No. 1 or SEQ ID No. 4, of the sequences complementary thereto, or of the RNA sequences corresponding to SEQ ID No. 1 or SEQ ID No. 4.
  • the probes or primers characterized in that they comprise a sequence of a nucleic acid according to the invention, are also part of the invention.
  • the present invention also relates to the primers or the probes according to the invention which may make it possible in particular to demonstrate or to distinguish the variant nucleic acid sequences, or to identify the genomic sequence of the genes the cDNA of which is represented by SEQ ID No. 1 or SEQ ID No. 4, in particular using an amplification method such as the PCR method or a related method.
  • the invention also relates to the use of a nucleic acid sequence according to the invention, as a probe or primer, for detecting, identifying, assaying or amplifying a nucleic acid sequence.
  • the polynucleotides which can be used as a probe or as a primer in methods for detecting, identifying, assaying or amplifying a nucleic acid sequence are a minimum of 15 bases, preferably 20 bases, or better still 25 to 30 bases in length.
  • the probes and primers according to the invention may be labeled directly or indirectly with a radioactive or nonradioactive compound using methods well known to those skilled in the art, in order to obtain a detectable and/or quantifiable signal.
  • polynucleotide sequences according to the invention which are unlabeled can be used directly as a probe or primer.
  • sequences are generally labeled so as to obtain sequences which can be used in many applications.
  • the primers or the probes according to the invention are labeled with radioactive elements or with nonradio-active molecules.
  • the nonradioactive entities are selected from ligands such as biotin, avidin, streptavidin or dioxygenin, haptens, dyes and luminescent agents, such as radioluminescent, chemiluminescent, bioluminescent, fluorescent or phosphorescent agents.
  • the polynucleotides according to the invention may thus be used as a primer and/or probe in methods using in particular the PCR (polymerase chain reaction) technique (Rolfs et al., 1991).
  • This technique requires choosing pairs of oligonucleotide primers bordering the fragment which must be amplified.
  • the amplified fragments can be identified, for example after agarose or polyacrylamide gel electrophoresis, or after a chromatographic technique such as gel filtration or ion exchange chromatography, and then sequenced.
  • the specificity of the amplification can be controlled using, as primers, the nucleotide sequences of polynucleotides of the invention and, as matrices, plasmids containing these sequences or else the derived amplification products.
  • the amplified nucleotide fragments may be used as reagents in hybridization reactions in order to demonstrate the presence, in a biological sample, of a target nucleic acid of sequence complementary to that of said amplified nucleotide fragments.
  • the invention is also directed toward the nucleic acids which can be obtained by amplification using primers according to the invention.
  • PCR-like is intended to denote all the methods using direct or indirect reproductions of nucleic acid sequences, or else in which the labeling systems have been amplified; these techniques are of course, known. In general, they involve amplifying the DNA with a polymerase; when the sample of origin is an RNA a reverse transcription should be carried out beforehand.
  • the target polynucleotide to be detected is an mRNA
  • an enzyme of the reverse transcriptase type is advantageously used, prior to carrying out an amplification reaction using the primers according to the invention or to carrying out a method of detection using the probes of the invention, in order to obtain a cDNA from the mRNA contained in the biological sample.
  • the cDNA obtained will then serve as a target for the primers or the probes used in the amplification or detection method according to the invention.
  • the probe hybridization technique may be carried out in many ways (Matthews et al., 1988).
  • the most general method consists in immobilizing the nucleic acid extracted from the cells of various tissues or from cells in culture, on a support (such as nitrocellulose, nylon or polystyrene), and in incubating the immobilized target nucleic acid with the probe, under well-defined conditions. After hybridization, the excess probe is removed and the hybrid molecules formed are detected using the appropriate method (measuring the radioactivity, the fluorescence or the enzymatic activity linked to the probe).
  • the latter may be used as capture probes.
  • a probe termed “capture probe”
  • capture probe is immobilized on a support and is used to capture, by specific hybridization, the target nucleic acid obtained from the biological sample to be tested, and the target nucleic acid is then detected using a second probe, termed “detection probe”, labeled with a readily detectable element.
  • antisense oligonucleotides i.e. oligonucleotides
  • the structure of which ensures, by hybridization with the target sequence, inhibition of expression of the corresponding product Mention should also be made of sense oligonucleotides, which, by interacting with proteins involved in regulating the expression of the corresponding product, will induce either inhibition or activation of this expression.
  • the oligo-nucleotides of the invention may be used in vitro and in vivo.
  • the present invention also relates to an isolated polypeptide, characterized in that it comprises a polypeptide chosen from:
  • polypeptide is intended to denote proteins or peptides.
  • biologically active fragment is intended to mean a fragment having the same biological activity as the peptide fragment from which it is deduced, preferably within the same order of magnitude (to within a factor of 10).
  • the examples show that the IBD1 protein (SEQ ID No. 2) has a potential role in apoptosis phenomena.
  • a biologically active fragment of the IBD1 protein therefore consists of a polypeptide derived from SEQ ID No. 2, also having a role in apoptosis.
  • the examples below propose biological functions for the IBD1 and IBD1prox proteins, as a function of the peptide domains of these proteins, and thus allow those skilled in the art to identify the biologically active fragments.
  • a polypeptide according to the invention is a polypeptide consisting of the sequence SEQ ID No. 2 (corresponding to the protein encoded by the IBD1 gene) or of the sequence SEQ ID No. 5 (corresponding to the protein encoded by IBD1prox) or of a sequence having at least 80% identity with SEQ ID No. 2 or SEQ ID No. 5 after optimal alignment.
  • sequence of the polypeptide has a percentage identity of at least 80%, after optimal alignment, with the sequence SEQ ID No. 2 or SEQ ID No. 5, preferably 90%, more preferably 98%.
  • polypeptide the amino acid sequence of which has a percentage identity of at least 80%, preferably 90%, more preferably 98%, after optimal alignment, with a reference sequence
  • polypeptides having certain modifications compared to the reference polypeptide such as in particular one or more deletions and/or truncations, an extension, a chimeric fusion and/or one or more substitutions.
  • polypeptides the amino acid sequence of which has a percentage identity of at least 80%, preferably 90%, more preferably 98%, after optimal alignment, with the sequence SEQ ID No. 2 or SEQ ID No. 5 or with a fragment thereof according to the invention
  • the present invention also relates to the cloning and/or expression vectors comprising a nucleic acid or encoding a polypeptide according to the invention.
  • a vector may also contain the elements required for the expression and, optionally, the secretion of the polypeptide in a host cell.
  • a host cell is also a subject of the invention.
  • vectors characterized in that they comprise a promoter and/or regulator sequence according to the invention are also part of the invention.
  • Said vectors preferably comprise a promoter, translation initiation and termination signals, and also regions suitable for regulating transcription. It must be possible for them to be maintained stably in the cell and they may optionally contain particular signals specifying secretion of the translated protein.
  • nucleic acid sequences according to the invention may be inserted into vectors which replicate autonomously in the chosen host, or vectors which integrate in the chosen host.
  • the viral vectors possibly being in particular adenoviruses (Perricaudet et al., 1992), retroviruses, lentiviruses, poxviruses or herpesviruses (Epstein et al., 1992).
  • adenoviruses Perricaudet et al., 1992
  • retroviruses retroviruses
  • lentiviruses lentiviruses
  • poxviruses poxviruses
  • herpesviruses Epstein et al., 1992
  • viruses are, for example, retroviruses (Temin, 1986), or AAVs (Carter, 1993).
  • naked polynucleotides such as naked DNA or naked RNA according to the technology developed by the company VICAL, bacterial artificial chromosomes (BACs), yeast artificial chromosomes (YACs) for expression in yeast, mouse artificial chromosomes (MACs) for expression in murine cells and, preferably, human artificial chromosomes (HACs) for expression in human cells.
  • BACs bacterial artificial chromosomes
  • YACs yeast artificial chromosomes
  • MACs mouse artificial chromosomes
  • HACs human artificial chromosomes
  • Such vectors are prepared according to the methods commonly used by those skilled in the art, and the clones resulting therefrom can be introduced into a suitable host using standard methods, such as, for example, lipofection, electroporation, heat shock, transformation after chemical permeabilization of the membrane, or cell fusion.
  • the invention also comprises host cells, in particular the eukaryotic and prokaryotic cells, transformed with the vectors according to the invention, and also the transgenic animals, preferably the mammals, except humans, comprising one of said transformed cells according to the invention.
  • These animals may be used as models, for studying the etiology of inflammatory and/or immune diseases, in particular of the inflammatory diseases of the digestive tract, or for studying cancers.
  • bacterial cells which can be used for the purpose of the present invention
  • yeast cells Buckholz, 1993
  • animal cells in particular mammalian cell cultures
  • mammalian cell cultures Edwards and Aruffo, 1993
  • Chinese hamster ovary CHO
  • insect cells in which it is possible to use methods employing, for example, baculo viruses (Luckow, 1993).
  • a preferred cellular host for expressing the proteins of the invention consists of COS cells.
  • mammals according to the invention animals such as rodents, in particular mice, rats or rabbits, expressing a polypeptide according to the invention are preferred.
  • mammals preference is also given to animals such as mice, rats or rabbits, characterized in that the gene encoding the protein of sequence SEQ ID No. 2 or SEQ ID No. 5, or the sequence of which is encoded by the homologous gene in these animals, is not functional, has been knocked out or has at least one mutation.
  • transgenic animals are obtained, for example, by homologous recombination on embryonic stem cells, transfer of these stem cells to embryos, selection of the chimeras affected in the reproductive lines, and growth of said chimeras.
  • the transgenic animals according to the invention may thus overexpress the gene encoding the protein according to the invention, or their homologous gene, or express said gene into which a mutation is introduced.
  • These transgenic animals in particular mice, are obtained, for example, by transfection of a copy of this gene under the control of a promoter which is strong and ubiquitous, or selective for a tissue type, or after viral transcription.
  • the transgenic animals according to the invention may be made deficient for the gene encoding one of the polypeptides of sequence SEQ ID No. 2 or SEQ ID No. 5, or their homologous genes, by inactivation using the LOXP/CRE recombinase system (Rohlmann et al., 1996) or any other system for inactivating the expression of this gene.
  • the cells and mammals according to the invention can be used in a method for producing a polypeptide according to the invention, as described below, and may also be used as a model for analysis.
  • the cells or mammals transformed as described above can also be used as models in order to study the interactions between the polypeptides according to the invention, and the chemical or protein compounds involved directly or indirectly in the activities of the polypeptides according to the invention, this being in order to study the various mechanisms and interactions involved.
  • polypeptides according to the invention may in particular be used for selecting products which interact with the polypeptides according to the invention, in particular the protein of sequence SEQ ID No. 2 or SEQ ID No. 5 or variants thereof according to the invention, as a cofactor or as an inhibitor, in particular a competitive inhibitor, or which have an agonist or antagonist activity with respect to the activity of the polypeptides according to the invention.
  • said transformed cells or transgenic animals are used as a model in particular for selecting products for combating pathological conditions associated with abnormal expression of this gene.
  • the invention also relates to the use of a cell, of a mammal or of a polypeptide according to the invention, for screening chemical or biochemical compounds which may interact directly or indirectly with the polypeptides according to the invention, and/or which are capable of modulating the expression or the activity of these polypeptides.
  • the invention also relates to a method for screening compounds capable of interacting, in vitro or in vivo, with a nucleic acid according to the invention, using a nucleic acid, a cell or a mammal according to the invention, and detecting the formation of a complex between the candidate compounds and the nucleic acid according to the invention.
  • the invention also relates to the use of a nucleic acid sequence according to the invention, for synthesizing recombinant polypeptides.
  • the method for producing a polypeptide of the invention in recombinant form is characterized in that the transformed cells, in particular the cells or mammals of the present invention, are cultured under conditions which allow the expression of a recombinant polypeptide encoded by a nucleic acid sequence according to the invention, and in that said recombinant polypeptide is recovered.
  • the recombinant polypeptides characterized in that they can be obtained using said method of production, are also part of the invention.
  • the recombinant polypeptides obtained as indicated above can be in both glycosylated and nonglycosylated form, and may or may not have the natural tertiary structure.
  • sequences of the recombinant polypeptides may also be modified in order to improve their solubility, in particular in aqueous solvents.
  • polypeptides may be produced using the nucleic acid sequences defined above, according to the techniques for producing recombinant polypeptides known to those skilled in the art.
  • the nucleic acid sequence used is placed under the control of signals which allow its expression in a cellular host.
  • An effective system for producing a recombinant polypeptide requires having a vector and a host cell according to the invention.
  • These cells can be obtained by introducing into host cells a nucleotide sequence inserted into a vector as defined above, and then culturing said cells under conditions which allow the replication and/or expression of the transfected nucleotide sequence.
  • the methods used for purifying a recombinant polypeptide are known to those skilled in the art.
  • the recombinant polypeptide may be purified from cell lysates and extracts or from the culture medium supernatant, by methods used individually or in combination, such as fractionation, chromatography methods, immunoaffinity techniques using specific monoclonal or polyclonal antibodies, etc.
  • polypeptides according to the present invention can also be obtained by chemical synthesis using one of the many known forms of peptide synthesis, for example techniques using solid phases (see in particular Stewart et al., 1984) or techniques using partial solid phases, by fragment condensation or by conventional synthesis in solution.
  • polypeptides obtained by chemical synthesis and which may comprise corresponding unnatural amino acids are also included in the invention.
  • the mono- or polyclonal antibodies, or fragments thereof, chimeric antibodies or immunoconjugates, characterized in that they are capable of specifically recognizing a polypeptide according to the invention, are part of the invention.
  • polyclonal antibodies may be obtained from a serum of an animal immunized against the polypeptides according to the invention, in particular produced by genetic recombination or by peptide synthesis, according to the usual procedures.
  • the mono- or polyclonal antibodies, or fragments thereof, chimeric antibodies or immunoconjugates characterized in that they are capable of specifically recognizing the polypeptides of sequence SEQ ID No. 2 or SEQ ID No. 5 are particularly preferred.
  • the specific monoclonal antibodies may be obtained according to the conventional method of hybridoma culture described by Köhler and Milstein (1975).
  • the antibodies according to the invention are, for example, chimeric antibodies, humanized antibodies, or Fab or F(ab′) 2 fragments. They may also be in the form of immunoconjugates or of labeled antibodies, in order to obtain a detectable and/or quantifiable signal.
  • the invention also relates to methods for detecting and/or purifying a polypeptide according to the invention, characterized in that they use an antibody according to the invention.
  • the invention also comprises purified polypeptides, characterized in that they are obtained using a method according to the invention.
  • the antibodies of the invention in particular the monoclonal antibodies, may also be used for detecting these polypeptides in a biological sample.
  • polypeptides according to the invention in particular the polypeptides of sequence SEQ ID No. 2 or SEQ ID No. 5, or a variant thereof, on specific tissue sections, for example using immunofluorescence, gold labeling and/or enzymatic immunoconjugates.
  • polypeptides may in particular make it possible to demonstrate abnormal expression of these polypeptides in the biological specimens or tissues.
  • the antibodies of the invention may advantageously be used in any situation where the expression of a polypeptide according to the invention, normal or mutated, must be observed.
  • a method for detecting a polypeptide according to the invention, in a biological sample comprising the Steps of bringing the biological sample into contact with an antibody according to the invention and demonstrating the antigen-antibody complex formed, is also a subject of the invention, as is a kit for carrying out such a method.
  • a kit for carrying out such a method in particular contains:
  • the antibodies according to the invention may also be used in the treatment of an inflammatory and/or immune disease, or of a cancer, in humans, when abnormal expression of the IBD1 gene or of the IBD1prox gene is observed.
  • Abnormal expression means overexpression or the expression of a mutated protein.
  • antibodies may be obtained directly from human serum, or may be obtained from animals immunized with polypeptides according to the invention, and then “humanized”, and may be used as such or in the preparation of a medicinal product intended for the treatment of the abovementioned diseases.
  • the methods for determining an allelic variability, a mutation, a deletion, a loss of heterozygocity or any genetic abnormability of the gene according to the invention characterized in that they use a nucleic acid sequence, a polypeptide or an antibody according to the invention, are also part of the invention.
  • the invention in fact provides the sequence of the IBD1 and IBD1prox genes involved in inflammatory and/or immune diseases, and in particular IBDs.
  • One of the teachings of the invention is to specify the mutations, in these nucleic acid or polypeptide sequences, which are associated with a phenotype corresponding to one of these inflammatory and/or immune diseases.
  • mutations can be detected directly by analysis of the nucleic acid and of the sequences according to the invention (genomic DNA, RNA or cDNA), but also via the polypeptides according to the invention.
  • the use of an antibody according to the invention which recognizes an epitope bearing a mutation makes it possible to distinguish between a “healthy” protein and a protein “associated with a pathological condition”.
  • IBD1 has an important role in a large number of inflammatory and/or immune diseases.
  • IBD1 can be associated with granulomatous inflammatory diseases.
  • Blau's syndrome and CD are in fact diseases which are part of this family. It is therefore hoped that variations in the IBD1 gene will be found for the other diseases of the same family (sarcoidosis, Behcet's disease, etc.).
  • IBD1 in part explain this susceptibility to cancer and define new carcinogenic pathways.
  • the precise description of the mutations which can be observed in the IBD1 gene thus makes it possible to lay down the foundations of a molecular diagnosis for the inflammatory or immune diseases in which this role is demonstrated. Such an approach, based on searching for mutations in the gene, will make it possible to contribute to the diagnosis of these diseases and possibly to reduce the extent of certain additional examinations which are invasive or expensive.
  • the invention lays down the foundations of such a molecular diagnosis based on searching for mutations in IBD1.
  • the molecular diagnosis of inflammatory diseases should also make it possible to improve the nosological classification of these diseases and to more clearly define subgroups of particular diseases by their clinical characteristics, the progressive nature of the disease or the response to certain treatments.
  • the dismantling of the existing mutations may thus make it possible to classify the currently undetermined forms of colitis which represent more than 10% of inflammatory bowel diseases.
  • Such an approach will make it possible to propose an early treatment suitable for each patient.
  • such an approach makes it possible to hope that it will eventually be possible to define an individualized treatment for the disease, depending on the genetic area of each disease, including curative and preventive measures.
  • the genes SEQ ID No. 3 or SEQ ID No. 6 may also be studied.
  • This method of diagnosis and/or of prognostic assessment may be used preventively (a study of predisposition to inflammatory diseases or to cancer), or in order to serve in establishing and/or confirming a clinical condition in a patient.
  • the inflammatory disease is an inflammatory disease of the digestive tract
  • the cancer is a cancer of the digestive tract (small intestine or colon).
