WO2002018640A2 - Gene called aladin, involved in allgrove syndrome, its expression product and their applications - Google Patents

Gene called aladin, involved in allgrove syndrome, its expression product and their applications Download PDF

Info

Publication number
WO2002018640A2
WO2002018640A2 PCT/IB2001/001574 IB0101574W WO0218640A2 WO 2002018640 A2 WO2002018640 A2 WO 2002018640A2 IB 0101574 W IB0101574 W IB 0101574W WO 0218640 A2 WO0218640 A2 WO 0218640A2
Authority
WO
WIPO (PCT)
Prior art keywords
aladin
gene
seq
sequence
syndrome
Prior art date
Application number
PCT/IB2001/001574
Other languages
French (fr)
Other versions
WO2002018640A3 (en
Inventor
Anastella Tullio-Pelet
Rémi SALOMON
Smail Hadj-Rabia
Stanislas Lyonnet
Arnold Munnich
Original Assignee
Institut National De La Sante Et De La Recherche Medicale (Inserm)
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 Institut National De La Sante Et De La Recherche Medicale (Inserm) filed Critical Institut National De La Sante Et De La Recherche Medicale (Inserm)
Publication of WO2002018640A2 publication Critical patent/WO2002018640A2/en
Publication of WO2002018640A3 publication Critical patent/WO2002018640A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • 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
    • 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 the identification of a novel gene, called ALADIN, involved in AUgrove syndrome and of the ALADIN gene products, as well as to the diagnostic and therapeutic applications thereof.
  • AUgrove syndrome is an autosomal recessive condition causing Adrenal insufficiency, Achalasia and Alacrima (Triple A). Life-threatening hypoglycemic episodes and severe feeding difficulties related to achalasia and gastric atonia frequently occur in the first decade.
  • Triple A syndrome results from the mutation of a gene involved in the development of the autonomic nervous system, namely : i) the cholinergic innervation of most affected organs, ii) the abnormal regulation of postural systolic blood pressure, and iii) the occasional anomaly of pupillary reflexes. Late-onset progressive neurological symptoms including cerebellar ataxia, peripheral neuropathy and mild dementia have suggested that the central nervous system could be involved in the disease as well.
  • the disease gene has been mapped to chromosome 12q13 (Weber et al, 1996 ; Stratakis et al, 1997).
  • the transcript contains an open-reading frame (ORF) which encodes a protein of 547 amino acids the sequence of which is close to a sequence incidentally disclosed by Sugano et al, (EMBL/Genbank/DDBJ databases/ access number AK 000833) on the one hand, and by Li et al (EMBL/Genbank/DDBJ databases/AF 226048) on the other hand. Determination of exon-intron boundaries was performed through sequence comparison between cDNA clones and genomic DNA, which led to the identification of 16 exons.
  • ORF open-reading frame
  • a subject of the present invention is thus an isolated nucleic acid the sequence of which is selected from the group consisting of sequences SEQ ID n° 1 to SEQ ID n° 17.
  • SEQ ID n° 1 represents a fragment of the genomic DNA of the ALADIN gene including exon 1 (nucleotide 102 to 224).
  • SEQ ID n° 2 represents a fragment of the genomic DNA of the ALADIN gene including exon 2 (nucleotide 101 to 228).
  • SEQ ID n° 3 represents a fragment of the genomic DNA of the ALADIN gene including exon 3 (nucleotide 101 to 156).
  • SEQ ID n° 4 represents a fragment of the genomic DNA of the
  • ALADIN gene including exon 4 (nucleotide 101 to 192).
  • SEQ ID n° 5 represents a fragment of the genomic DNA of the ALADIN gene including exon 5 (nucleotide 101 to 147).
  • SEQ ID n° 6 represents a fragment of the genomic DNA of the ALADIN gene including exon 6 (nucleotide 101 to 199).
  • SEQ ID n° 7 represents a fragment of the genomic DNA of the ALADIN gene including exon 7 (nucleotide 101 to 244).
  • SEQ ID n° 8 represents a fragment of the genomic DNA of the ALADIN gene including exon 8 (nucleotide 101 to 221).