  • the teaching of the invention in fact makes it possible to determine the mutations which exhibit a linkage disequilibrium with inflammatory diseases of the digestive tract, and which are therefore associated with such diseases.
  • the analysis may be carried out by sequencing all or part of the gene, or by other methods known to those skilled in the art. Methods based on PCR, for example PCR-SSCP, which makes it possible to detect point mutations, may in particular be used.
  • the analysis may also be carried out by attaching a probe according to the invention, corresponding to one of the sequences SEQ ID No. 1, 3, 4 or 6, to a DNA chip, and hybridization on these microplates.
  • a DNA chip containing a sequence according to the invention is also one of the subjects of the invention.
  • a protein chip containing an amino acid sequence according to the invention is also a subject of the invention.
  • Such a protein chip makes it possible to study the interactions between the polypeptides according to the invention and other proteins or chemical compounds, and may thus be useful for screening compounds which interact with the polypeptides according to the invention.
  • the protein chips according to the invention may also be used to detect the presence of antibodies directed against the polypeptides according to the invention in the serum of patients.
  • a protein chip containing an antibody according to the invention may also be used.
  • RNA in particular by Northern blotting or with RT-PCR experiments, with probes or primers according to the invention
  • protein expressed in particular by Western blotting, using antibodies according to the invention.
  • the gene tested is preferably the gene of sequence SEQ ID No. 1, the inflammatory disease for which the intention is to predict susceptibility being a disease of the digestive tract, in particular Crohn's disease or ulcerative colitis. If the intention is to detect a cancer, it is preferably colon cancer.
  • the invention also relates to methods for obtaining an allele of the IBD1 gene, associated with a detectable phenotype, comprising the following steps:
  • Such a method may be followed by a step of sequencing all or part of the nucleic acid encoding the IBD1 protein, which makes it possible to predict susceptibility to inflammatory disease or of a cancer.
  • the agent capable of specifically detecting a nucleic acid encoding the IBD1 protein is advantageously an oligonucleotide probe according to the invention, which may be made up of DNA, RNA or PNA, which may or may not be modified.
  • the modifications may include radioactive or fluorescent labeling, or may be due to modifications in the bonds between the bases (phosphorothioates or methyl phosphonates, for example).
  • Step b) of the method described above may also be an amplification step as described above.
  • the invention also relates to a method for detecting and/or assaying a nucleic acid according to the invention, in a biological sample, comprising the following steps of bringing a probe according to the invention into contact with a biological sample, and detecting and/or assaying the hybrid formed between said polynucleotide and the nucleic acid of the biological sample.
  • kit of reagents comprising:
  • Such a kit may also contain positive or negative controls in order to ensure the quality of the results obtained.
  • nucleic acid in order to detect and/or assay a nucleic acid according to the invention, those skilled in the art may also perform an amplification step using primers chosen from the sequences according to the invention.
  • the invention also relates to the compounds chosen from a nucleic acid, a polypeptide, a vector, a cell or an antibody according to the invention, or the compounds obtained using the screening methods according to the invention, as a medicinal product, in particular for preventing and/or treating an inflammatory and/or immune disease, or a cancer, associated with the presence of at least one mutation of the gene corresponding to SEQ ID No. 1 or SEQ ID No. 4, preferably an inflammatory disease of the digestive tract, in particular Crohn's disease or ulcerative colitis.
  • FIG. 1 Nonparametric genetic linkage tests for Crohn's disease in the pericentromeric region of chromosome 16 (according to Hugot et al., 1996). Multipoint linkage analysis based on identity by decendance for the markers of the pericentromeric region of chromosome 16. The genetic distances between markers were estimated using the CRIMAP program. The lod score (MAPMAKER/SIBS) is indicated on the left-hand figure. Two pseudoprobability tests were developed and reported on the right-hand figure. The first (Tz) is analogous to the test of the means. The second (Tz2) is analogous to the test of the proportion of affected pairs sharing two alleles.
  • FIG. 3 Diagrammatic representation of the protein encoded by IBD1.
  • the protein encoded by IBD1 is represented horizontally.
  • the various domains of which it is composed are indicated on the figure with the amino acid reference number corresponding to the start and to the end of each domain.
  • the protein consists of a CARD domain, a nucleotide-binding domain (NBD) and leucine-rich motifs (LRR).
  • FIG. 4 Diagrammatic representation of the IBD1/NOD2 protein in three variants associated with CD.
  • the translation produced deduced from the cDNA sequence of the IBD1 candidate gene is identical to that of NOD2 (Ogura et al., 2000).
  • the polypeptide contains 2 CARD domains (CAspase Recruitment Domains), a nucleotide-binding domain (NBD) and 10 repeats of 27 amino acids, leucine-rich motifs (LRR).
  • the consensus sequence of the ATP/GTP-binding site of the motif A (P loop) of the NBD is indicated with a black circle.
  • the sequence changes encoded by the three main variants associated with CD are SNP 8 (R675W), SNP 12 (G881R) and SNP 13 (frame shift 980).
  • the frame shift changes a leucine codon to a proline codon at position 980, which is immediately followed by a stop codon.
  • the first step toward identifying the IBD1 gene was to reduce the size of the genetic region of interest, initially centered on the marker D16S411 located between D16S409 and D16S419 (Hugot et al., 1996 and FIG. 1 ).
  • a group of close markers was used in order to more clearly specify the genetic region, and made it possible to complete the genetic linkage analyses and to search for a genetic linkage disequilibrium with the disease.
  • the families comprising sick individuals suffering from UC were excluded from the study.
  • markers of the micro-satellite type were studied. These markers together made up a high resolution map with an average distance between markers of the order of 1cM in the genetic region of interest. The characteristics of the markers studied are given in table 1.
  • Each marker is listed according to international nomenclature and mostly by the name proposed by the laboratory of origin. The markers appear according to their order on the chromosome (from 16p to 16q). The genetic distance between the markers (in Kosambi centiMorgans, calculated from the experimental data using the Crimap program) is indicated in the second column. The first polymorphic marker is taken randomly as a reference point. The oligonucleotides which were used for the polymerase chain reaction (PCR) are indicated in the third column.
  • the genotyping of these microsatellite markers was based on automatic sequencer technology using fluorescent primers. Briefly, after amplification, the fluorescent polymerase chain reaction (PCR) products were loaded onto a polyacrylamide gel on an automatic sequencer according to the manufacturer's recommendations (Perkin Elmer). The size of the alleles for each individual was deduced using the Genescan® and Genotyper® software. The data were then kept on an integrated computer base containing the genealogical, phenotypic and genetic data. They were then used for the genetic linkage analyses.
  • PCR polymerase chain reaction
  • the typing data for these 26 polymorphism markers were also analyzed so as to search for a transmission disequilibrium.
  • Two groups of 108 and 76 families with one or more sick individuals suffering from CD were studied.
  • the statistical test for transmission disequilibrium has been described by Spielman et al. (1993). In this study, only one sick individual per family was taken into account, and the value of p was corrected by the number of alleles tested for each marker studied.
  • the chromosomal DNA segments were identified based on certain polymorphism markers used in fine genetic mapping (D16S411, D16S416, D16S541, D16S770, D16S2623, D16S3035, D16S3117 and D16S3136).
  • BAC bacterial artificial chromosome
  • the average size of the BACs is 136 kb.
  • the BAC of this contig containing the polymorphism marker D16S3136 (called hb87b10), the size of which was 163761 bp, was sequenced according to the “shotgun” method. Briefly, the BAC DNA was fragmented by sonication. The DNA fragments thus generated were subjected to agarose gel electrophoresis and those with a size greater than 1.5 kb were eluted in order to be analyzed. These fragments were then cloned into the m13 phage, which was itself introduced into bacteria made competent, by electroporation. After culturing, the DNA of the clones was recovered and sequenced by automatic sequencing methods using fluorescent primers of the m13 vector on an automatic sequencer.
  • SNPs biallelic polymorphism markers
  • the typing data were then analyzed using a transmission disequilibrium test (TDT computer program of the GENEHUNTER software version 2).
  • TDT computer program of the GENEHUNTER software version 2 For the families comprising several affected relatives, a single sufferer was taken into account for the analysis. In fact, if several related sufferers are taken into account, this poses the problem of nonindependence of the data in the statistical calculations and can induce an inflation of the value of the test.
  • the sufferer used for the analysis was drawn by lots, within each family, using an automatic randomization procedure. Given this randomization, the value of the statistical test obtained represented only one possible sample derived from the group of families studied. So as not to limit the analysis to this one possible sample, and in order to understand more clearly the soundness of the results obtained, for each test, about one hundred random samples were thus generated and analyzed.
  • the markers were studied separately and then grouped according to their order on the chromosomal segment (KIAA0849ex9 (locus 1), hb27G11F (locus 2), Ctg22Ex1 (locus 3), SNP1 (locus 4), ctg2931-3ac/ola (locus 5), ctg2931-5ag/ola (locus 6), SNP3-2931 (locus 7), Ctg25Ex1 (locus 8), CTG35ExA (locus 9), ctg35ExC (locus 10), d16s3136 (locus 11), hb133D1f (locus 12), D16S3035 (locus 13), ADCY7int7 (locus 14)) (table 2).
  • the haplotypes comprising 2, 3 and 4 consecutive markers were thus analyzed still using the same strategy (100 random samples, taking a single affected individual for each family).
  • haplotypes the most strongly associated with Crohn's disease themselves also extend over this region. Thus, for the majority of the random samples, the transmission test was positive (p ⁇ 0.01) for haplotypes combining the following markers:
  • the susceptibility haplotype most at risk is defined by the loci 7 to 10. This is the haplotype 1-2-1-2 (table 2).
  • the markers tested are, as expected, in linkage disequilibrium with respect to one another.
  • the EST hs135201 (UniGene) made it possible to identify a cDNA not appearing on the available genetic databases (Genbank). It therefore corresponds to a new human gene. Comparison of the sequence of the cDNA and of the genomic DNA showed that this gene consists of 11 exons and 10 introns. An additional exon, positioned 5′ to the cDNA identified, is predicted by analysis of the sequence with the Grail program. These exons are very homologous to the first exons of the CARD4/NOD1 gene. Taking into consideration all of the exons identified and the putative additional exon, this new gene appears to have a genomic structure very close to that of CARD4/NOD1.
  • the cDNA reproduced in the annex (SEQ ID No. 1) therefore comprises all of the identified sequence plus the sequence predicted by the computer modeling, the complementary DNA beginning randomly at the first ATG codon of the predicted coding sequence. On this basis, the gene would therefore comprise 12 exons and 11 introns.
  • the intron-exon structure of the gene is reported on SEQ ID No. 3.
  • the protein sequence deduced from the nucleotide sequence comprises 1041 amino acids (SEQ ID No. 2). This sequence has not been found on the biological databases either (Genpept, pir, swissprot).
  • the IBD1 gene therefore effectively comprises only 11 exons and 10 introns and encodes a protein of 1013 amino acids (i.e. 28 amino acids less than initially determined).
  • the LRR domain of the protein makes it possible to affiliate the protein to a family of proteins involved in intracellular signaling and present both in plants and in animals.
  • CARD4/NOD1 The similarity between this new gene and CARD4/NOD1 suggests that, like CARD4/NOD1, the IBD1 protein is involved in the regulation of apoptosis and of the activation of NF-kappa B (Bertin et al., 1999; Inohara et al., 1999).
  • the regulation of cellular apoptosis and activation of NF-kappa B are intracellular signaling pathways which are essential in immune reactions. Specifically, these signal translation pathways are the effector pathways of the proteins of the TNF (Tumor Necrosis Factor) receptor family involved in cell-cell interactions and the cellular response to the various mediators of inflammation (cytokines).
  • TNF Tumor Necrosis Factor
  • NF-kB plays a pivotal role in inflammatory bowel diseases and is activated in lamina intestinal mononuclear cells in Crohn's disease (Schreiber et al., 1998).
  • the treatment of Crohn's disease is based on the use of sulfasalazine and glucocorticoids, which are both known to be NF-kB inhibitors (Auphan et al., 1995 and Wahl et al., 1998).
  • the IBD1 candidate gene encodes a protein very similar to NOD2, a member of the CED4/APAF1 superfamily (Ogura et al., 2000).
  • the nucleotide and protein sequences of IBD1 and NOD2 in reality only diverge for a small portion right at the start of the two reported sequences.
  • the tissue expressions of Nod2 and IBD1 can, in addition, be superimposed.
  • These two genes (proteins) can therefore be considered to be identical.
  • LRR domain of Nod2 has binding activity for bacterial lipopolysaccharides (LPS) (Inohara et al., 2000) and that deletion thereof stimulates the NFkB pathway. This result confirms the data of the invention.
  • LPS bacterial lipopolysaccharides
  • the tissue expression of IBD1 was then studied by Northern blotting.
  • a 4.5 kb transcript is visible in most human tissues.
  • the size of the transcript is in accordance with the size predicted by the cDNA.
  • the 4.5 kb transcript appears to be very poorly abundant in the small intestine and the colon. It is, on the other hand, very strongly expressed in white blood cells. This is in agreement with clinical data on transplants which suggest that Crohn's disease is potentially a disease associated with circulating immune cells. In fact, bowel transplantation does not prevent recurrence on the transplant in Crohn's disease, whereas bone marrow transplantation appears to have a beneficial effect on the progression of the disease.
  • the promoter of the IBD1 gene has not currently been identified with precision. It is, however, reasonable to think, by analogy with a very large number of genes, that this promoter lies, at least partly, immediately upstream of the gene, in the 5′ portion thereof.
  • This genetic region contains transcribed sequences, as witnessed by the presence of ESTs (HUMGS01037, AA835524, hs.105242, SHGC17274, hs.146128, hs.122983, hs.87280).
  • the ATCC clones containing these sequences were sequenced and analyzed in the laboratory, making it possible to demonstrate an exon and intron organization with possible alternative splicings. These data suggest the existence of another gene (named IBD1prox due to its proximity to IBD1).
  • the partial sequence of the complementary DNA of IBD1prox is reported (SEQ ID No. 4), as is its intron-exon structure, on SEQ ID No. 6.
  • IBD1prox corresponds to the anonymous EST HUMGS01037, the RNA of which is expressed more strongly in differentiated leukocytic lines than in undifferentiated lines.
  • this gene may have a role in inflammation and cell differentiation. It may therefore also, itself, be considered to be a good candidate for susceptibility to IBD.
  • the association between CD and the polymorphism ctg35ExC located on the coding sequence of IBD1prox supports this hypothesis even though this polymorphism does not cause any sequence variation at the protein level.
  • IBD1 and IBD1prox are not currently established. However, the considerable proximity between the two genes may reflect an interaction between them. In this case, the “head-to-tail” location of these genes suggests that they may have common or interdependent methods of regulation.
  • the coding sequence and the intron-exon junctions of the gene were sequenced from exon 2 to exon 12 inclusive, in 70 independent individuals, namely: 50 sick individuals suffering from CD, 10 sick individuals suffering from UC, 1 sick individual suffering from Blau's syndrome and 9 healthy controls.
  • the sick individuals studied were mostly familial forms of the disease and were often carriers of the susceptibility haplotype defined by the transmission disequilibrium studies.
  • the healthy controls were of Caucasian origin.
  • the mutations other than silent mutations observed in each exon are reported. They are indicated by the variation in the peptide chain. For each mutation and for each phenotype studied, the number of times where the mutation is observed, related to the number of chromosomes tested, is indicated.
  • sequence variants were observed in 43/50 CD versus 5/9 healthy controls, and 6/10 UC.
  • the existence of one or more sequence variation(s) appeared to be associated with the CD phenotype.
  • sequence variations often existed in the same individual suffering from CD, suggesting a sometimes recessive effect of the gene for CD.
  • no composite heterozygote or homozygote was observed among the patients suffering from UC or among the healthy controls.
  • non-silent variants were present both in the sick individuals suffering from UC or from CD and in the healthy individuals. They were the variants S269P, N290S, R703W and V956I located in exons 2, 4 and 9. Further information therefore appears to be necessary before selecting a possible functional role for these sequence variants.
  • V956I is a conservative sequence variation (aliphatic amino acids).
  • sequence variant S269P corresponds to a variation in amino acid class (hydroxylated to immuno acid) at the beginning of the nucleotide-binding domain.
  • This sequence variant and CD are in transmission disequilibrium. It is in fact the polymorphism Snp3 (cf. above).
  • R703W results in a modification of the amino acid class (aromatic instead of basic). This modification occurs in the intermediate region between the NBD and LRR domains, which is a region conserved between IBD1 and CARD4/NOD1. A functional role may therefore be suspected for this polymorphism.
  • This sequence variation (corresponding to the polymorphic site Snpl) is transmitted to sick individuals suffering from CD more often than at random (cf. above), confirming that this polymorphism is associated with CD. It is possible that the presence of this mutant in healthy individuals reflects incomplete penetration of the mutation as is expected for complex genetic diseases such as chronic inflammatory bowel diseases.
  • the variant R704C located immediately next to R703W, could be identified in both CD and UC. It also, itself, corresponds to a nonconservative variation of the protein (sulfur-containing amino acid instead of basic amino acid) on the same protein region, suggesting a functional effect for R704C which is as important as that for R703W.
  • the mutation G909R occurs on the last amino acid of the sixth LRR motif. It replaces an aliphatic amino acid with a basic amino acid. This variation is potentially important given the usually neutral or polar nature of the amino acids in the terminal position of the leucine-rich motifs (both for IBD1 and for NOD1/CARD4) and the conserved nature of this amino acid on the IBD1 and NOD1/CARD4 proteins.
  • the promoter of the gene is not currently determined. In all probability, however, it is likely to be located in the 5′ region upstream of the gene. According to this hypothesis, the sequence variants observed in this region may have a functional effect. This may explain the very strong association between CD and certain polymorphic loci, such as ctg35ExC or Ctg25Ex1.
  • the invention thus provides the first description of mutations in the family of genes containing a CARD domain in humans.
  • the frequency of these mutations in various inflammatory diseases shows that the IBD1 gene has an essential role in normal and pathological inflammatory processes.
  • This invention provides new paths of understanding and of research in the field of the physiopathology of normal and pathological inflammatory processes. As a result, it makes it possible to envision the development of new pharmaceutical molecules which regulate the effector pathways controlled by IBD1 and which are useful in the treatment of inflammatory diseases and in the regulation of inflammatory processes in general.
  • V928I (formerly V956I) is not significantly associated with one or the other of the inflammatory bowel diseases, and does not therefore appear to have an important role in the disease.
  • the mutation S241P (formerly S269P) is in linkage disequilibrium with the other main mutations and does not appear to play an important role, by itself, in susceptibility to inflammatory bowel diseases (data not shown).
  • the IBD1prox gene located in the promoter region of IBD1, and the partial sequence of which is disclosed in the present invention, may also, itself, have an important role in the regulation of cellular apoptosis and of the inflammatory process, as suggested by its differential expression in mature cells of the immune system.
  • the strong association reported in this work between the polymorphism marker ctg35ExC (located in the transcribed region of the gene) and Crohn's disease also pleads very strongly in favor of this hypothesis.
  • Inflammatory bowel diseases are complex genetic diseases for which, until now, no susceptibility gene had been identified with certainty.
  • the invention has made it possible to identify the first gene for susceptibility to Crohn's disease, using a positional cloning (or reverse genetics) approach. This is the first genetic location obtained using such an approach for a complex genetic disease, which demonstrates its usefulness and its feasibility, at least in certain cases in complex genetic diseases.
  • the present invention also relates to a purified or isolated nucleic acid, characterized in that it encodes a polypeptide possessing a continuous fragment of at least 200 amino acids of a protein chosen from SEQ ID No. 2 and SEQ ID No. 5.

Abstract

The invention concerns genes involved in inflammatory and/or immune diseases and some cancers, in particular intestinal cryptogenic inflammatory diseases, and proteins coded by said genes. The invention also concerns methods for diagnosing inflammatory diseases.

Description

    CROSS-REFERENCES TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 12/370,543, filed Feb. 12, 2009, which application is a continuation of U.S. application Ser. No. 10/240,046, filed Jan. 15, 2003, now U.S. Pat. No. 7,592,437 which issued on Sep. 22, 2009, which application was a National Stage application of PCT/FR01/00935, filed Mar. 27, 2001, which application claims priority to FR 0003832, filed Mar. 27, 2000, all of which are incorporated by reference in their entirety.
  • The present invention relates to genes involved in inflammatory and/or immune diseases and certain cancers, in particularly cryptogenetic inflammatory bowel diseases, and also to the proteins encoded by these genes. The present invention also relates to methods for diagnosing inflammatory diseases.
  • Cryptogenetic inflammatory bowel diseases (IBDs) are diseases characterized by an inflammation of the digestive tract, the cause of which is unknown. Depending on the location and the characteristics of the inflammation, two different nosological entities are distinguished: ulcerative colitis (UC) and Crohn's disease (CD). UC was described by S Wilkes in 1865, whereas the first case of regional ileitis was reported by Crohn in 1932. In reality, it is possible that these two diseases go back much further.
  • IBDs are chronic diseases which evolve throughout life and which affect approximately 1 to 2 individuals per 1000 inhabitants in western countries, which represents between 60000 and 100000 individuals suffering from these diseases in France. They are diseases which appear in young individuals (peak instance is in the third decade), progressing via attacks interspersed with remissions, with frequent complications such as undernutrition, retarded growth in children, bone demineralization and, in the end, malignant degeneration to colon cancer. No specific treatment exists. Conventional therapeutics make use of anti-inflammatories, of immunosuppressors and of surgery. All these therapeutic means are, themselves, a source of considerable iatrogenic morbidity. For all these reasons, IBDs appear to be a considerable public health problem.
  • The etiology of IBDs is currently unknown. Environmental factors are involved in the occurrence of the disease, as witnessed by the secular increase in incidence of the disease and the incomplete concordance in monozygous twins. The only environmental risk factors currently known are 1) tobacco, the role of which is harmful in CD and beneficial in UC, and 2) appendectomy which has a protective role for UC.
  • Genetic predisposition has been suspected for a long time due to the existence of ethnic and familial aggregation of these diseases. In fact, IBDs are more common in the Caucasian population, and in particular in the Jewish population of central Europe. Familial forms represent from 6 to 20% of IBD cases. They are particularly common when the disease begins early. However, it is studies in twins which have made it possible to confirm the genetic nature of these diseases. In fact, the concordance rate between twins for these diseases is greater in monozygous twins than in dizygous twins, which pleads strongly in favor of a hereditary component to IBDs, in particular to CD. In all probability, IBDs are complex genetic diseases involving several different genes, interacting with one another and with environmental factors. IBDs can therefore be classified within the context of multifactor diseases.
  • Two major strategies have been developed in order to demonstrate the IBD-susceptibility genes. The first is based on the analysis of genes which are candidates for physiopathological reasons. Thus, many genes have been proposed as potentially important for IBDs. They are often genes which have a role in inflammation and the immune response. Mention may be made of the HLA, TAP, TNF and MICA genes, lymphocyte T receptor, ICAM1, interleukin 1, CCR5, etc. Other genes participate in diverse functions, such as GAI2, motilin, MRAMP, HMLH1, etc. In reality, none of the various candidate genes studied has currently definitively proved itself to have a role in the occurrence of IBDs.
  • The recent development of human genome maps using highly polymorphic genetic markers has enabled geneticists to develop a nontargeted approach over the entire genome. This approach, also called reverse genetics or positional cloning, makes no hypothesis regarding the genes involved in the disease and attempts to discover them through systematic screening of the genome. The method most used for complex genetic diseases is based on studying identity by decendance of the affected individuals of the same family. This value is calculated for a large number (300-400) of polymorphism markers distributed evenly (every 10cM) over the genome). In the case of excess identity between affected individuals, the marker(s) tested indicate(s) a region supposed to contain a gene for susceptibility to the disease. In the case of complex genetic diseases, since the model underlying the genetic predisposition (number of genes and relative importance of each of them) is unknown, the statistical methods to be used will have to be adjusted.
  • The present invention relates to the demonstration of the nucleic acid sequence of genes involved in IBDs, and other inflammatory diseases, and also the use of these nucleic acid sequences.
  • In the context of the present invention, preliminary studies by the inventors have already made it possible to locate a CD-susceptibility gene. Specifically, the inventors (Hugot et al., 1996) have shown that a CD-susceptibility gene is located in the pericentromeric region of chromosome 16 (FIG. 1). It was the first gene for susceptibility to a complex genetic disease located by positional cloning and satisfying the strict criteria proposed in the literature (Lander and Kruglyak, 1995). This gene was named IBD1 (for inflammatory bowel disease 1). Since then, other locations have been proposed by other authors, in particular on chromosomes 12, 1, 3, 6 and 7 (Satsangi et al., 1996; Cho et al., 1998). Although they have been located, it has currently not been possible to identify any of these IBD-susceptibility genes.
  • Some authors have not been able to replicate this location (Rioux et al., 1998). This is not, however, surprising in the case of complex genetic diseases in which genetic heterogeneity is probable.
  • It is interesting to note that, according to the same approach of positional cloning, locations have also been proposed on chromosome 16 for several immune and inflammatory diseases, such as ankylosing spondylarthritis, Blau's syndrome, psoriasis, etc. (Becker et al., 1998; Tromp et al., 1996). All these diseases may then share the same gene (or the same group of genes) located on chromosome 16.
  • A maximum of genetic linkage tests is virtually always located at the same position, in the region of D16S409 or D16S411, separated only by 2cM. This result contradicts the considerable size (usually greater than 20cM) of the confidence interval which can be attributed to the genetic location according to an approach using nonparametric linkage analyses.
  • Comparison of the statistical tests used in the studies by the inventors shows that the tests based on complete identity by decendance (Tz2) are better than the tests based on the mean of identity by decendance (Tz) (FIG. 1). Such a difference can be explained by a recessive effect of IBD1.
  • Several genes known to be in the pericentromeric region of chromosome 16, such as the interleukin 4 receptor, CD19, CD43 or CD11, appear to be good potential candidates for CD. Preliminary results do not however plead in favor of these genes being involved in CD.
  • In particular, the present invention provides not only the sequence of IBD1 gene, but also the partial sequence of another gene, called IBD1prox due to it being located in proximity to IBD, and demonstrated as reported in the examples below. These genes, the cDNA sequence of which corresponds, respectively, to SEQ ID No. 1 and SEQ ID No. 4, are therefore potentially involved in many inflammatory and/or immune diseases and also in cancers.
  • The peptide sequence expressed by the IBD1 and IBD1prox genes is represented by SEQ ID No. 2 and SEQ ID No. 5, respectively; the genomic sequence of these genes is represented by SEQ ID No. 3 and SEQ ID No. 6, respectively.
  • Thus, a subject of the present invention is a purified or isolated nucleic acid, characterized in that it Comprises a nucleic acid sequence chosen from the following group of sequences:
    • a) SEQ ID No. 1, SEQ ID No. 3, SEQ ID No. 4 and SEQ ID No. 6;
    • b) the sequence of a fragment of at least 15 consecutive nucleotides of a sequence chosen from SEQ ID No. 1, SEQ ID No. 3, SEQ ID No. 4 or SEQ ID No. 6;
    • c) a nucleic acid sequence having a percentage identity of at least 80%, after optimal alignment, with a sequence defined in a) or b);
    • d) a nucleic acid sequence which hybridizes, under high stringency conditions, with a nucleic acid sequence defined in a) or b);
    • e) the complementary sequence or the RNA sequence corresponding to a sequence as defined in a), b), c) or d).
  • The nucleic acid sequence according to the invention defined in c) has a percentage identity of at least 80%, after optimal alignment, with a sequence as defined in a) or b) above, preferably 90%, most preferably 98%.
  • The terms “nucleic acid”, “nucleic acid sequence”, “polynucleotide”, “oligonucleotide”, “polynucleotide sequence” and “nucleotide sequence”, terms which will be employed indifferently in the present description, are intended to denote a precise series of nucleotides, which may or may not be modified, making it possible to define a fragment or a region of a nucleic acid, which may or may not comprise unnatural nucleotides, and which may correspond equally to a double-stranded DNA, a single-stranded DNA and transcription products of said DNAs. Thus, the nucleic acid sequences according to the invention also encompass PNAs (Peptide Nucleic Acids), or the like.
  • It should be understood that the present invention does not relate to the nucleotide sequences in their natural chromosomal environment, that is to say in the natural state. They are sequences which have been isolated and/or purified, that is to say they have been taken directly or indirectly, for example by copying, their environment having been at least partially modified. Thus, nucleic acids obtained by chemical synthesis are also intended to be denoted.
  • For the purpose of the present invention, the term “percentage identity” between two nucleic acid or amino acid sequences is intended to denote a percentage of nucleotides or of amino acid residues which are identical between the two sequences to be compared, obtained after the best alignment, this percentage being purely statistical and the differences between the two sequences being distributed randomly and over their entire length. The term “best alignment” or “optimal alignment” is intended to denote the alignment for which the percentage identity determined as below is highest. Sequence comparisons between two nucleic acid or amino acid sequences are conventionally carried out by comparing these sequences after having aligned them optimally, said comparison being carried out by segment or by “window of comparison” so as to identify and compare local regions of sequence similarity. The optimal alignment of the sequences for the comparison may be carried out, besides manually, by means of the local homology algorithm of Smith and Waterman (1981), by means of the local homology algorithm of Neddleman and Wunsch (1970), by means of the similarity search method of Pearson and Lipman (1988), by means of computer programs using these algorithms (GAP, BESTFIT, BLAST P, BLAST N, FASTA and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.). In order to obtain the optimal alignment, the BLAST program is preferably used, with the BLOSUM 62 matrix. The PAM or PAM250 matrices may also be used.
  • The percentage identity between two nucleic acid or amino acid sequences is determined by comparing these two sequences aligned optimally, the nucleic acid or amino acid sequence to be compared possibly comprising additions or deletions with respect to the reference sequence for optimal alignment between these two sequences. The percentage identity is calculated by determining the number of identical positions for which the nucleotide or the amino acid residue is identical between the two sequences, dividing this number of identical positions by the total number of positions compared and multiplying the resultant number by 100 so as to obtain the percentage identity between these two sequences.
  • The expression “nucleic acid sequences having a percentage identity of at least 80%, preferably 90%, more preferably 98%, after optimal alignment with a reference sequence” is intended to denote the nucleic acid sequences which, compared to the reference nucleic acid sequence, have certain modifications, such as in particular a deletion, a truncation, an extension, a chimeric fusion and/or a substitution, in particular of the point type, and the nucleic acid sequence of which exhibits at least 80%, preferably 90%, more preferably 98%, identity, after optimal alignment, with the reference nucleic acid sequence. They are preferably sequences whose complementary sequences are capable of hybridizing specifically with the sequence SEQ ID No. 1 or SEQ ID No. 4 of the invention. Preferably, the specific or high stringency hybridization conditions will be such that they ensure at least 80%, preferably 90%, more preferably 98%, identity, after optimal alignment, between one of the two sequences and the sequence complementary to the other.
  • Hybridization under high stringency conditions means that the conditions of temperature and of ionic strength are chosen such that they allow the hybridization between two complementary DNA fragments to be maintained. By way of illustration, high stringency conditions for the hybridization step for the purposes of defining the polynucleotide fragments described above are advantageously as follows.
  • The DNA-DNA or DNA-RNA hybridization is carried out in two steps: (1) prehybridization at 42° C. for 3 hours in phosphate buffer (20 mM, pH 7.5) containing 5×SSC (1×SSC corresponds to a solution of 0.15 M NaCl+0.015 M sodium citrate), 50% of formamide, 7% of sodium dodecyl sulfate (SDS), 10×Denhardt's, 5% of dextran sulfate and 1% of salmon sperm DNA; (2) hybridization per se for 20 hours at a temperature which depends on the length of the probe (i.e.: 42° C. for a probe >100 nucleotides in length), followed by 2 washes of 20 minutes at 20° C. in 2×SSC+2% SDS and 1 wash of 20 minutes at 20° C. in 0.1×SSC+0.1% SDS. The final wash is carried out in 0.1×SSC+0.1% SDS for 30 minutes at 60° C. for a probe >100 nucleotides in length. The high stringency hybridization conditions described above for a polynucleotide of defined length may be adjusted by those skilled in the art for longer or shorter oligonucleotides, according to the teaching of Sambrook et al., 1989.
  • Among the nucleic acid sequences having a percentage identity of at least 80%, preferably 90%, more preferably 98%, after optimal alignment, with the sequence according to the invention, preference is also given to the variant nucleic acid sequences of SEQ ID No. 1 or of SEQ ID No. 4, or of fragments thereof, that is to say all the nucleic acid sequences corresponding to allelic variants, that is to say individual variations of the sequence SEQ ID No. 1 or SEQ ID No. 4. These natural mutated sequences correspond to polymorphisms present in mammals, in particular in humans and, in particular, to polymorphisms which may lead to the occurrence of a pathological condition. Preferably, the present invention relates to the variant nucleic acid sequences in which the mutations lead to a modification of the amino acid sequence of the polypeptide, or of fragments thereof, encoded by the normal sequence of SEQ ID No. 1 or SEQ ID No. 4.
  • The expression “variant nucleic acid sequence” is also intended to denote any RNA or cDNA resulting from a mutation and/or variation of a splice site of the genomic nucleic acid sequence the cDNA of which has the sequence SEQ ID No. 1 or SEQ ID No. 4.
  • The invention preferably relates to a purified or isolated nucleic acid according to the present invention, characterized in that it comprises or consists of one of the sequences SEQ ID No. 1 or SEQ ID No. 4, of the sequences complementary thereto, or of the RNA sequences corresponding to SEQ ID No. 1 or SEQ ID No. 4.
  • The probes or primers, characterized in that they comprise a sequence of a nucleic acid according to the invention, are also part of the invention.
  • Thus, the present invention also relates to the primers or the probes according to the invention which may make it possible in particular to demonstrate or to distinguish the variant nucleic acid sequences, or to identify the genomic sequence of the genes the cDNA of which is represented by SEQ ID No. 1 or SEQ ID No. 4, in particular using an amplification method such as the PCR method or a related method.
  • The invention also relates to the use of a nucleic acid sequence according to the invention, as a probe or primer, for detecting, identifying, assaying or amplifying a nucleic acid sequence.
  • According to the invention, the polynucleotides which can be used as a probe or as a primer in methods for detecting, identifying, assaying or amplifying a nucleic acid sequence are a minimum of 15 bases, preferably 20 bases, or better still 25 to 30 bases in length.
  • The probes and primers according to the invention may be labeled directly or indirectly with a radioactive or nonradioactive compound using methods well known to those skilled in the art, in order to obtain a detectable and/or quantifiable signal.
  • The polynucleotide sequences according to the invention which are unlabeled can be used directly as a probe or primer.
  • The sequences are generally labeled so as to obtain sequences which can be used in many applications. The primers or the probes according to the invention are labeled with radioactive elements or with nonradio-active molecules.
  • Among the radioactive isotopes used, mention may be made of 32P, 33P, 35S, 3H or 125I. The nonradioactive entities are selected from ligands such as biotin, avidin, streptavidin or dioxygenin, haptens, dyes and luminescent agents, such as radioluminescent, chemiluminescent, bioluminescent, fluorescent or phosphorescent agents.
  • The polynucleotides according to the invention may thus be used as a primer and/or probe in methods using in particular the PCR (polymerase chain reaction) technique (Rolfs et al., 1991). This technique requires choosing pairs of oligonucleotide primers bordering the fragment which must be amplified. Reference may, for example, be made to the technique described in U.S. Pat. No. 4,683,202. The amplified fragments can be identified, for example after agarose or polyacrylamide gel electrophoresis, or after a chromatographic technique such as gel filtration or ion exchange chromatography, and then sequenced. The specificity of the amplification can be controlled using, as primers, the nucleotide sequences of polynucleotides of the invention and, as matrices, plasmids containing these sequences or else the derived amplification products. The amplified nucleotide fragments may be used as reagents in hybridization reactions in order to demonstrate the presence, in a biological sample, of a target nucleic acid of sequence complementary to that of said amplified nucleotide fragments.
  • The invention is also directed toward the nucleic acids which can be obtained by amplification using primers according to the invention.
  • Other techniques for amplifying the target nucleic acid may advantageously be employed as an alternative to PCR (PCR-like) using a pair of primers of nucleotide sequences according to the invention. The term “PCR-like” is intended to denote all the methods using direct or indirect reproductions of nucleic acid sequences, or else in which the labeling systems have been amplified; these techniques are of course, known. In general, they involve amplifying the DNA with a polymerase; when the sample of origin is an RNA a reverse transcription should be carried out beforehand. A large number of methods currently exist for this amplification, such as, for example, the SDA (strand displacement amplification) technique (Walker et al., 1992), the TAS (transcription-based amplification system) technique described by Kwoh et al. (1989), the 3SR (self-sustained sequence replication) technique described by Guatelli et al. (1990), the NASBA (nucleic acid sequence based amplification) technique described by Kievitis et al. (1991), the TMA (transcription mediated amplification) technique, the LCR (ligase chain reaction) technique described by Landegren et al. (1988), the RCR (repair chain reaction) technique described by Segev (1992), the CPR (cycling probe reaction) technique described by Duck et al. (1990), and the Q-beta-replicase amplification technique described by Miele et al. (1983). Some of these techniques have since been improved.
  • When the target polynucleotide to be detected is an mRNA, an enzyme of the reverse transcriptase type is advantageously used, prior to carrying out an amplification reaction using the primers according to the invention or to carrying out a method of detection using the probes of the invention, in order to obtain a cDNA from the mRNA contained in the biological sample. The cDNA obtained will then serve as a target for the primers or the probes used in the amplification or detection method according to the invention.
  • The probe hybridization technique may be carried out in many ways (Matthews et al., 1988). The most general method consists in immobilizing the nucleic acid extracted from the cells of various tissues or from cells in culture, on a support (such as nitrocellulose, nylon or polystyrene), and in incubating the immobilized target nucleic acid with the probe, under well-defined conditions. After hybridization, the excess probe is removed and the hybrid molecules formed are detected using the appropriate method (measuring the radioactivity, the fluorescence or the enzymatic activity linked to the probe).
  • According to another embodiment of the nucleic acid probes according to the invention, the latter may be used as capture probes. In this case, a probe, termed “capture probe”, is immobilized on a support and is used to capture, by specific hybridization, the target nucleic acid obtained from the biological sample to be tested, and the target nucleic acid is then detected using a second probe, termed “detection probe”, labeled with a readily detectable element.
  • Among the advantageous nucleic acid fragments, mention should thus be made in particular of antisense oligonucleotides, i.e. oligonucleotides, the structure of which ensures, by hybridization with the target sequence, inhibition of expression of the corresponding product. Mention should also be made of sense oligonucleotides, which, by interacting with proteins involved in regulating the expression of the corresponding product, will induce either inhibition or activation of this expression.
  • In both cases (sense and antisense), the oligo-nucleotides of the invention may be used in vitro and in vivo.
  • The present invention also relates to an isolated polypeptide, characterized in that it comprises a polypeptide chosen from:
    • a) a polypeptide of sequence SEQ ID No. 2 or SEQ ID No. 5;
    • b) a variant polypeptide of a polypeptide of sequence defined in a);
    • c) a polypeptide homologous to a polypeptide defined in a) or b), comprising at least 80% identity with said polypeptide of a);
    • d) a fragment of at least 15 consecutive amino acids of a polypeptide defined in a), b) or c);
    • e) a biologically active fragment of a polypeptide defined in a), b) or c).
  • For the purpose of the present invention, the term “polypeptide” is intended to denote proteins or peptides.
  • The expression “biologically active fragment” is intended to mean a fragment having the same biological activity as the peptide fragment from which it is deduced, preferably within the same order of magnitude (to within a factor of 10). Thus, the examples show that the IBD1 protein (SEQ ID No. 2) has a potential role in apoptosis phenomena. A biologically active fragment of the IBD1 protein therefore consists of a polypeptide derived from SEQ ID No. 2, also having a role in apoptosis. The examples below propose biological functions for the IBD1 and IBD1prox proteins, as a function of the peptide domains of these proteins, and thus allow those skilled in the art to identify the biologically active fragments.
  • Preferably, a polypeptide according to the invention is a polypeptide consisting of the sequence SEQ ID No. 2 (corresponding to the protein encoded by the IBD1 gene) or of the sequence SEQ ID No. 5 (corresponding to the protein encoded by IBD1prox) or of a sequence having at least 80% identity with SEQ ID No. 2 or SEQ ID No. 5 after optimal alignment.