  • SEQ ID n° 9 represents a fragment of the genomic DNA of the
  • ALADIN gene including exon 9 (nucleotide 101 to 225).
  • SEQ ID n° 10 represents a fragment of the genomic DNA of the ALADIN gene including exon 10 (nucleotide 99 to 159).
  • SEQ ID n° 11 represents a fragment of the genomic DNA of the ALADIN gene including exon 11 (nucleotide 101 to 195).
  • SEQ ID n° 12 represents a fragment of the genomic DNA of the _4 4D/N gene including exon 12 (nucleotide 105 to 293).
  • SEQ ID n° 13 represents a fragment of the genomic DNA of the ALADIN gene including exon 13 (nucleotide 101 to 168).
  • SEQ ID n° 14 represents a fragment of the genomic DNA of the ALADIN gene including exon 14 (nucleotide 101 to 182).
  • SEQ ID n° 15 represents a fragment of the genomic DNA of the
  • ALADIN gene including exon 15 (nucleotide 101 to 185).
  • SEQ ID n° 16 represents a fragment of the genomic DNA of the ALADIN gene including exon 16 (nucleotide 101 to 358).
  • SEQ ID n° 17 is the promoter of the ALADIN gene.
  • the invention further relates to a method of in vitro diagnosis of a disorder of AUgrove syndrome's family, wherein : - either one or more mutation(s) is detected in the ALADIN gene defined as comprising any sequences from SEQ ID n° 1 to n° 17, by means of at least one nucleotide probe or primer which specifically hybridizes with any of said sequences ;
  • the present invention also provides pharmaceutical compositions comprising an ALADIN polypeptide, or a nucleic acid encoding said polypeptide.
  • a further subject-matter of the invention is a pharmaceutical composition comprising an anti-sense sequence or an antibody directed against the ALADIN gene product.
  • Figure 1 shows mutant Triple A chromosome haplotypes in North
  • FIG. 1b physical map of the Triple A critical region shows STSs, polymorphic markers, BACs and genes.
  • BAG clone AC021103 that contains the Triple A ORF is 170 kb in length.
  • Genomic organization of the ALADIN gene is shown underneath (ATG: translation initiation codon; TAG: stop codon). Introns (filled line) and exons (rectangles) are drawn to scale. The position of the mutations is indicated.
  • Figure 2 shows a sequence analysis and identification of ALADIN gene mutations in Triple A patients. One representative pedigree is shown for each of the five mutations identified. DNA sequence of a control and a homozygote patient (mutant) are presented. Heterozygosity for sequence alterations have been confirmed by DNA sequence analysis in both parents for each of the kindreds studied.
  • Figure 3a shows prediction of the WD-repeat structure of ALADIN gene product according to the bmerc program. The most significant WD-repeat is indicated by an arrow. Residues in marked columns are predicted to fold into ⁇ -strands.
  • Figure 3b shows relative expression pattern of the ALADIN mRNA as compared to UBIQUITINE (AU: arbitrary unit).
  • the nucleic acid of the invention preferably comprises, or in a preferred embodiment consists in, sequence SEQ ID n° 1 to SEQ ID n° 17.
  • the invention is also directed to the intron and exon sequences included in listed sequences SEQ ID n° 1 to n° 17.
  • homologous nucleotide sequences are also part of the present invention.
  • Such homologous sequences are defined as : i) sequences which exhibit a similarity of at least 70 % in comparison with SEQ ID n° 1 to n° 17 ; or ii) sequence which hybridize with SEQ ID n° 1 to n° 17 or their complementary sequences, under stringent conditions of hybridization.
  • such homologous sequences show at least 75 % of similarity, preferably at least 85 %, more preferably at least 90 % of similarity with SEQ ID n° 1 to n° 17.
  • a homologous nucleotide sequence of the invention specifically hybridizes to the sequences to which it refers or to their complementary sequences under stringent conditions. Parameters that define the conditions of stringency depend upon the temperature at which 50 % of annealed strands separate (T m ).
  • T m is calculated as follows :
  • hybridization temperature is preferably around from 5°C to 10° C below the calculated T m
  • hybridization buffer solutions that are used are preferably solutions with high ionic strength, such as an aqueous 6 X SSC solution for example.
  • Said homologous sequences include mammalian genes coding for the ALADIN geneproduct, preferably of primate, cattle, sheep, swine, or rodent, as well as allelic variants. Such homologous nucleic acids can be readily retrieved by polymerase chain reaction (PCR) amplification of genomic DNA extracts by conventional methods.
  • oligonucleotide primers matching upstream and downstream of the 5' and 3' ends of the domain to be amplified.
  • Suitable primers can be designed according to the nucleotide sequences SEQ ID n° 1 to SEQ ID n° 17.
  • a primer can consist of 10 to 40, preferably 15 to 25 nucleotides. It may also be advantageous to select primers containing C and G nucleotides in a proportion sufficient to ensure efficient hybridization ; e.g, an amount of C and G nucleotides of at least 40 %, preferably 50 % of the total nucleotide amount.
  • such primers may be selected from the group consisting of sequences SEQ ID n° 20 to SEQ ID n° 53.
  • nucleic acid sequences of the invention are useful for the detection of an abnormality, such as a mutation, in the ALADIN gene or in the transcripts of the ALADIN gene. Such an analysis allows in vitro diagnosis of disorders associated with said abnormality.
  • a subject of the present invention is a method of in vitro diagnosis of a disorder associated with an abnormality in the ALADIN gene or in the transcripts of the ALADIN gene, wherein one or more mutation(s), preferably inducing a modification of the expression of the ALADIN gene, is detected in the ALADIN gene or in the transcripts of the ALADIN gene.
  • the authors of the present invention have more particularly investigated the abnormalities in ALADIN gene or in the transcripts of the ALADIN gene which are responsible for AUgrove syndrome.
  • ND not determined.
  • the present invention relates to methods of in vitro diagnosis wherein the nucleic acid sequences of the invention or probes or primers derived thereof are used to detect aberrant synthesis or genetic abnormalities such as genetic rearrangement at the ALADIN gene level.
  • the present invention is more particularly directed to a method of in vitro diagnosis comprising the steps of :
  • the method of the invention can also be applied to the detection of an abnormality in the transcript of the ALADIN gene, by amplifying the mRNAs contained in a biological sample, for example by RT-PCR.
  • - producing cDNA from mRNA contained in a biological sample - contacting said cDNA with specific oligonucleotides (primers) permitting the amplification of all or part of the transcript of the ALADIN gene, under conditions permitting a hybridization of the primers with said cDNA ;
  • - detecting the amplification products ; - comparing the amplified products as obtained to the amplified products obtained with a normal control biological sample, and thereby detecting a possible abnormality in the transcript of the ALADIN gene.
  • This comparison of the amplified products obtained from the biological sample with the amplified products obtained with a normal biological sample can be carried out for example by specific probe hybridization, by sequencing or by restriction site analysis.
  • One skilled in the art knows very well the standard methods for analysing the DNA contained in a biological sample and for diagnosing a genetic disorder. Many strategies for genotypic analysis are available (Antonarakis et al., 1989, Cooper et al., 1991 ).
  • DGGE Denaturing Gradient Gel Electrophoresis
  • SSCP Single Strand Conformation Polymorphism
  • DHPLC Method Denaturing high-performance liquid chromatography
  • RT-PCR method may be advantageously used for detecting abnormalities in the ALADIN transcript, as it allows to visualize the consequences of a splicing mutation such as exon skipping or aberrant splicing due to the activation of a cryptic site.
  • This method is preferably followed by direct sequencing as well.
  • the more recently developped technique using DNA chip can also be advantageously implemented for detecting an abnormality in the ALADIN gene (Bellis et al., 1997).
  • the cloning of the ALADIN gene, as well as the identification of various mutations responsible for various disorders according to the invention, allow direct or semi-direct diagnosis.
  • the specificity and reliability of such diagnosis methods are more particularly appreciable for prenatal diagnosis.