  • The sequence of the polypeptide has a percentage identity of at least 80%, after optimal alignment, with the sequence SEQ ID No. 2 or SEQ ID No. 5, preferably 90%, more preferably 98%.
  • The expression “polypeptide, the amino acid sequence of which has a percentage identity of at least 80%, preferably 90%, more preferably 98%, after optimal alignment, with a reference sequence” is intended to denote the polypeptides having certain modifications compared to the reference polypeptide, such as in particular one or more deletions and/or truncations, an extension, a chimeric fusion and/or one or more substitutions.
  • Among the polypeptides, the amino acid sequence of which has a percentage identity of at least 80%, preferably 90%, more preferably 98%, after optimal alignment, with the sequence SEQ ID No. 2 or SEQ ID No. 5 or with a fragment thereof according to the invention, preference is given to the variant polypeptides encoded by the variant nucleic acid sequences as defined previously, in particular the polypeptides, the amino acid sequence of which has at least one mutation corresponding in particular to a truncation, deletion, substitution and/or addition of at least one amino acid residue compared with the sequence SEQ ID No. 2 or SEQ ID No. 5 or with a fragment thereof, more preferably the variant polypeptides having a mutation associated with the pathological condition.
  • The present invention also relates to the cloning and/or expression vectors comprising a nucleic acid or encoding a polypeptide according to the invention. Such a vector may also contain the elements required for the expression and, optionally, the secretion of the polypeptide in a host cell. Such a host cell is also a subject of the invention.
  • The vectors characterized in that they comprise a promoter and/or regulator sequence according to the invention are also part of the invention.
  • Said vectors preferably comprise a promoter, translation initiation and termination signals, and also regions suitable for regulating transcription. It must be possible for them to be maintained stably in the cell and they may optionally contain particular signals specifying secretion of the translated protein.
  • These various control signals are chosen as a function of the cellular host used. To this effect, the nucleic acid sequences according to the invention may be inserted into vectors which replicate autonomously in the chosen host, or vectors which integrate in the chosen host.
  • Among the systems which replicate autonomously, use is preferably made, depending on the host cell, of systems of the plasmid or viral type, the viral vectors possibly being in particular adenoviruses (Perricaudet et al., 1992), retroviruses, lentiviruses, poxviruses or herpesviruses (Epstein et al., 1992). Those skilled in the art are aware of the technology which can be used for each of these systems.
  • When integration of the sequence into the chromosomes of the host cell is desired, use may be made, for example, of systems of the plasmid or viral type; such viruses are, for example, retroviruses (Temin, 1986), or AAVs (Carter, 1993).
  • Among the nonviral vectors, preference is given to naked polynucleotides such as naked DNA or naked RNA according to the technology developed by the company VICAL, bacterial artificial chromosomes (BACs), yeast artificial chromosomes (YACs) for expression in yeast, mouse artificial chromosomes (MACs) for expression in murine cells and, preferably, human artificial chromosomes (HACs) for expression in human cells.
  • Such vectors are prepared according to the methods commonly used by those skilled in the art, and the clones resulting therefrom can be introduced into a suitable host using standard methods, such as, for example, lipofection, electroporation, heat shock, transformation after chemical permeabilization of the membrane, or cell fusion.
  • The invention also comprises host cells, in particular the eukaryotic and prokaryotic cells, transformed with the vectors according to the invention, and also the transgenic animals, preferably the mammals, except humans, comprising one of said transformed cells according to the invention. These animals may be used as models, for studying the etiology of inflammatory and/or immune diseases, in particular of the inflammatory diseases of the digestive tract, or for studying cancers.
  • Among the cells which can be used for the purpose of the present invention, mention may be made of bacterial cells (Olins and Lee, 1993), but also yeast cells (Buckholz, 1993) as well as animal cells, in particular mammalian cell cultures (Edwards and Aruffo, 1993), and especially Chinese hamster ovary (CHO) cells. Mention may also be made of insect cells in which it is possible to use methods employing, for example, baculo viruses (Luckow, 1993). A preferred cellular host for expressing the proteins of the invention consists of COS cells.
  • Among the mammals according to the invention, animals such as rodents, in particular mice, rats or rabbits, expressing a polypeptide according to the invention are preferred.
  • Among the mammals according to the invention, preference is also given to animals such as mice, rats or rabbits, characterized in that the gene encoding the protein of sequence SEQ ID No. 2 or SEQ ID No. 5, or the sequence of which is encoded by the homologous gene in these animals, is not functional, has been knocked out or has at least one mutation.
  • These transgenic animals are obtained, for example, by homologous recombination on embryonic stem cells, transfer of these stem cells to embryos, selection of the chimeras affected in the reproductive lines, and growth of said chimeras.
  • The transgenic animals according to the invention may thus overexpress the gene encoding the protein according to the invention, or their homologous gene, or express said gene into which a mutation is introduced. These transgenic animals, in particular mice, are obtained, for example, by transfection of a copy of this gene under the control of a promoter which is strong and ubiquitous, or selective for a tissue type, or after viral transcription.
  • Alternatively, the transgenic animals according to the invention may be made deficient for the gene encoding one of the polypeptides of sequence SEQ ID No. 2 or SEQ ID No. 5, or their homologous genes, by inactivation using the LOXP/CRE recombinase system (Rohlmann et al., 1996) or any other system for inactivating the expression of this gene.
  • The cells and mammals according to the invention can be used in a method for producing a polypeptide according to the invention, as described below, and may also be used as a model for analysis.
  • The cells or mammals transformed as described above can also be used as models in order to study the interactions between the polypeptides according to the invention, and the chemical or protein compounds involved directly or indirectly in the activities of the polypeptides according to the invention, this being in order to study the various mechanisms and interactions involved.
  • They may in particular be used for selecting products which interact with the polypeptides according to the invention, in particular the protein of sequence SEQ ID No. 2 or SEQ ID No. 5 or variants thereof according to the invention, as a cofactor or as an inhibitor, in particular a competitive inhibitor, or which have an agonist or antagonist activity with respect to the activity of the polypeptides according to the invention. Preferably, said transformed cells or transgenic animals are used as a model in particular for selecting products for combating pathological conditions associated with abnormal expression of this gene.
  • The invention also relates to the use of a cell, of a mammal or of a polypeptide according to the invention, for screening chemical or biochemical compounds which may interact directly or indirectly with the polypeptides according to the invention, and/or which are capable of modulating the expression or the activity of these polypeptides.
  • Similarly, the invention also relates to a method for screening compounds capable of interacting, in vitro or in vivo, with a nucleic acid according to the invention, using a nucleic acid, a cell or a mammal according to the invention, and detecting the formation of a complex between the candidate compounds and the nucleic acid according to the invention.
  • The compounds thus selected are also subjects of the invention.
  • The invention also relates to the use of a nucleic acid sequence according to the invention, for synthesizing recombinant polypeptides.
  • The method for producing a polypeptide of the invention in recombinant form, which is itself included in the present invention, is characterized in that the transformed cells, in particular the cells or mammals of the present invention, are cultured under conditions which allow the expression of a recombinant polypeptide encoded by a nucleic acid sequence according to the invention, and in that said recombinant polypeptide is recovered.
  • The recombinant polypeptides, characterized in that they can be obtained using said method of production, are also part of the invention.
  • The recombinant polypeptides obtained as indicated above can be in both glycosylated and nonglycosylated form, and may or may not have the natural tertiary structure.
  • The sequences of the recombinant polypeptides may also be modified in order to improve their solubility, in particular in aqueous solvents.
  • Such modifications are known to those skilled in the art, such as, for example, deletion of hydrophobic domains or substitution of hydrophobic amino acids with hydrophilic amino acids.
  • These polypeptides may be produced using the nucleic acid sequences defined above, according to the techniques for producing recombinant polypeptides known to those skilled in the art. In this case, the nucleic acid sequence used is placed under the control of signals which allow its expression in a cellular host.
  • An effective system for producing a recombinant polypeptide requires having a vector and a host cell according to the invention.
  • These cells can be obtained by introducing into host cells a nucleotide sequence inserted into a vector as defined above, and then culturing said cells under conditions which allow the replication and/or expression of the transfected nucleotide sequence.
  • The methods used for purifying a recombinant polypeptide are known to those skilled in the art. The recombinant polypeptide may be purified from cell lysates and extracts or from the culture medium supernatant, by methods used individually or in combination, such as fractionation, chromatography methods, immunoaffinity techniques using specific monoclonal or polyclonal antibodies, etc.
  • The polypeptides according to the present invention can also be obtained by chemical synthesis using one of the many known forms of peptide synthesis, for example techniques using solid phases (see in particular Stewart et al., 1984) or techniques using partial solid phases, by fragment condensation or by conventional synthesis in solution.
  • The polypeptides obtained by chemical synthesis and which may comprise corresponding unnatural amino acids are also included in the invention.
  • The mono- or polyclonal antibodies, or fragments thereof, chimeric antibodies or immunoconjugates, characterized in that they are capable of specifically recognizing a polypeptide according to the invention, are part of the invention.
  • Specific polyclonal antibodies may be obtained from a serum of an animal immunized against the polypeptides according to the invention, in particular produced by genetic recombination or by peptide synthesis, according to the usual procedures.
  • The advantage of antibodies which specifically recognize certain polypeptides, variants or immunogenic fragments thereof according to the invention is in particular noted.
  • The mono- or polyclonal antibodies, or fragments thereof, chimeric antibodies or immunoconjugates characterized in that they are capable of specifically recognizing the polypeptides of sequence SEQ ID No. 2 or SEQ ID No. 5 are particularly preferred.
  • The specific monoclonal antibodies may be obtained according to the conventional method of hybridoma culture described by Köhler and Milstein (1975).
  • The antibodies according to the invention are, for example, chimeric antibodies, humanized antibodies, or Fab or F(ab′)2 fragments. They may also be in the form of immunoconjugates or of labeled antibodies, in order to obtain a detectable and/or quantifiable signal.
  • The invention also relates to methods for detecting and/or purifying a polypeptide according to the invention, characterized in that they use an antibody according to the invention.
  • The invention also comprises purified polypeptides, characterized in that they are obtained using a method according to the invention.
  • Moreover, besides their use for purifying the polypeptides, the antibodies of the invention, in particular the monoclonal antibodies, may also be used for detecting these polypeptides in a biological sample.
  • They thus constitute a means for the immunocytochemical or immunohistochemical analysis of the expression of the polypeptides according to the invention, in particular the polypeptides of sequence SEQ ID No. 2 or SEQ ID No. 5, or a variant thereof, on specific tissue sections, for example using immunofluorescence, gold labeling and/or enzymatic immunoconjugates.
  • They may in particular make it possible to demonstrate abnormal expression of these polypeptides in the biological specimens or tissues.
  • More generally, the antibodies of the invention may advantageously be used in any situation where the expression of a polypeptide according to the invention, normal or mutated, must be observed.
  • Thus, a method for detecting a polypeptide according to the invention, in a biological sample, comprising the Steps of bringing the biological sample into contact with an antibody according to the invention and demonstrating the antigen-antibody complex formed, is also a subject of the invention, as is a kit for carrying out such a method. Such a kit in particular contains:
      • a) a monoclonal or polyclonal antibody according to the invention;
      • b) optionally, reagents for constituting a medium suitable for the immunoreaction;
      • c) the reagents for detecting the antigen-antibody complex produced during the immunoreaction.
  • The antibodies according to the invention may also be used in the treatment of an inflammatory and/or immune disease, or of a cancer, in humans, when abnormal expression of the IBD1 gene or of the IBD1prox gene is observed. Abnormal expression means overexpression or the expression of a mutated protein.
  • These antibodies may be obtained directly from human serum, or may be obtained from animals immunized with polypeptides according to the invention, and then “humanized”, and may be used as such or in the preparation of a medicinal product intended for the treatment of the abovementioned diseases.
  • The methods for determining an allelic variability, a mutation, a deletion, a loss of heterozygocity or any genetic abnormability of the gene according to the invention, characterized in that they use a nucleic acid sequence, a polypeptide or an antibody according to the invention, are also part of the invention.
  • The invention in fact provides the sequence of the IBD1 and IBD1prox genes involved in inflammatory and/or immune diseases, and in particular IBDs. One of the teachings of the invention is to specify the mutations, in these nucleic acid or polypeptide sequences, which are associated with a phenotype corresponding to one of these inflammatory and/or immune diseases.
  • These mutations can be detected directly by analysis of the nucleic acid and of the sequences according to the invention (genomic DNA, RNA or cDNA), but also via the polypeptides according to the invention. In particular, the use of an antibody according to the invention which recognizes an epitope bearing a mutation makes it possible to distinguish between a “healthy” protein and a protein “associated with a pathological condition”.
  • Thus, the study of the IBD1 gene in various inflammatory and/or immune human diseases thus shows that sequence variants of this gene exist in Crohn's disease, ulcerative colitis and Blau's syndrome, as demonstrated by the examples. These sequence variations result in considerable variations in the deduced protein sequence. In fact, they are either located on very conserved sites of the protein in important functional domains, or they result in the synthesis of a truncated protein. It is therefore extremely probable that these deleterious modifications lead to a modification of the function of the protein and therefore have a causal effect in the occurrence of these diseases.
  • The variety of diseases in which these mutations are observed suggests that the IBD1 gene is potentially important in many inflammatory and/or immune diseases. This result should be compared with the fact that the pericentromeric region of chromosome 16 has been described as containing genes for susceptibility to various human diseases, such as ankylosing spondylarthritis or psoriatic arthropathy. It may therefore be considered that IBD1 has an important role in a large number of inflammatory and/or immune diseases.
  • In particular, IBD1 can be associated with granulomatous inflammatory diseases. Blau's syndrome and CD are in fact diseases which are part of this family. It is therefore hoped that variations in the IBD1 gene will be found for the other diseases of the same family (sarcoidosis, Behcet's disease, etc.).
  • In addition, the involvement of IBD1 in the cellular pathways leading to apoptosis raises the question of its possible carcinogenic role. In fact, it is expected that a dysregulation of IBD1 may result in a predisposition to cancer. This hypothesis is supported by the fact that a predisposition to colon cancer exists in inflammatory bowel diseases. IBD1 may in part explain this susceptibility to cancer and define new carcinogenic pathways.
  • The precise description of the mutations which can be observed in the IBD1 gene thus makes it possible to lay down the foundations of a molecular diagnosis for the inflammatory or immune diseases in which this role is demonstrated. Such an approach, based on searching for mutations in the gene, will make it possible to contribute to the diagnosis of these diseases and possibly to reduce the extent of certain additional examinations which are invasive or expensive. The invention lays down the foundations of such a molecular diagnosis based on searching for mutations in IBD1.
  • The molecular diagnosis of inflammatory diseases should also make it possible to improve the nosological classification of these diseases and to more clearly define subgroups of particular diseases by their clinical characteristics, the progressive nature of the disease or the response to certain treatments. By way of example, the dismantling of the existing mutations may thus make it possible to classify the currently undetermined forms of colitis which represent more than 10% of inflammatory bowel diseases. Such an approach will make it possible to propose an early treatment suitable for each patient. In general, such an approach makes it possible to hope that it will eventually be possible to define an individualized treatment for the disease, depending on the genetic area of each disease, including curative and preventive measures.
  • In particular, preference is given to a method of diagnosis and/or of prognostic assessment of an inflammatory disease or of a cancer, characterized in that the presence of at least one mutation and/or a deleterious modification of expression of the gene corresponding to SEQ ID No. 1 or SEQ ID No. 4 is determined, using a biological specimen from a patient, by analyzing all or part of a nucleic acid sequence corresponding to said gene. The genes SEQ ID No. 3 or SEQ ID No. 6 may also be studied.
  • This method of diagnosis and/or of prognostic assessment may be used preventively (a study of predisposition to inflammatory diseases or to cancer), or in order to serve in establishing and/or confirming a clinical condition in a patient.
  • Preferably, the inflammatory disease is an inflammatory disease of the digestive tract, and the cancer is a cancer of the digestive tract (small intestine or colon).
  • The teaching of the invention in fact makes it possible to determine the mutations which exhibit a linkage disequilibrium with inflammatory diseases of the digestive tract, and which are therefore associated with such diseases.
  • The analysis may be carried out by sequencing all or part of the gene, or by other methods known to those skilled in the art. Methods based on PCR, for example PCR-SSCP, which makes it possible to detect point mutations, may in particular be used.
  • The analysis may also be carried out by attaching a probe according to the invention, corresponding to one of the sequences SEQ ID No. 1, 3, 4 or 6, to a DNA chip, and hybridization on these microplates. A DNA chip containing a sequence according to the invention is also one of the subjects of the invention.
  • Similarly, a protein chip containing an amino acid sequence according to the invention is also a subject of the invention. Such a protein chip makes it possible to study the interactions between the polypeptides according to the invention and other proteins or chemical compounds, and may thus be useful for screening compounds which interact with the polypeptides according to the invention. The protein chips according to the invention may also be used to detect the presence of antibodies directed against the polypeptides according to the invention in the serum of patients. A protein chip containing an antibody according to the invention may also be used.
  • Those skilled in the art are also able to carry out techniques for studying the deleterious modification of the expression of a gene, for example by studying the mRNA (in particular by Northern blotting or with RT-PCR experiments, with probes or primers according to the invention), or the protein expressed, in particular by Western blotting, using antibodies according to the invention.
  • The gene tested is preferably the gene of sequence SEQ ID No. 1, the inflammatory disease for which the intention is to predict susceptibility being a disease of the digestive tract, in particular Crohn's disease or ulcerative colitis. If the intention is to detect a cancer, it is preferably colon cancer.
  • The invention also relates to methods for obtaining an allele of the IBD1 gene, associated with a detectable phenotype, comprising the following steps:
      • a) obtaining a nucleic acid sample from an individual expressing said detectable phenotype;
      • b) bringing said nucleic acid sample into contact with an agent capable of specifically detecting a nucleic acid encoding the IBD1 protein;
      • c) isolating said nucleic acid encoding the IBD1 protein.
  • Such a method may be followed by a step of sequencing all or part of the nucleic acid encoding the IBD1 protein, which makes it possible to predict susceptibility to inflammatory disease or of a cancer.
  • The agent capable of specifically detecting a nucleic acid encoding the IBD1 protein is advantageously an oligonucleotide probe according to the invention, which may be made up of DNA, RNA or PNA, which may or may not be modified. The modifications may include radioactive or fluorescent labeling, or may be due to modifications in the bonds between the bases (phosphorothioates or methyl phosphonates, for example). Those skilled in the art are aware of the protocols for isolating a specific DNA sequence. Step b) of the method described above may also be an amplification step as described above.
  • The invention also relates to a method for detecting and/or assaying a nucleic acid according to the invention, in a biological sample, comprising the following steps of bringing a probe according to the invention into contact with a biological sample, and detecting and/or assaying the hybrid formed between said polynucleotide and the nucleic acid of the biological sample.
  • Those skilled in the art are capable of carrying out such a method, and may in particular use a kit of reagents, comprising:
      • a) a polynucleotide according to the invention, used as a probe;
      • b) the reagents required for carrying out a hybridization reaction between said probe and the nucleic acid of the biological sample;
      • c) the reagents required for detecting and/or assaying the hybrid formed between said probe and the nucleic acid of the biological sample;
        which is also a subject of the invention.
  • Such a kit may also contain positive or negative controls in order to ensure the quality of the results obtained.
  • However, in order to detect and/or assay a nucleic acid according to the invention, those skilled in the art may also perform an amplification step using primers chosen from the sequences according to the invention.