  • the nucleic acid sequences of the present invention represent a highly interesting tool for genetic counseling.
  • the ALADIN gene of the present invention codes for polypeptide having the aminoacid sequence encoded shown in SEQ ID n° 19.
  • This polypeptide can be prepared by any of the standard methods of purification of soluble proteins, by peptide synthesis or by genetic engineering.
  • Said techniques comprise the insertion of a nucleic acid sequence of SEQ ID n° 18 into an expression vector, such as a plasmid, and the transformation of host cells with the expression vector, by any of the methods available to the skilled person, like for instance electroporation.
  • Said expression vector contains a promoter sequence, signals for initiation and termination of translation, as well as appropriate regions for regulation of translation. Its insertion into the host cell may be transient or stable.
  • Host cells may be prokaryotic or eukaryotic, including but not limiting to bacteria, yeasts, insect cells, mammalian cells, including cell lines which are commercially available.
  • This polypeptide can be used to produce specific monoclonal or polyclonal antibodies, or fragments thereof, or chimeric or immunoconjugate antibobies.
  • Polyclonal antibodies can be obtained from serum of an animal immunized against the ALADIN gene product, which can be produced by genetic engineering for example, as above described, according to standard methods well-known by one skilled in the art.
  • Monoclonal antibodies can be obtained according to the standard method of hybridoma culture (Kohler and Milstein, 1975).
  • Said antibodies are particularly useful for detecting or purifying an ALADIN gene product polypeptide according to the invention in a biological sample.
  • a further subject of the present invention is thus a method of diagnosis comprising the steps of :
  • This immunoassay can be carried out according to any standard technique well-known by one skilled in the art, such as ELISA (Enzyme-Linked Immunosorbent Assays), E.I.A. (Enzyme-lmmunoAssays), R.I.A. (RadiolmmunoAssays) and the like. Sandwich-type immunoassays are particularly advantageous.
  • the antibodies directed against the ALADIN gene product can be detectably labelled, either directly (for example by means of coupling with a radioisotope, an enzyme, biotine, ...) or indirectly (for example by means of a secondary labelled antibody).
  • Another subject of the present invention is a pharmaceutical composition
  • a pharmaceutical composition comprising, as an active agent, the ALADIN polypeptide, in association with a pharmaceutically acceptable carrier.
  • a further subject of the present invention is a pharmaceutical composition
  • a pharmaceutical composition comprising, as an active agent, a nucleic acid encoding said polypeptide and a pharmaceutically acceptable carrier.
  • Said nucleic acid preferably inserted in a vector, may be administered in a naked form or in association with transfection facilitating agents.
  • a further subject of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising, as an active agent, an anti-sense sequence capable of specifically hybridizing with SEQ ID n° 18, in association with a pharmaceutically acceptable carrier.
  • a still further subject of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising, as an active agent, an antibody directed against said the ALADIN gene product, in association with a pharmaceutically acceptable carrier.
  • compositions of the invention are useful for preventing and/or treating disorders, wherein the ALADIN gene or its expression product is implicated, as above described.
  • compositions of the invention may be administered to a mammal, preferably to a human, in need of a such treatment, according to a dosage which may vary widely as a function of the age, weight and state of health of the patient, the nature and severity of the complaint and the route of administration.
  • the appropriate unit forms of administration comprise oral forms such as tablets, gelatin capsules, powders, granules and oral suspensions or solutions, sublingual and buccai administration forms, subcutaneous, intramuscular, intravenous, intranasal or intraoccular administration forms and rectal administration forms.
  • a further subject of the present invention is a method of preventing and/or treating disorders, wherein the ALADIN gene or its expression product is implicated, which comprises administering to a subject in need of a such treatment an amount of a pharmaceutical composition as above defined effective to prevent and/or alleviate said disorders.
  • Inclusion criteria were : i) laboratory evidence of glucocorticoid deficiency, ii) alacrima with positive Schirmer test, and iii) achalasia of the cardia with positive manometry or barium study, in at least one family member.
  • Genotyping Genomic DNA was extracted from blood samples with informed consent and linkage studies were performed using microsatellite DNA markers from the Genethon genetic map and the Genome Database (map available on http://www.cephb.fr/cqi-bin/wdb/ceph/systeme/form - http://www.cephb.fr/cgi- bin/wdb/ceph/systeme/form - http://research.marshfieldclinic.org/genetics/) (Dib et al, 1996). Marker informativity varied from 61 to 83 %. Genotyping with microsatellite markers was performed as described, in a 25- ⁇ l total reaction volume containing 100 ng genomic DNA (Belin et al, 1998).
  • the common ancestral haplotype was reconstructed and the age of the Triple A mutation was estimated by measuring the number of generations elapsed since the appearance of the founder mutation in the North African population, based on the earliest common ancestor as proposed by Picollo et al, (1996) (ABEL program).
  • the YACs of the 12q13 region were selected from the CEPH-Genethon database (Chumakov et al, 1993 ; Burke et al, 1991) using polymorphic markers mapped to the critical region.
  • a human genomic BAC library of the CNS was screened by PCR using microsatellites, STS and ESTs.
  • the length of BAC and YAC clones was evaluated by field inversion gel electrophoresis (FIGE Mapper, Biorad, Richmond, CA, USA) under the following conditions: forward : 180V, reverse : 120 V, with a 0.1-2 s switch time for 16 h at 20°C.
  • the authors of the invention designed STSs by directly sequencing the ends of BAC clones.
  • the BAC DNA was isolated and directly sequenced (Osoegawa) and sequence data were integrated to the CNS contig.
  • BAC AC0ACA566F07 contained both the D12S1618 and D12S1604 loci that were in partial and complete linkage disequilibrium with the Triple A mutation respectively (Fig. 1b). This suggested that this BAC encompassed the centromeric boundary of the Triple A gene. Extensive sequencing revealed that five genes map to BAC AC0ACA566F07 (Fig.
  • BAC ACO566F07 The Centre National de Sequencage (CNS) BAC library was further screened using the most telomeric STS of BAC ACO566F07 (Fig. 1b).
  • This EST (1,792 bp) derives from a human adipose tissue cDNA library and encodes an hitherto unknown protein (NEDO human cDNA sequencing project, submission 2-15-2000).
  • Genomic sequence alignment and exon-prediction programs revealed that the AK000833 EST is transcribed from a 16 exon gene, mapping 30 kb telomeric to D12S1604 (Fig. 1b). This gene was regarded as a candidate in Triple A syndrome, based on fine linkage disequilibrium mapping.
  • EXAMPLE 2 Identification of mutations in unrelated patients with AUgrove syndrome
  • the other two mutations were located in canonical consensus splice sites, namely the acceptor site of intron 4 (IVS4 -2 A ⁇ G) and the donor site of intron 14 (IVS14 +1 G ⁇ A, Fig. 3 and Table 1).
  • the IVS4 -2 A ⁇ G mutation resulted in the retention of intron 4 in the mutant mRNA and in a premature translation termination 47 codons downstream exon 4.
  • the IVS14 +1 G ⁇ A mutation produced various abnormal transcripts, which resulted in premature translation termination upstream exon 16. This latter mutation was found in 9/10 families of North African ancestry carrying allele 5 at the D12S1604 locus, thus confirming our prediction of linkage disequilibrium.
  • the gene product is expected to contain four putative WD- repeats (Fig. 3a).
  • Northern blot and dot blot studies showed that the gene is widely expressed but much stronger signals were observed in endocrine and neuroendocrine derivatives (adrenal and pituitary), the cerebellum and the corpus callosum (Fig. 3b).
  • the MC2R could play a role in addressing of the MC2R to the membrane as suggested by the involvement of several WD-repeat proteins in intracellular membrane trafficking (Smith et al, 1999). Furthermore, since the MC2R is expressed in the adrenal cortex only, the ALADIN gene product may be associated with other membrane receptors in other tissues such as the lacrymal glands, oesophageal cardia, and brain.