  • Finally, the invention also relates to the compounds chosen from a nucleic acid, a polypeptide, a vector, a cell or an antibody according to the invention, or the compounds obtained using the screening methods according to the invention, as a medicinal product, in particular for preventing and/or treating an inflammatory and/or immune disease, or a cancer, associated with the presence of at least one mutation of the gene corresponding to SEQ ID No. 1 or SEQ ID No. 4, preferably an inflammatory disease of the digestive tract, in particular Crohn's disease or ulcerative colitis.
  • The following examples make it possible to understand more clearly the advantages of the invention, and should not be considered to limit the scope of the invention.
  • DESCRIPTION OF THE FIGURES
  • FIG. 1: Nonparametric genetic linkage tests for Crohn's disease in the pericentromeric region of chromosome 16 (according to Hugot et al., 1996). Multipoint linkage analysis based on identity by decendance for the markers of the pericentromeric region of chromosome 16. The genetic distances between markers were estimated using the CRIMAP program. The lod score (MAPMAKER/SIBS) is indicated on the left-hand figure. Two pseudoprobability tests were developed and reported on the right-hand figure. The first (Tz) is analogous to the test of the means. The second (Tz2) is analogous to the test of the proportion of affected pairs sharing two alleles.
  • FIG. 2: Multipoint nonparametric genetic linkage analysis. 78 families with several relatives suffering from Crohn's disease were genotyped for 26 polymorphism markers in the pericentromeric region of chromosome 16. The location of each marker is symbolized by an arrow. The order of the markers and the distance separating them derive from the analysis of the experimental data with the Crimap software. The arrows under the curve indicate the markers SPN, D16S409 and D16S411 used in the first study published (Hugot et al., 1996). The arrows located at the top of the figure correspond to the markers D16S3136, D16S541, D16S3117, D16S416 and D16S770 located at the maximum of the genetic linkage test. The typing data were analyzed using the multipoint nonparametric analysis program of the Genehunter software version 1.3. The maximum NPL score is 3.33 (p=0.0004).
  • FIG. 3: Diagrammatic representation of the protein encoded by IBD1. The protein encoded by IBD1 is represented horizontally. The various domains of which it is composed are indicated on the figure with the amino acid reference number corresponding to the start and to the end of each domain. The protein consists of a CARD domain, a nucleotide-binding domain (NBD) and leucine-rich motifs (LRR).
  • FIG. 4: Diagrammatic representation of the IBD1/NOD2 protein in three variants associated with CD.
  • A: The translation produced deduced from the cDNA sequence of the IBD1 candidate gene is identical to that of NOD2 (Ogura et al., 2000). The polypeptide contains 2 CARD domains (CAspase Recruitment Domains), a nucleotide-binding domain (NBD) and 10 repeats of 27 amino acids, leucine-rich motifs (LRR). The consensus sequence of the ATP/GTP-binding site of the motif A (P loop) of the NBD is indicated with a black circle. The sequence changes encoded by the three main variants associated with CD are SNP 8 (R675W), SNP 12 (G881R) and SNP 13 (frame shift 980). The frame shift changes a leucine codon to a proline codon at position 980, which is immediately followed by a stop codon.
  • B: Rare missense variants of NOD2 in 457 CD patients, 159 UC patients and 103 unaffected, unrelated individuals. The positions of the rare missense variants are indicated for the three groups. The scale on the left indicates the number of each variant identified in the groups under investigation and that on the right measures the frequency of the mutation. The allelic frequencies of the polymorphism V928I was not significantly different (0.92:0.08) in the three groups and the corresponding genotypes were in Hardy-Weinberg equilibrium.
  • EXAMPLES Example 1 Fine Location of IBD1
  • The first step toward identifying the IBD1 gene was to reduce the size of the genetic region of interest, initially centered on the marker D16S411 located between D16S409 and D16S419 (Hugot et al., 1996 and FIG. 1). A group of close markers (high resolution genetic map) was used in order to more clearly specify the genetic region, and made it possible to complete the genetic linkage analyses and to search for a genetic linkage disequilibrium with the disease.
  • The study related to 78 families comprising at least 2 relatives suffering from CD, which corresponded to 119 affected pairs. The families comprising sick individuals suffering from UC were excluded from the study.
  • Twenty-six genetic polymorphism markers of the micro-satellite type were studied. These markers together made up a high resolution map with an average distance between markers of the order of 1cM in the genetic region of interest. The characteristics of the markers studied are given in table 1.
  • TABLE 1
    Polymorphic markers of the microsatellite
    type used for the fine location of IBD1
    Name of polymorphism Cumulative
    marker distance (cM) PCR primers
    D16S3120 0 SEQ ID No. 7
    (AFM326vc5) SEQ ID No. 8
    D16S298 2.9 SEQ ID No. 9
    (AFMa189wg5) SEQ ID No. 10
    D16S299 3.4 SEQ ID No. 11
    SEQ ID No. 12
    SPN 3.9 SEQ ID No. 13
    SEQ ID No. 14
    D16S383 4.3 SEQ ID No. 15
    SEQ ID No. 16
    D16S753 4.9 SEQ ID No. 17
    (GGAA3G05) SEQ ID No. 18
    D16S3044 5.8 SEQ ID No. 19
    (AFMa222za9) SEQ ID No. 20
    D16S409 5.8 SEQ ID No. 21
    (AFM161xa1) SEQ ID No. 22
    D16S3105 6.1 SEQ ID No. 23
    (AFMb341zc5) SEQ ID No. 24
    D16S261 6.8 SEQ ID No. 25
    (MFD24) SEQ ID No. 26
    D16S540 6.9 SEQ ID No. 27
    (GATA7B02) SEQ ID No. 28
    D16S3080 7 SEQ ID No. 29
    (AFMb068zb9) SEQ ID No. 30
    D16S517 7 SEQ ID No. 31
    (AFMa132we9) SEQ ID No. 32
    D16S411 8 SEQ ID No. 33
    (AFM186xa3) SEQ ID No. 34
    D16S3035 10.4 SEQ ID No. 35
    (AFMa189wg5) SEQ ID No. 36
    D16S3136 10.4 SEQ ID No. 37
    (AFMa061xe5) SEQ ID No. 38
    D16S541 11.4 SEQ ID No. 39
    (GATA7E02) SEQ ID No. 40
    D16S3117 11.5 SEQ ID No. 41
    (AFM288wb1) SEQ ID No. 42
    D16S416 12.4 SEQ ID No. 43
    (AFM210yg3) SEQ ID No. 44
    D16S770 13.2 SEQ ID No. 45
    (GGAA20G02) SEQ ID No. 46
    D16S2623 15 SEQ ID No. 47
    (GATA81B12) SEQ ID No. 48
    D16S390 16.5 SEQ ID No. 49
    SEQ ID No. 50
    D16S419 20.4 SEQ ID No. 51
    (AFM225zf2) SEQ ID No. 52
    D16S771 21.8 SEQ ID No. 53
    (GGAA23C09) SEQ ID No. 54
    D16S408 25.6 SEQ ID No. 55
    (AFM137xf8) SEQ ID No. 56
    D16S508 38.4 SEQ ID No. 57
    (AFM304xf1) SEQ ID No. 58
  • Each marker is listed according to international nomenclature and mostly by the name proposed by the laboratory of origin. The markers appear according to their order on the chromosome (from 16p to 16q). The genetic distance between the markers (in Kosambi centiMorgans, calculated from the experimental data using the Crimap program) is indicated in the second column. The first polymorphic marker is taken randomly as a reference point. The oligonucleotides which were used for the polymerase chain reaction (PCR) are indicated in the third column.
  • The genotyping of these microsatellite markers was based on automatic sequencer technology using fluorescent primers. Briefly, after amplification, the fluorescent polymerase chain reaction (PCR) products were loaded onto a polyacrylamide gel on an automatic sequencer according to the manufacturer's recommendations (Perkin Elmer). The size of the alleles for each individual was deduced using the Genescan® and Genotyper® software. The data were then kept on an integrated computer base containing the genealogical, phenotypic and genetic data. They were then used for the genetic linkage analyses.
  • Several quality controls were carried out throughout the genotyping procedure:
      • independent double reading of the genotyping data,
      • use of a standard DNA as an internal control for each electrophoretic migration,
      • control of the size range for each allele observed,
      • search for mendelian transmission errors,
      • calculation of the genetic distance between markers (CRIMAP program) and comparison of this distance with the data from the literature,
      • further typing of the markers for which recombination between close markers was observed.
  • The genotyping data were analyzed by multipoint non-parametric genetic linkage methods (GENEHUNTER program version 1.3). The informativeness of the marker system was greater than 80% for the region studied. The test maximum (NPL=3.33; P=0.0004) was obtained for the markers D16S541, D16S3117, D16S770 and D16S416 (FIG. 2).
  • The typing data for these 26 polymorphism markers were also analyzed so as to search for a transmission disequilibrium. Two groups of 108 and 76 families with one or more sick individuals suffering from CD were studied. The statistical test for transmission disequilibrium has been described by Spielman et al. (1993). In this study, only one sick individual per family was taken into account, and the value of p was corrected by the number of alleles tested for each marker studied.
  • A transmission disequilibrium was observed for alleles 4 and 5 (size 205 and 207 base pairs, respectively) of the marker D16S3136 (p=0.05 and p=0.01, respectively).
  • These results, which suggest an association between the marker D16S3136 and CD, led to the construction of a physical map of the genetic region centered on D16S3136 and to establishment of the sequence of a large genomic DNA segment (BAC) containing this polymorphic site. It was then possible to identify and analyze a larger number of polymorphism markers in the region of D16S3136, and also to define and study the transcribed sequences present in the region.
  • Example 2 Physical Mapping of the IBD1 Region
  • A contig of genomic DNA fragments, centered on the markers D16S3136, D16S3117, D16S770 and D16S416, was generated from the human genomic DNA libraries of the Jean Dausset foundation/CEPH. The chromosomal DNA segments were identified based on certain polymorphism markers used in fine genetic mapping (D16S411, D16S416, D16S541, D16S770, D16S2623, D16S3035, D16S3117 and D16S3136). For each marker, a bacterial artificial chromosome (BAC) library was screened by PCR so as to search for clones containing the marker sequence. Depending on whether or not the sequences tested were present on the BAC clones, it was then possible to organize the clones among one another using the Segmap software version 3.35.
  • It was possible to establish, for the BACs, a continuous organization (contig) covering the genetic region of interest, according to a method known to those skilled in the art (Rouquier et al., 1994; Kim et al., 1996; Asakawa et at., 1997). To do this, the ends of the BACs identified were sequenced and these new sequence data were then used to repeatedly screen the BAC libraries. At each screening, the BAC contig then progressed by a step until a continuum of overlapping clones was obtained. The size of each BAC contributing to the contig was deduced from its migration profile on a pulsed field agarose gel.
  • A BAC contig containing 101 BACs and extending over an overall distance of more than 2.5 Mb, with an average redundancy of 5.5 BACs at each point of the contig, was thus constructed. The average size of the BACs is 136 kb.
  • Example 3 Sequencing of BAC hb87b10
  • The BAC of this contig containing the polymorphism marker D16S3136 (called hb87b10), the size of which was 163761 bp, was sequenced according to the “shotgun” method. Briefly, the BAC DNA was fragmented by sonication. The DNA fragments thus generated were subjected to agarose gel electrophoresis and those with a size greater than 1.5 kb were eluted in order to be analyzed. These fragments were then cloned into the m13 phage, which was itself introduced into bacteria made competent, by electroporation. After culturing, the DNA of the clones was recovered and sequenced by automatic sequencing methods using fluorescent primers of the m13 vector on an automatic sequencer.
  • 1526 different sequences with an average size of 600 bp were generated, which were organized with respect to one another using the Polyphredphrap® software, resulting in a sequence contig covering the entire BAC. The sequence thus generated had an average redundancy of 5.5 genomic equivalents. The rare (n=5) sequence gaps not represented in the m13 clone library were filled by generating specific PCR primers, on either side of these gaps, and analyzing the PCR product derived from the genomic DNA of a healthy individual.
  • Sequence homologies with sequences available in public genetic databases (Genbank) were sought. No known gene could be identified in this region of 163 kb. Several ESTs were positioned, suggesting that unknown genes were contained in this sequence. These ESTs derived from the public genetic databases (Genbank, GDB, Unigene, dbEST) bore the following references: AI167910, A1011720, Rn24957, Mm30219, hs132289, AA236306, hs87296, AA055131, hs151708, AA417809, AA417810, hs61309, hs116424, HUMGS01037, AA835524, hs105242, SHGC17274, hs146128, hs122983, hs87280 and hs135201. The search for putative exons using the GRAIL computer program made it possible to identify several potential exons, polyadenylation sites and promoter sequences.
  • Example 4 Transmission Disequilibrium Studies
  • 12 biallelic polymorphism markers (SNPs) were identified in a region extending over approximately 250 kb and centered on the BAC hb87b10. These polymorphisms were generated by analyzing the sequence of ten or so independent sick individuals suffering from CD. The sequencing was mostly carried out at known ESTs positioned on the BAC or in the region thereof. Putative exons, predicted by the GRAIL computer program, were also analyzed. The characteristics of the polymorphic markers thus identified are given in table 2.
  • TABLE 2
    Characteristics of biallelic polymorphism
    markers studied in the region of IBD1
    I II III IV V VI
    1 KIAA0849ex9 AS-PCR SEQ ID No. 88 to 90 116
    2 hb27G11F PCR- BsrI SEQ ID No. 86, 87 185
    RFLP 116
    69
    3 Ctg22Ex1 PCR- RsaI SEQ ID No. 84, 85 381
    RFLP 313
    69
    4 SNP1 AS-PCR SEQ ID No. 81 to 83 410
    5 ctg2931- LO SEQ ID No. 78 to 80 51
    3ac/ola 49
    6 ctg2931- LO SEQ ID No. 75 to 77 44
    5ag/ola 42
    7 SNP3-2931 AS-PCR SEQ ID No. 72 to 74 245
    8 Ctg25Ex1 PCR- BsteII SEQ ID No. 70, 71 207
    RFLP 122
    85
    9 CTG35ExA AS-PCR SEQ ID No. 67 to 69 333
    10 ctg35ExC AS-PCR SEQ ID No. 64 to 66 198
    11 D16S3136 SEQ ID No. 37, 38
    12 hb133D1f PCR- TaqI SEQ ID No. 62, 63 369
    RFLP 295
    74
    13 D16S3035 SEQ ID No. 35, 36
    14 ADCY7int7 AS-PCR SEQ ID No. 59 to 61 140
    AS-PCR: allele-specific PCR; LO: ligation of oligonucleotides
  • The 12 biallelic polymorphism markers newly described in this study are listed in this table. For each one of them, the following are indicated:
      • the locus (column I)
      • the name (column II)
      • the genotyping technique used (column III)
      • the restriction enzyme possibly used (column IV)
      • the oligonucleotide primers used for the polymerase chain reaction or for the ligation (column V)
      • the size of the products expected during typing
        (column VI)
  • 199 families comprising 1 or more sick individuals suffering from CD were typed for these 12 polymorphism markers and also for the markers D16S3035 and D16S3136 located on the BAC hb87b10. The families comprising sick individuals suffering from UC were not taken into account. The methods for typing the polymorphisms studied were variable depending on the type of polymorphism, using:
      • the PCR-RFLP technique (amplification followed by enzymatic digestion of the PCR product) when the polymorphism was located on an enzymatic restriction site.
      • PCR with primers specific for the polymorphic site: differential amplification of two alleles using primers specific for each allele.
      • Oligoligation test: differential ligation using oligonucleotides specific for each allele, followed by polyacrylamide gel electrophoresis.
  • The typing data were then analyzed using a transmission disequilibrium test (TDT computer program of the GENEHUNTER software version 2). For the families comprising several affected relatives, a single sufferer was taken into account for the analysis. In fact, if several related sufferers are taken into account, this poses the problem of nonindependence of the data in the statistical calculations and can induce an inflation of the value of the test. The sufferer used for the analysis was drawn by lots, within each family, using an automatic randomization procedure. Given this randomization, the value of the statistical test obtained represented only one possible sample derived from the group of families studied. So as not to limit the analysis to this one possible sample, and in order to understand more clearly the soundness of the results obtained, for each test, about one hundred random samples were thus generated and analyzed.
  • The markers were studied separately and then grouped according to their order on the chromosomal segment (KIAA0849ex9 (locus 1), hb27G11F (locus 2), Ctg22Ex1 (locus 3), SNP1 (locus 4), ctg2931-3ac/ola (locus 5), ctg2931-5ag/ola (locus 6), SNP3-2931 (locus 7), Ctg25Ex1 (locus 8), CTG35ExA (locus 9), ctg35ExC (locus 10), d16s3136 (locus 11), hb133D1f (locus 12), D16S3035 (locus 13), ADCY7int7 (locus 14)) (table 2). The haplotypes comprising 2, 3 and 4 consecutive markers were thus analyzed still using the same strategy (100 random samples, taking a single affected individual for each family).
  • For each sample tested, only the genotypes (or haplotypes) carried by at least 10 parental chromosomes were taken into account. On average, 250 different tests were thus carried out for each sample. It was then possible to deduce the number of tests expected to be positive for each significance threshold and to compare this distribution to the distribution observed. For the healthy individuals, the distribution of the tests is not different from that expected on a random basis (χ2=2.85, ddl=4, p=0.58). For the sick individuals, on the other hand, there is an excess of positive tests, reflecting the existence of a transmission disequilibrium in the region studied.
  • The results of the transmission disequilibrium test for each polymorphism marker taken separately or for the haplotypes showing the strongest transmission disequilibriums showed that the following markers and the disease are in linkage disequilibrium: Ctg22Ex1 (locus 3), SNP1 (locus 4), ctg2931-5ag/ola (locus 6), SNP3-2931 (locus 7), Ctg25Ex1 (locus 8) and ctg35ExC (locus 10). These markers extend over a region of approximately 50 kb (positions 74736 to 124285 on the sequence of hb87b10).
  • The haplotypes the most strongly associated with Crohn's disease themselves also extend over this region. Thus, for the majority of the random samples, the transmission test was positive (p<0.01) for haplotypes combining the following markers:
      • locus 5-6, locus 6-7, locus 7-8, locus 8-9, locus 9-10, locus 10-11
      • locus 5-6-7, locus 6-7-8, locus 7-8-9, locus 8-9-10, locus 9-10-11
      • locus 5-6-7-8, locus 6-7-8-9, locus 7-8-9-10.
  • The susceptibility haplotype most at risk is defined by the loci 7 to 10. This is the haplotype 1-2-1-2 (table 2).
  • The markers tested are, as expected, in linkage disequilibrium with respect to one another.
  • More recently, a new test, the Pedigree Disequilibrium Test (PDT), published in July 2000 (Martin et al., 2000), was used to understand more clearly the meaning of the results obtained with the TDT computer program. This new statistic in fact makes it possible to use all of the information available in a family, both from the sick individuals and from the healthy individuals, and to counterbalance the importance of each relative in an overall statistic for each family. The values of p corresponding to the PDT tests and obtained for an enlarged group of 235 families with one or more relatives suffering from Crohn's disease are given in table 3. This new analysis confirms that the region of the BAC hb87b10 is indeed associated with Crohn's disease.
  • TABLE 3
    Results of the PDT tests carried out on 235
    families suffering from Crohn's disease
    LOCUS VALUE p OF THE PDT TEST
    KIAA0849ex9 NS
    hb27g11f 0.05
    ctg22ex1 0.01
    SNP1 0.001
    ctg2931-3ac/ola NS
    ctg2931-5ag/ola 0.0001
    SNP3-2931 0.0001
    ctg25ex1 0.0006
    ctg35exA NS
    ctg35exC 0.00002
    D16S3136 NS
    hb133d1f NS
    D16S3035 NS
    (NS: not significant)
  • Example 5 Identification of the IBD1 Gene
  • The published EST groups (Unigene references: Hs135201, Hs87280, Hs122983, Hs146128, H5105242, Hs116424, Hs61309, Hs151708, Hs87296 and Hs132289) present on the BAC hb87b10 were studied in the search for a more complete complementary DNA (cDNA) sequence. For IBD1prox, the clones available in public libraries were sequenced and the sequences were organized with respect to one another. For IBD1, a peripheral blood complementary DNA library (Stratagene human blood cDNA lambda zapexpress ref 938202) was screened with the PCR products generated from known ESTs according to the methods proposed by the manufacturer. The sequence of the cDNAs thus identified was then used for further screening of the cDNA library, and so on, until the presented cDNA was obtained.
  • The EST hs135201 (UniGene) made it possible to identify a cDNA not appearing on the available genetic databases (Genbank). It therefore corresponds to a new human gene. Comparison of the sequence of the cDNA and of the genomic DNA showed that this gene consists of 11 exons and 10 introns. An additional exon, positioned 5′ to the cDNA identified, is predicted by analysis of the sequence with the Grail program. These exons are very homologous to the first exons of the CARD4/NOD1 gene. Taking into consideration all of the exons identified and the putative additional exon, this new gene appears to have a genomic structure very close to that of CARD4/NOD1. Moreover, a transcription initiation site appears upstream of the first putative exon. For all of these reasons, the putative exon was considered to contribute to this new gene. The cDNA reproduced in the annex (SEQ ID No. 1) therefore comprises all of the identified sequence plus the sequence predicted by the computer modeling, the complementary DNA beginning randomly at the first ATG codon of the predicted coding sequence. On this basis, the gene would therefore comprise 12 exons and 11 introns. The intron-exon structure of the gene is reported on SEQ ID No. 3.
  • The protein sequence deduced from the nucleotide sequence comprises 1041 amino acids (SEQ ID No. 2). This sequence has not been found on the biological databases either (Genpept, pir, swissprot).
  • Now, more recently, it has not been possible to confirm the putative exon described above. The IBD1 gene therefore effectively comprises only 11 exons and 10 introns and encodes a protein of 1013 amino acids (i.e. 28 amino acids less than initially determined).
  • The study of the deduced protein sequence shows that this gene contains three different functional domains (FIG. 3):
      • A CARD domain (Caspase Recruitment Domain) known to be involved in the interaction between proteins regulating apoptosis and activation of the NFkappa B pathway. The CARD domain makes it possible to classify this new protein in the CARD protein family, the most longstanding members of which are CED4, APAF1 and RICK.
      • An NBD domain (Nucleotide-Binding Domain) comprising an ATP-recognition site and a magnesium-binding site. The protein should therefore very probably have kinase activity.
      • An LRR domain (Leucine-Rich Domain) presumed to participate in the interaction between proteins, by analogy with other described protein domains.
  • Moreover, the LRR domain of the protein makes it possible to affiliate the protein to a family of proteins involved in intracellular signaling and present both in plants and in animals.
  • Comparison of this new gene with previously identified genes available in the public databases shows that this gene is very homologous to CARD4/NOD1 (Bertin et al., 1999; Inohara et al., 1999). This homology relates to the sequence of the complementary DNA, the intron-exon structure of the gene and the protein sequence. The sequence identity of the two complementary DNAs is 58%. A similarity is also observed at the level of the intron-exon structure. The sequence homology at the protein level is of the order of 40%.
  • The similarity between this new gene and CARD4/NOD1 suggests that, like CARD4/NOD1, the IBD1 protein is involved in the regulation of apoptosis and of the activation of NF-kappa B (Bertin et al., 1999; Inohara et al., 1999). The regulation of cellular apoptosis and activation of NF-kappa B are intracellular signaling pathways which are essential in immune reactions. Specifically, these signal translation pathways are the effector pathways of the proteins of the TNF (Tumor Necrosis Factor) receptor family involved in cell-cell interactions and the cellular response to the various mediators of inflammation (cytokines). The new gene therefore appears to be potentially important in the inflammatory reaction in general.
  • Several bodies of proof support bacteria-induced deregulation of NF-kB in Crohn's disease. First of all, spontaneous susceptibility to IBD in mice has been associated with mutations in Tlr4, a molecule known to bind to LPS via its LRR domain (Poltorak et al., 1998 and Sundberg et al., 1994) and to be a member of the activators of the NF-kB family. Second, treatment with antibiotics causes a provisional improvement in patients suffering from CD, giving credit to the hypothesis that enteric bacteria may play an etiological role in Crohn's disease (McKay, 1999). Third, NF-kB plays a pivotal role in inflammatory bowel diseases and is activated in lamina propria mononuclear cells in Crohn's disease (Schreiber et al., 1998). Fourth, the treatment of Crohn's disease is based on the use of sulfasalazine and glucocorticoids, which are both known to be NF-kB inhibitors (Auphan et al., 1995 and Wahl et al., 1998).
  • Even more recently, it has been shown that the IBD1 candidate gene encodes a protein very similar to NOD2, a member of the CED4/APAF1 superfamily (Ogura et al., 2000). The nucleotide and protein sequences of IBD1 and NOD2 in reality only diverge for a small portion right at the start of the two reported sequences. The tissue expressions of Nod2 and IBD1 can, in addition, be superimposed. These two genes (proteins) can therefore be considered to be identical. It has been demonstrated that the LRR domain of Nod2 has binding activity for bacterial lipopolysaccharides (LPS) (Inohara et al., 2000) and that deletion thereof stimulates the NFkB pathway. This result confirms the data of the invention.
  • The tissue expression of IBD1 was then studied by Northern blotting. A 4.5 kb transcript is visible in most human tissues. The size of the transcript is in accordance with the size predicted by the cDNA. The 4.5 kb transcript appears to be very poorly abundant in the small intestine and the colon. It is, on the other hand, very strongly expressed in white blood cells. This is in agreement with clinical data on transplants which suggest that Crohn's disease is potentially a disease associated with circulating immune cells. In fact, bowel transplantation does not prevent recurrence on the transplant in Crohn's disease, whereas bone marrow transplantation appears to have a beneficial effect on the progression of the disease.
  • Certain data also call to mind alternative splicing, which may prove to be an important element in the possibility of generating mutants which may play a role in the development of inflammatory diseases.
  • The promoter of the IBD1 gene has not currently been identified with precision. It is, however, reasonable to think, by analogy with a very large number of genes, that this promoter lies, at least partly, immediately upstream of the gene, in the 5′ portion thereof. This genetic region contains transcribed sequences, as witnessed by the presence of ESTs (HUMGS01037, AA835524, hs.105242, SHGC17274, hs.146128, hs.122983, hs.87280). The ATCC clones containing these sequences were sequenced and analyzed in the laboratory, making it possible to demonstrate an exon and intron organization with possible alternative splicings. These data suggest the existence of another gene (named IBD1prox due to its proximity to IBD1). The partial sequence of the complementary DNA of IBD1prox is reported (SEQ ID No. 4), as is its intron-exon structure, on SEQ ID No. 6.
  • Translation of the cDNAs corresponding to IBD1prox results in a protein containing a homeobox. Analysis of several cDNAs of the gene suggests, however, the existence of alternative splicings. IBD1prox, according to one of the possible alternative splicings, corresponds to the anonymous EST HUMGS01037, the RNA of which is expressed more strongly in differentiated leukocytic lines than in undifferentiated lines.
  • Thus, it is possible that this gene may have a role in inflammation and cell differentiation. It may therefore also, itself, be considered to be a good candidate for susceptibility to IBD. The association between CD and the polymorphism ctg35ExC located on the coding sequence of IBD1prox supports this hypothesis even though this polymorphism does not cause any sequence variation at the protein level.
  • Finally, more recently, the existence of a genetic linkage in families suffering from Crohn's disease and not comprising any mutation in the IBD1 gene also, itself, suggests that IBD1prox has a role in addition to IBD1 in genetic predisposition to the disease.
  • The functional relationship between IBD1 and IBD1prox is not currently established. However, the considerable proximity between the two genes may reflect an interaction between them. In this case, the “head-to-tail” location of these genes suggests that they may have common or interdependent methods of regulation.
  • Example 6 Identification of IBD1 Gene Mutations in Inflammatory Diseases
  • In order to confirm the role of IBD1 in inflammatory diseases, the coding sequence and the intron-exon junctions of the gene were sequenced from exon 2 to exon 12 inclusive, in 70 independent individuals, namely: 50 sick individuals suffering from CD, 10 sick individuals suffering from UC, 1 sick individual suffering from Blau's syndrome and 9 healthy controls. The sick individuals studied were mostly familial forms of the disease and were often carriers of the susceptibility haplotype defined by the transmission disequilibrium studies. The healthy controls were of Caucasian origin.
  • It was thus possible to identify 24 sequence variants on this group of 70 unrelated individuals (table 3).
  • The nomenclature of the mutations reported refers to the initial sequence of the protein comprising 1 041 amino acids. The more recently proposed nomenclature is easily deduced by removing 28 amino acids from the initial sequence, and therefore corresponds to a protein comprising 1 013 amino acids (cf. example 5).
  • TABLE 4
    Mutations observed in the IBD1 gene
    Nucleotide Protein Crohn's Ulcerative Health
    Exon variant variant disease colitis controls
    1 Not tested
    2 G417A Silent
    2 C537G Silent
    3 None
    4 T805C S269P 48/100  6/20 3/18
    4 A869G N290S 0 0 1/18
    4 C905T A302V 1/100 0 0
    4 C1283T P428L 1/100 0 0
    4 C1284A Silent
    4 C1287T Silent
    4 T1380C Silent
    4 T1764G Silent
    4 G1837A A613T 1/100 0 0
    4 C2107T R703W 10/10  1/20 1/18
    4 C2110T R704C 4/10  1/20 0
    5 G2365A R792Q 1/100 0 0
    5 G2370A V794M 0 1/20 0
    5 G2530A E844K 1/10  0 0
    6 A2558G N853S 1/100 0 0
    6 A2590G M864V 1/100 0 0
    7 None
    8 G2725C G909R 7/100 0 0
    8 C2756A A919D 1/100 0 0
    9 G2866A V9561 2/100 1/20 3/18
    10 C2928T Silent
    11 3022insC Stop 20/100  0 0
    12 none
  • The mutations other than silent mutations observed in each exon are reported. They are indicated by the variation in the peptide chain. For each mutation and for each phenotype studied, the number of times where the mutation is observed, related to the number of chromosomes tested, is indicated.
  • No functional sequence variant was identified in exons 1 to 3 (corresponding to the CARD domain of the protein). Exons 7 and 12 did not show any sequence variation either. Certain variants corresponded to polymorphisms already identified and typed for transmission disequilibrium studies, namely:
      • Snp3-2931: nucleotide variant T805C, protein variant S269P
      • ctg2931-5ag/ola: nucleotide variant T1380C (silent)
      • ctg2931-3ac/ola: nucleotide variant T1746G (silent)
      • SNP1: nucleotide variant C2107T, protein variant R703W.
  • Several sequence variations were silent (G417A, C537G, C1284A, C1287T, T1380C, T1764G and C2928T) and did not lead to any modification of the protein sequence. They were not studied further here.
  • For the 16 non-silent sequence variations, protein sequence variants were observed in 43/50 CD versus 5/9 healthy controls, and 6/10 UC. The existence of one or more sequence variation(s) appeared to be associated with the CD phenotype. Several sequence variations often existed in the same individual suffering from CD, suggesting a sometimes recessive effect of the gene for CD. On the other hand, no composite heterozygote or homozygote was observed among the patients suffering from UC or among the healthy controls.
  • Some non-silent variants were present both in the sick individuals suffering from UC or from CD and in the healthy individuals. They were the variants S269P, N290S, R703W and V956I located in exons 2, 4 and 9. Further information therefore appears to be necessary before selecting a possible functional role for these sequence variants.
  • V956I is a conservative sequence variation (aliphatic amino acids).
  • The sequence variant S269P corresponds to a variation in amino acid class (hydroxylated to immuno acid) at the beginning of the nucleotide-binding domain. This sequence variant and CD are in transmission disequilibrium. It is in fact the polymorphism Snp3 (cf. above).
  • R703W results in a modification of the amino acid class (aromatic instead of basic). This modification occurs in the intermediate region between the NBD and LRR domains, which is a region conserved between IBD1 and CARD4/NOD1. A functional role may therefore be suspected for this polymorphism. This sequence variation (corresponding to the polymorphic site Snpl) is transmitted to sick individuals suffering from CD more often than at random (cf. above), confirming that this polymorphism is associated with CD. It is possible that the presence of this mutant in healthy individuals reflects incomplete penetration of the mutation as is expected for complex genetic diseases such as chronic inflammatory bowel diseases.
  • The variant R704C, located immediately next to R703W, could be identified in both CD and UC. It also, itself, corresponds to a nonconservative variation of the protein (sulfur-containing amino acid instead of basic amino acid) on the same protein region, suggesting a functional effect for R704C which is as important as that for R703W.
  • Other sequence variations are specific for CD, for UC or for Blau's syndrome.
  • Some sequence variations are, on the contrary, rare, present in one or a few sick individuals (A613T, R704C, E844K, N853S, M864V, A919D). They are always variations leading to nonconservative modifications of the protein in leucine-rich domains, at positions which are important within these domains. These various elements suggest that these variations have a functional role.
  • Two sequence variations (G909R and L1008P*) are found in quite a large number of Crohn's diseases (respectively 7/50 and 16/50) whereas they are not detected in the controls or in the individuals suffering from UC.
  • The deletion/insertion of a guanosine at codon 1008 results in transformation of the third leucine of the alpha helix of the last LRR to proline followed by a STOP codon (L1008P*). This sequence variation therefore leads to an important modification of the protein: decrease in size of the protein (protein having a truncated LRR domain) and modification of a very conserved amino acid (leucine). This sequence modification is associated with CD, as witnessed by a transmission disequilibrium study in 16 families carrying the mutation (P=0.008).
  • The mutation G909R occurs on the last amino acid of the sixth LRR motif. It replaces an aliphatic amino acid with a basic amino acid. This variation is potentially important given the usually neutral or polar nature of the amino acids in the terminal position of the leucine-rich motifs (both for IBD1 and for NOD1/CARD4) and the conserved nature of this amino acid on the IBD1 and NOD1/CARD4 proteins.
  • In Blau's syndrome, the sick individuals (n=2) of the family studied carried a specific sequence variation (L470F) located in exon 4 and corresponding to the NBD domain of the protein. In this series, this sequence variant was specific for Blau's syndrome.
  • In UC, several sequence variants not found in healthy individuals were also identified. The proportion of sick individuals carrying a mutation was smaller than for CD, as expected given the less strongly established linkage between IBD1 and UC, and the supposedly less genetic nature of the latter disease. Sequence variations were common to CD and to UC (R703W, R704C). Others, on the other hand, appeared to be specific for UC (V794M). This observation makes it possible to confirm that CD and UC are diseases which, at least partly, share the same genetic predisposition. It lays down the foundations of a nosological classification for IBDs.
  • The study of the sequence variants of the IBD1 gene has therefore made it possible to identify several variants having a very probable functional effect (for example: truncated protein) and associated with Crohn's disease, with UC and with Blau's syndrome.
  • The promoter of the gene is not currently determined. In all probability, however, it is likely to be located in the 5′ region upstream of the gene. According to this hypothesis, the sequence variants observed in this region may have a functional effect. This may explain the very strong association between CD and certain polymorphic loci, such as ctg35ExC or Ctg25Ex1.
  • The invention thus provides the first description of mutations in the family of genes containing a CARD domain in humans. The frequency of these mutations in various inflammatory diseases shows that the IBD1 gene has an essential role in normal and pathological inflammatory processes. This invention provides new paths of understanding and of research in the field of the physiopathology of normal and pathological inflammatory processes. As a result, it makes it possible to envision the development of new pharmaceutical molecules which regulate the effector pathways controlled by IBD1 and which are useful in the treatment of inflammatory diseases and in the regulation of inflammatory processes in general.
  • Example 7 Bases for a Biological Diagnosis of Susceptibility to Crohn's Disease
  • More recently, 457 independent patients suffering from Crohn's disease, 159 independent patients suffering from ulcerative colitis and 103 healthy controls were studied in the search for mutations. This study made it possible to confirm the mutations previously reported and to identify additional mutations, reported in FIG. 4. The main mutations were then genotyped in 235 families suffering from Crohn's disease. This more recent study is reported using, as reference, the shorter protein sequence (1 013 amino acids, see example 5), but the prior nomenclature for the mutations is easily deduced from the latter by adding 28 to the number indicating the position of the amino acids.
  • Among the 5 most common mutations, the conservative mutation V928I (formerly V956I) is not significantly associated with one or the other of the inflammatory bowel diseases, and does not therefore appear to have an important role in the disease.
  • The mutation S241P (formerly S269P) is in linkage disequilibrium with the other main mutations and does not appear to play an important role, by itself, in susceptibility to inflammatory bowel diseases (data not shown).
  • Conversely, the other 3 mutations, R675W (formerly R703W), G881R (formerly G909R) and 980fs (formerly L1008P*), are significantly associated with Crohn's disease but not with ulcerative colitis (cf. below). The location in the LRR, or in its immediate proximity, of the 3 common mutations pleads very strongly in favor of a functional mechanism involving this protein domain, probably via a defect in negative regulation of NFkB by the mutated protein. The other mutations are more rare (FIG. 4). These cumulative mutations are present in 17% of the individuals suffering from Crohn's disease versus, respectively, 4% and 5% of the healthy individuals or individuals suffering from ulcerative colitis. A large number of rare mutations are also located in the LRR.
  • The intrafamily studies of the three polymorphisms most common in Crohn's disease show that all three are associated with the disease (table 5). As expected, for a mutation supposed to be very deleterious, the polymorphism most strongly associated is the truncating mutation. These three polymorphisms are independently associated with Crohn's disease, since it was not possible to identify, on 235 families, chromosomes carrying more than one of these three mutations. The independent nature of these associations considerably supports the hypothesis that the IBD1 gene is clearly involved in genetic predisposition to Crohn's disease.
  • TABLE 5
    Study of the 3 common polymorphisms of IBD1 in
    235 families suffering from Crohn's disease
    MUTATION VALUE p OF THE PDT TEST
    R675W 0.001
    G881R 0.003
    980fs 0.000006
  • The case-control studies confirm this association (table 6). They show that the mutations most common in Crohn's disease are not common in ulcerative colitis.
  • TABLE 6
    Case-control study of the 3 common polymorphisms of IBD1 in inflammatory bowel diseases
    No. OF FREQUENCY OF FREQUENCY OF FREQUENCY OF TOTAL
    CHROMOSOMES THE ALLELE AT THE ALLELE AT THE ALLELE AT ALLELES
    MUTATION STUDIED RISK R675W RISK G881R RISK 980fs AT RISK
    Healthy 206 0.04 0.01 0.02 0.07
    controls
    Ulcerative 318 0.03 0.00 0.01 0.05
    colitis
    Crohn's 936 0.11 0.06 0.12 0.29
    disease
  • The study of the dose-effect of these mutations shows that individuals carrying a mutation in the homozygous or composite heterozygous state exhibit a much greater risk of developing the disease than individuals who are not carrying or are heterozygous for these mutations (table 7).
  • TABLE 7
    Relative and absolute risk of Crohn's disease
    attributable as a function of the genotype of IBD1
    In the general population, a risk of Crohn's disease of
    0.001 has been taken as a reference, and it has been presumed
    that the mutations are in Hardy-Weinberg equilibrium.
    Distribution
    GENOTYPE
    SIMPLE COMPOSITE
    No HETERO- HOMO- HETERO-
    VARIANT ZYGOTE ZYGOTE ZYGOTE
    Healthy 88 15 0 0
    Ulcerative 145 13 1 0
    colitis
    Crohn's 267 133 28 40
    disease
    Attributable
    risk of CD:
    Relative risk 1 3 38 44
    Absolute risk 0.0007 0.002 0.03 0.03
  • The studies mentioned above confirm the prior preliminary data and provide the detailed bases for a biological diagnosis of Crohn's disease by studying the IBD1 variants. In fact, this work:
    • 1) defines the mutations, the frequency of which is greater than 0.001 in a mixed Caucasian population;
    • 2) defines the frequency of the mutations observed and makes it possible to define 3 main mutations associated with Crohn's disease. Thus, it is possible, by virtue of this work, to define a strategy for studying the gene in order to search for morbid variants, namely: firstly, typing the 3 main mutations; secondly, searching for mutations in the last 7 exons; thirdly, searching for other sequence variants;
    • 3) defines the practical modalities for searching for these mutations by pointing out their position and their nature. In fact, it is then easy for those skilled in the art to develop typing and sequencing methods according to their personal expertise. Mention may in particular be made of the possibility of genotyping the three main mutations by PCR followed by enzymatic digestion and electrophoresis, study of the migration profiles by dHPLC, DGGE or SSCP, oligoligation, microsequencing, etc.;
    • 4) demonstrates the independence of the most common mutations which are not observed on the same chromosome in this extended and varied population. This information makes it possible to reliably classify the individuals who are composite heterozygotes (having two mutations) as carriers with a double dose of intragenic variations;
    • 5) demonstrates that the great majority of the mutations only lead to a null or minimal effect on the risk of ulcerative colitis. This result makes it possible to envision assisting the clinician in the differential diagnosis between these two diseases. In fact, in approximately 10% of cases, inflammatory bowel diseases remain unclassified despite biological, radiological and endoscopic examination;
    • 6) defines a relative and absolute risk of disease for the most common genotypes. This result lays down the foundations of a predictive diagnosis potentially useful in an approach of preventive monitoring and intervention in populations at risk, in particular the relatives of sick individuals;
    • 7) demonstrates the existence of a dose-effect for the IBD1 gene and confirms the partly recessive nature of genetic predisposition to Crohn's disease. It therefore makes it possible to lay the foundations for genetic counseling and for intra-familial preclinical diagnosis.
  • Finally, it should be noted that an additional mutation of the NBD domain was isolated in a second family carrying Blau's syndrome. The rareness of the two events in 2 different families is sufficient to confirm the involvement of this gene in Blau's syndrome and in granulomatous diseases in general.
  • All of these data provide a diagnostic tool which is directly applicable and of use to the practitioner in his or her daily practice.
  • The IBD1prox gene, located in the promoter region of IBD1, and the partial sequence of which is disclosed in the present invention, may also, itself, have an important role in the regulation of cellular apoptosis and of the inflammatory process, as suggested by its differential expression in mature cells of the immune system. The strong association reported in this work between the polymorphism marker ctg35ExC (located in the transcribed region of the gene) and Crohn's disease also pleads very strongly in favor of this hypothesis.
  • Inflammatory bowel diseases are complex genetic diseases for which, until now, no susceptibility gene had been identified with certainty. The invention has made it possible to identify the first gene for susceptibility to Crohn's disease, using a positional cloning (or reverse genetics) approach. This is the first genetic location obtained using such an approach for a complex genetic disease, which demonstrates its usefulness and its feasibility, at least in certain cases in complex genetic diseases.
  • The present invention also relates to a purified or isolated nucleic acid, characterized in that it encodes a polypeptide possessing a continuous fragment of at least 200 amino acids of a protein chosen from SEQ ID No. 2 and SEQ ID No. 5.
  • REFERENCES
    • Auphan et al. (1995), Science 270, 286-90.
    • Asakawa et al. (1997), Gene, 191, 69.
    • Becker et al. (1998), Proc. Natl. Acad. Sci. USA, 95, 9979.
    • Bertin et al. (1999), J. Biol. Chem., 274, 12955.
    • Buckholz (1993), Curr. Op. Biotechnology 4, 538.
    • Carter, (1993), Curr. Op. Biotechnology 3, 533.
    • Cho et al. (1998), Proc. Natl. Acad. Sci. USA, 95, 7502.
    • Duck et al. (1990), Biotechniques, 9, 142.
    • Edwards and Aruffo (1993), Curr. Op. Biotechnology, 4, 558.
    • Epstein (1992), Medecine/Sciences, 8, 902.
    • Guatelli et al. (1990), Proc. Natl. Acad. Sci. USA 87: 1874.
    • Hugot et al. (1996), Nature, 379, 821.
    • Inohara et al. (1999), J. Biol. Chem., 274, 14560.
    • Inohara et al. (2000), J. Biol. Chem.
    • Kievitis et al. (1991), J. Virol. Methods, 35, 273.
    • Kim et al. (1996), Genomics, 34, 213.
    • Köhler and Milstein (1975), Nature, 256, 495.
    • Kwoh et al. (1989), Proc. Natl. Acad. Sci. USA, 86, 1173.
    • Landegren et al. (1988), Science 241, 1077.
    • Lander and Kruglyak (1995), Nat. Genet., 11, 241.
    • Luckow (1993), Curr. Op. Biotechnology 4, 564.
    • Martin et al. (2000), Am. J. Hum. Genet. 67: 146-54.
    • Matthews et al. (1988), Anal. Biochem., 169, 1-25.
    • McKay (1999), Gastroenterol. 13, 509-516.
    • Miele et al. (1983), J. Mol. Biol., 171, 281.
    • Neddleman and Wunsch (1970), J. Mol. Biol. 48: 443.
    • Ogura et al. (2000), J. Biol. Chem.
    • Olins and Lee (1993), Curr. Op. Biotechnology 4: 520.
    • Perricaudet et al. (1992), La Recherche 23: 471.
    • Pearson and Lipman (1988), Proc. Natl. Acad. Sci. USA 85: 2444.
    • Poltorak et al. (1998), Sciences 282, 2085-8.
    • Rioux et al. (1998), Gastroenterology, 115: 1062.
    • Rohlmann et al. (1996), Nature Biotech. 14: 1562.
    • Rolfs, A. et al. (1991), Berlin: Springer-Verlag.
    • Rouquier et al. (1994), Anal. Biochem. 217, 205.
    • Sambrook et al. (1989), Molecular cloning: a laboratory manual, 2nd ed. Cold Spring Harbor Lab., Cold Spring Harbor, N.Y..
    • Satsangi et al. (1996), Nat. Genet., 14: 199.
    • Schreiber et al. (1998), Gut 42, 477-84.
    • Segev (1992), Kessler C. Springer Verlag, Berlin, N.Y., 197-205.
    • Smith and Waterman (1981) Ad. App. Math. 2: 482.
    • Steward and Yound (1984), Solid phase peptides synthesis, Pierce Chem. Company, Rockford, 111, 2nd ed. (1984).
    • Spielman et al. (1993), Am. J. Hum. Genet., 52, 506.
    • Sundberg et al. (1994), Gastroenterology, 107, 1726-35.
    • Temin (1986), Retrovirus vectors for gene transfer. In Kucherlapati R., ed. Gene Transfer, New York, Plenum Press, 149-187.
    • Tromp et al. (1996), Am. J. Hum. Genet., 59: 1097.
    • Wahl et al. (1998), B. J. Olin. Invest. 101, 1163-74.
    • Walker (1992), Nucleic Acids Res. 20: 1691.