  • Linkage and homozygosity mapping allowed the authors of the invention to reduce the Triple A gene interval to 3.9 cM only, while strong linkage disequilibrium further reduced this critical region to a 180 kb BAC contig, as all North African patients shared a rare genotype at the D12S1604 locus mapping to this BAC contig.
  • EST AK00833 maps 30 kb telomeric to D12S1604 locus on the contig, ii) is strongly expressed in organs and tissues targeted in the disease, and iii) is consistently mutated in the patients, it is now clear that this EST, now referred to as ALADIN (for ALacrima - Achalasia - ADrenal INsuffisiency - Neurologic disorders), corresponds to the disease causing gene.
  • ALADIN for ALacrima - Achalasia - ADrenal INsuffisiency - Neurologic disorders

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Analytical Chemistry (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • General Health & Medical Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Microbiology (AREA)
  • Pathology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Cell Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to the identification of a novel gene, called ALADIN involved in Allgrove syndrome, and of the ALADIN gene products, as well as to the diagnostic and therapeutic applications thereof. More particularly, the invention relates to a method of in vitro diagnosis of a disorder of Allgrove syndrome's family, wherein either one or more mutation(s) is detected in the ALADIN gene, or an abnormal expression of ALADIN gene product is detected.

Description

Gene called ALADIN, involved in AUgrove syndrome, its expression product and their applications
FIELD OF THE INVENTION
The present invention relates to the identification of a novel gene, called ALADIN, involved in AUgrove syndrome and of the ALADIN gene products, as well as to the diagnostic and therapeutic applications thereof.
BACKGROUND OF THE INVENTION
AUgrove syndrome is an autosomal recessive condition causing Adrenal insufficiency, Achalasia and Alacrima (Triple A). Life-threatening hypoglycemic episodes and severe feeding difficulties related to achalasia and gastric atonia frequently occur in the first decade. Several lines of evidence suggest that Triple A syndrome results from the mutation of a gene involved in the development of the autonomic nervous system, namely : i) the cholinergic innervation of most affected organs, ii) the abnormal regulation of postural systolic blood pressure, and iii) the occasional anomaly of pupillary reflexes. Late-onset progressive neurological symptoms including cerebellar ataxia, peripheral neuropathy and mild dementia have suggested that the central nervous system could be involved in the disease as well.
The disease gene has been mapped to chromosome 12q13 (Weber et al, 1996 ; Stratakis et al, 1997).
SUMMARY OF THE INVENTION
Linkage analyses and homozygosity mapping allowed the authors of the present invention to refine the mapping of the Triple A locus to the 3.9 cM interval defined by two loci. They have at last cloned the gene responsible for AUgrove syndrome, which they have called ALADIN gene for ALacrima - Achalasia - ADrenal INsuffisiency - Neurologic disorders. The transcript contains an open-reading frame (ORF) which encodes a protein of 547 amino acids the sequence of which is close to a sequence incidentally disclosed by Sugano et al, (EMBL/Genbank/DDBJ databases/ access number AK 000833) on the one hand, and by Li et al (EMBL/Genbank/DDBJ databases/AF 226048) on the other hand. Determination of exon-intron boundaries was performed through sequence comparison between cDNA clones and genomic DNA, which led to the identification of 16 exons.
A subject of the present invention is thus an isolated nucleic acid the sequence of which is selected from the group consisting of sequences SEQ ID n° 1 to SEQ ID n° 17.
SEQ ID n° 1 represents a fragment of the genomic DNA of the ALADIN gene including exon 1 (nucleotide 102 to 224).
SEQ ID n° 2 represents a fragment of the genomic DNA of the ALADIN gene including exon 2 (nucleotide 101 to 228).
SEQ ID n° 3 represents a fragment of the genomic DNA of the ALADIN gene including exon 3 (nucleotide 101 to 156). SEQ ID n° 4 represents a fragment of the genomic DNA of the
ALADIN gene including exon 4 (nucleotide 101 to 192).
SEQ ID n° 5 represents a fragment of the genomic DNA of the ALADIN gene including exon 5 (nucleotide 101 to 147).