Claims (17)

1-25. (canceled)
26. A purified or isolated nucleic acid probe, wherein said nucleic acid probe is specific for a variant nucleic acid sequence selected from the group consisting of SEQ ID NO:3 having a C to T mutation at nucleotide 16467, SEQ ID NO:3 having a G to C mutation at nucleotide 27059, and SEQ ID NO:3 having a C insertion at nucleotide 34296.
27. The nucleic acid probe of claim 26, wherein said nucleic acid probe is specific for SEQ ID NO:3 having a C to T mutation at nucleotide 16467.
28. The nucleic acid probe of claim 26, wherein said nucleic acid probe is specific for SEQ ID NO:3 having a G to C mutation at nucleotide 27059.
29. The nucleic acid probe of claim 26, wherein said nucleic acid probe is specific for SEQ ID NO:3 having a C insertion at nucleotide 34296.
30. The nucleic acid probe of claim 26, wherein said nucleic acid probe is about 15 to 30 nucleotides in length.
31. The nucleic acid probe of claim 26, wherein said nucleic acid probe comprises a detectable label.
32. The nucleic acid probe of claim 31, wherein said detectable label is selected from the group consisting of a radioactive isotope, a ligand, and a luminescent agent.
33. The nucleic acid probe of claim 32, wherein said ligand is selected from the group consisting of biotin, avidin, streptavidin, dioxygenin, a hapten, and a dye.
34. The nucleic acid probe of claim 32, wherein said luminescent agent is selected from the group consisting of a radioluminescent agent, a chemiluminescent agent, a bioluminescent agent, a fluorescent agent, and a phosphorescent agent.
35. The nucleic acid probe of claim 26, wherein said nucleic acid probe is immobilized on a support.
36. A kit comprising the nucleic acid probe of claim 26.
37. The kit of claim 36, wherein said kit comprises nucleic acid probes specific for at least two of said variant nucleic acid sequences.
38. The kit of claim 36, wherein said kit comprises nucleic acid probes specific for three of said variant nucleic acid sequences.
39. A DNA chip comprising the nucleic acid probe of claim 26.
40. The DNA chip of claim 39, wherein said DNA chip comprises nucleic acid probes specific for at least two of said variant nucleic acid sequences.
41. The DNA chip of claim 39, wherein said DNA chip comprises nucleic acid probes specific for three of said variant nucleic acid sequences.
US13/406,487 2000-03-27 2012-02-27 Genes involved in inflammatory bowel diseases and use thereof Abandoned US20130035260A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/406,487 US20130035260A1 (en) 2000-03-27 2012-02-27 Genes involved in inflammatory bowel diseases and use thereof