SEQ ID n° 6 represents a fragment of the genomic DNA of the ALADIN gene including exon 6 (nucleotide 101 to 199).
SEQ ID n° 7 represents a fragment of the genomic DNA of the ALADIN gene including exon 7 (nucleotide 101 to 244).
SEQ ID n° 8 represents a fragment of the genomic DNA of the ALADIN gene including exon 8 (nucleotide 101 to 221). SEQ ID n° 9 represents a fragment of the genomic DNA of the
ALADIN gene including exon 9 (nucleotide 101 to 225).
SEQ ID n° 10 represents a fragment of the genomic DNA of the ALADIN gene including exon 10 (nucleotide 99 to 159).
SEQ ID n° 11 represents a fragment of the genomic DNA of the ALADIN gene including exon 11 (nucleotide 101 to 195).
SEQ ID n° 12 represents a fragment of the genomic DNA of the _4 4D/N gene including exon 12 (nucleotide 105 to 293). SEQ ID n° 13 represents a fragment of the genomic DNA of the ALADIN gene including exon 13 (nucleotide 101 to 168).
SEQ ID n° 14 represents a fragment of the genomic DNA of the ALADIN gene including exon 14 (nucleotide 101 to 182). SEQ ID n° 15 represents a fragment of the genomic DNA of the
ALADIN gene including exon 15 (nucleotide 101 to 185).
SEQ ID n° 16 represents a fragment of the genomic DNA of the ALADIN gene including exon 16 (nucleotide 101 to 358).
SEQ ID n° 17 is the promoter of the ALADIN gene. In the annexed sequence listing, the cDNA coding sequence
(SEQ ID n° 18), and the aminoacid corresponding sequence (SEQ ID n° 19) are also shown.
The invention further relates to a method of in vitro diagnosis of a disorder of AUgrove syndrome's family, wherein : - either one or more mutation(s) is detected in the ALADIN gene defined as comprising any sequences from SEQ ID n° 1 to n° 17, by means of at least one nucleotide probe or primer which specifically hybridizes with any of said sequences ;
- or an abnormal expression of ALADIN gene product defined as comprising aminoacid sequence SEQ ID n° 19 is detected.
The present invention also provides pharmaceutical compositions comprising an ALADIN polypeptide, or a nucleic acid encoding said polypeptide.
A further subject-matter of the invention is a pharmaceutical composition comprising an anti-sense sequence or an antibody directed against the ALADIN gene product. BRIEF DESCRIPTION OF THE DRAWINGS :
Figure 1 shows mutant Triple A chromosome haplotypes in North
African patients, physical map of the Triple A interval at 12 13, and genomic organization of ALADIN gene.
In Figure 1a, mutant haplotypes in North African families (16 chromosomes studied) at the Triple A locus show linkage disequilibrium with allele 5 at locus D12S1604.
In Figure 1b, physical map of the Triple A critical region shows STSs, polymorphic markers, BACs and genes. BAG clone AC021103 that contains the Triple A ORF is 170 kb in length. Genomic organization of the ALADIN gene is shown underneath (ATG: translation initiation codon; TAG: stop codon). Introns (filled line) and exons (rectangles) are drawn to scale. The position of the mutations is indicated.
Figure 2 shows a sequence analysis and identification of ALADIN gene mutations in Triple A patients. One representative pedigree is shown for each of the five mutations identified. DNA sequence of a control and a homozygote patient (mutant) are presented. Heterozygosity for sequence alterations have been confirmed by DNA sequence analysis in both parents for each of the kindreds studied.
Figure 3a shows prediction of the WD-repeat structure of ALADIN gene product according to the bmerc program. The most significant WD-repeat is indicated by an arrow. Residues in marked columns are predicted to fold into β-strands.
Figure 3b shows relative expression pattern of the ALADIN mRNA as compared to UBIQUITINE (AU: arbitrary unit).
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
The nucleic acid of the invention preferably comprises, or in a preferred embodiment consists in, sequence SEQ ID n° 1 to SEQ ID n° 17. The invention is also directed to the intron and exon sequences included in listed sequences SEQ ID n° 1 to n° 17.