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
FR0003832 2000-03-27
FR0003832A FR2806739B1 (en) 2000-03-27 2000-03-27 GENES INVOLVED IN INFLAMMATORY BOWEL DISEASES AND THEIR USE
US10/240,046 US7592437B2 (en) 2000-03-27 2001-03-27 Genes involved in intestinal inflammatory diseases and use thereof
PCT/FR2001/000935 WO2001072822A2 (en) 2000-03-27 2001-03-27 Genes involved in intestinal inflammatory diseases and use thereof
US12/370,543 US8137915B2 (en) 2000-03-27 2009-02-12 Genes involved in intestinal inflammatory diseases and use thereof
US13/406,487 US20130035260A1 (en) 2000-03-27 2012-02-27 Genes involved in inflammatory bowel diseases and use thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/370,543 Continuation US8137915B2 (en) 2000-03-27 2009-02-12 Genes involved in intestinal inflammatory diseases and use thereof

Publications (1)

Publication Number Publication Date
US20130035260A1 true US20130035260A1 (en) 2013-02-07

Family

ID=8848516

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/240,046 Expired - Lifetime US7592437B2 (en) 2000-03-27 2001-03-27 Genes involved in intestinal inflammatory diseases and use thereof
US12/370,543 Expired - Lifetime US8137915B2 (en) 2000-03-27 2009-02-12 Genes involved in intestinal inflammatory diseases and use thereof
US13/406,487 Abandoned US20130035260A1 (en) 2000-03-27 2012-02-27 Genes involved in inflammatory bowel diseases and use thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/240,046 Expired - Lifetime US7592437B2 (en) 2000-03-27 2001-03-27 Genes involved in intestinal inflammatory diseases and use thereof
US12/370,543 Expired - Lifetime US8137915B2 (en) 2000-03-27 2009-02-12 Genes involved in intestinal inflammatory diseases and use thereof

Country Status (11)

Country Link
US (3) US7592437B2 (en)
EP (1) EP1268784B1 (en)
JP (1) JP5150997B2 (en)
AT (1) ATE291086T1 (en)
AU (2) AU8727201A (en)
CA (1) CA2404448C (en)
DE (1) DE60109430T2 (en)
FR (1) FR2806739B1 (en)
NZ (1) NZ522011A (en)
WO (1) WO2001072822A2 (en)
ZA (1) ZA200207821B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11268149B2 (en) 2004-12-08 2022-03-08 Cedars-Sinai Medical Center Diagnosis and treatment of inflammatory bowel disease

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6818750B2 (en) 1999-09-01 2004-11-16 The Burnham Institute Card proteins involved in cell death regulation
US7163801B2 (en) 1999-09-01 2007-01-16 The Burnham Institute Methods for determining the prognosis for cancer patients using tucan
FR2806739B1 (en) * 2000-03-27 2005-02-18 Fond Jean Dausset Ceph GENES INVOLVED IN INFLAMMATORY BOWEL DISEASES AND THEIR USE
US7196182B2 (en) 2000-05-24 2007-03-27 The Burnham Institute Card domain containing polypeptides, encoding nucleic acids, and methods of use
EP1297129A2 (en) * 2000-05-24 2003-04-02 The Burnham Institute Card domain containing polypeptides, encoding nucleic acids, and methods of use
EP1404712B1 (en) * 2000-10-30 2014-06-18 The Regents Of The University Of Michigan Nod2 nucleic acids and proteins
DK1330525T3 (en) * 2000-10-30 2013-03-04 Univ Michigan NOD2 nucleic acids and proteins
US7060442B2 (en) * 2000-10-30 2006-06-13 Regents Of The University Of Michigan Modulators on Nod2 signaling
US20060154276A1 (en) 2004-05-13 2006-07-13 Prometheus Laboratories Inc. Methods of diagnosing inflammatory bowel disease
EP2359137A4 (en) 2008-11-11 2012-05-30 Prometheus Lab Inc Methods for prediction of inflammatory bowel disease (ibd) using serologic markers
WO2010120814A1 (en) * 2009-04-14 2010-10-21 Prometheus Laboratories Inc. Inflammatory bowel disease prognostics
AU2010266028B2 (en) 2009-06-25 2015-04-30 Société des Produits Nestlé S.A. Methods for diagnosing irritable bowel syndrome
WO2011060098A1 (en) 2009-11-10 2011-05-19 Prometheus Laboratories Inc. Methods for predicting post-surgery risk associated with ileal pouch-anal anastomosis
AU2011317149B2 (en) 2010-10-18 2015-05-07 Société des Produits Nestlé S.A. Methods for determining anti-drug antibody isotypes
DK2676137T3 (en) 2011-02-17 2015-01-19 Nestec Sa TESTS TO DETECT AUTO ANTIBODIES FOR ANTI-TNF PHARMACEUTICALS
CN103649336A (en) 2011-05-10 2014-03-19 雀巢产品技术援助有限公司 Methods of disease activity profiling for personalized therapy management
MX352274B (en) 2011-10-21 2017-11-16 Nestec Sa Methods for improving inflammatory bowel disease diagnosis.

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5474796A (en) * 1991-09-04 1995-12-12 Protogene Laboratories, Inc. Method and apparatus for conducting an array of chemical reactions on a support surface
US6060246A (en) * 1996-11-15 2000-05-09 Avi Biopharma, Inc. Reagent and method for isolation and detection of selected nucleic acid sequences
US6117986A (en) * 1998-06-10 2000-09-12 Intergen Company, L.P. Pyrimidines linked to a quencher
US20010053519A1 (en) * 1990-12-06 2001-12-20 Fodor Stephen P.A. Oligonucleotides
US7592437B2 (en) * 2000-03-27 2009-09-22 Fondation Jean Dausset-Ceph Genes involved in intestinal inflammatory diseases and use thereof

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999023255A1 (en) * 1997-10-31 1999-05-14 Cedars-Sinai Medical Center Methods of identifying and diagnosing inflammatory bowel disease or subtypes with the nramp locus
US6469140B1 (en) * 1998-02-06 2002-10-22 Millennium Pharmaceuticals, Inc. Molecules of the card-related protein family and uses thereof
US6262333B1 (en) * 1998-06-10 2001-07-17 Bayer Corporation Human genes and gene expression products
WO2000058473A2 (en) * 1999-03-31 2000-10-05 Curagen Corporation Nucleic acids including open reading frames encoding polypeptides; 'orfx'
EP1074617A3 (en) 1999-07-29 2004-04-21 Research Association for Biotechnology Primers for synthesising full-length cDNA and their use
MXPA02009271A (en) 2000-03-24 2003-03-12 Genetics Inst A novel p selectin glycoprotein ligand (psgl 1) binding protein and uses therefor.
US6812339B1 (en) * 2000-09-08 2004-11-02 Applera Corporation Polymorphisms in known genes associated with human disease, methods of detection and uses thereof
DK1330525T3 (en) * 2000-10-30 2013-03-04 Univ Michigan NOD2 nucleic acids and proteins
EP1404712B1 (en) * 2000-10-30 2014-06-18 The Regents Of The University Of Michigan Nod2 nucleic acids and proteins

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20010053519A1 (en) * 1990-12-06 2001-12-20 Fodor Stephen P.A. Oligonucleotides
US5474796A (en) * 1991-09-04 1995-12-12 Protogene Laboratories, Inc. Method and apparatus for conducting an array of chemical reactions on a support surface
US6060246A (en) * 1996-11-15 2000-05-09 Avi Biopharma, Inc. Reagent and method for isolation and detection of selected nucleic acid sequences
US6117986A (en) * 1998-06-10 2000-09-12 Intergen Company, L.P. Pyrimidines linked to a quencher
US7592437B2 (en) * 2000-03-27 2009-09-22 Fondation Jean Dausset-Ceph Genes involved in intestinal inflammatory diseases and use thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
NEB Catalog 1998/1999 pages 121 and 284. *
Rothstein (PNAS 1994 Vol 91 pages 4155-4159) *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11268149B2 (en) 2004-12-08 2022-03-08 Cedars-Sinai Medical Center Diagnosis and treatment of inflammatory bowel disease

Also Published As

Publication number Publication date
US8137915B2 (en) 2012-03-20
DE60109430T2 (en) 2006-04-06
CA2404448C (en) 2014-03-25
JP2003528631A (en) 2003-09-30
WO2001072822A2 (en) 2001-10-04
ZA200207821B (en) 2003-06-12
NZ522011A (en) 2005-11-25
WO2001072822A3 (en) 2002-07-18
AU2001287272B2 (en) 2006-09-21
AU8727201A (en) 2001-10-08
FR2806739A1 (en) 2001-09-28
FR2806739B1 (en) 2005-02-18
US20030190639A1 (en) 2003-10-09
EP1268784A2 (en) 2003-01-02
DE60109430D1 (en) 2005-04-21
ATE291086T1 (en) 2005-04-15
US7592437B2 (en) 2009-09-22
US20100159453A1 (en) 2010-06-24
EP1268784B1 (en) 2005-03-16
JP5150997B2 (en) 2013-02-27
CA2404448A1 (en) 2001-10-04
WO2001072822B1 (en) 2002-09-06

Similar Documents

Publication Publication Date Title
US8137915B2 (en) Genes involved in intestinal inflammatory diseases and use thereof
US9115400B2 (en) LMNA gene and its involvement in Hutchinson-Gilford Progeria Syndrome (HGPS) and arteriosclerosis
EP0756488B1 (en) Detection of alterations of tumor suppressor genes for the diagnosis of cancer
JP4017672B2 (en) Polymorphisms and novel genes in the region of human hemochromatosis genes
US8975018B2 (en) Method of detecting expanded CAG repeat region in spinocerebellar ataxia-2 gene
WO1997042314A1 (en) Nucleic acid encoding spinocerebellar ataxia-2 and products related thereto
JP5965594B2 (en) Cloning, expression and identification of the SPG gene capable of responding to the most common forms of autosomal dominant hereditary spastic paraplegia
JPH07143884A (en) Tumor suppressor gene merlin and its application
US20030082720A1 (en) Compositions methods and kits relating to treating and diagnosing hypertension
US6844431B1 (en) Nucleic acid encoding spinocerebellar ataxia-2 and products related thereto
JP2006506988A (en) Human type II diabetes gene located on chromosome 5q35-SLIT-3
JPH07289297A (en) Cancer-suppressing gene
US7151161B1 (en) Human genes of chromosome 11q13.3
US6440699B1 (en) Prostate cancer susceptible CA7 CG04 gene
US7700748B2 (en) VMGLOM gene and its mutations causing disorders with a vascular component
WO2001029266A2 (en) Identification of arsacs mutations and methods of use therefor
WO2001016291A2 (en) 07cg27 gene

Legal Events

Date Code Title Description
AS Assignment

Owner name: FONDATION JEAN DAUSSET-CEPH, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HUGOT, JEAN-PIERRE;THOMAS, GILLES;ZOUALI, MOHAMED;AND OTHERS;REEL/FRAME:031905/0387

Effective date: 20021220

STCB Information on status: application discontinuation

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