The homologous nucleotide sequences are also part of the present invention. Such homologous sequences are defined as : i) sequences which exhibit a similarity of at least 70 % in comparison with SEQ ID n° 1 to n° 17 ; or ii) sequence which hybridize with SEQ ID n° 1 to n° 17 or their complementary sequences, under stringent conditions of hybridization. Preferably, such homologous sequences show at least 75 % of similarity, preferably at least 85 %, more preferably at least 90 % of similarity with SEQ ID n° 1 to n° 17.
Preferably a homologous nucleotide sequence of the invention specifically hybridizes to the sequences to which it refers or to their complementary sequences under stringent conditions. Parameters that define the conditions of stringency depend upon the temperature at which 50 % of annealed strands separate (Tm).
For sequences comprising more than 30 nucleotides, Tm is calculated as follows : Tm = 81.5 + 0.41 (% G + C) + 16.6 Log (positive ion concentration) - 0.63 (% formamide) - (600/polynucleotide size in base pairs) (Sambrook et al, 1989).
For sequences comprising less than 30 nucleotides, Tm is calculated as follows :
Tm= 4(G + C) + 2 (A + T). Under appropriate stringent conditions avoiding the hybridization of non specific sequences, hybridization temperature is preferably around from 5°C to 10° C below the calculated Tm, and hybridization buffer solutions that are used are preferably solutions with high ionic strength, such as an aqueous 6 X SSC solution for example. Said homologous sequences include mammalian genes coding for the ALADIN geneproduct, preferably of primate, cattle, sheep, swine, or rodent, as well as allelic variants. Such homologous nucleic acids can be readily retrieved by polymerase chain reaction (PCR) amplification of genomic DNA extracts by conventional methods. This involves the use of synthetic oligonucleotide primers matching upstream and downstream of the 5' and 3' ends of the domain to be amplified. Suitable primers can be designed according to the nucleotide sequences SEQ ID n° 1 to SEQ ID n° 17.
Typically, a primer can consist of 10 to 40, preferably 15 to 25 nucleotides. It may also be advantageous to select primers containing C and G nucleotides in a proportion sufficient to ensure efficient hybridization ; e.g, an amount of C and G nucleotides of at least 40 %, preferably 50 % of the total nucleotide amount.
As an example, such primers may be selected from the group consisting of sequences SEQ ID n° 20 to SEQ ID n° 53.
The nucleic acid sequences of the invention are useful for the detection of an abnormality, such as a mutation, in the ALADIN gene or in the transcripts of the ALADIN gene. Such an analysis allows in vitro diagnosis of disorders associated with said abnormality.
A subject of the present invention is a method of in vitro diagnosis of a disorder associated with an abnormality in the ALADIN gene or in the transcripts of the ALADIN gene, wherein one or more mutation(s), preferably inducing a modification of the expression of the ALADIN gene, is detected in the ALADIN gene or in the transcripts of the ALADIN gene.
The authors of the present invention have more particularly investigated the abnormalities in ALADIN gene or in the transcripts of the ALADIN gene which are responsible for AUgrove syndrome.
The following table reports a non exhaustive spectrum of mutations in the ALADIN gene : Table 1 - ALADIN gene mutations
Family Geographic Number of Age at Clinical Consanguinity Allele at the Mutation Location / Nature origin affected diagnosis features D12S1604 locus (homozygous)
(years) 1 France 1 4 4A yes ND _VS4-2A→G intron 4 (splice acceptor)
2 Algeria 1 6 3A yes 5/5 1024C→T (R312X) exon 9 (nonsense)
3 Turquia 3 ND /ND /2 3A yes 2/2 1071insT exon 10 (frameshift)
4 Algeria 2 20 / 14 3A yes 5/5 IVS14+1G→A intron 14 (splice donor)
5 Algeria 2 ND 3A yes 5/5 id id
6 Algeria 1 ND 3A yes 5/5 id id
7 Algeria 1 9 3A yes 5/5 id id
8 Algeria 1 7 3A yes 5/5 id id
9 Algeria 2 9 / 3 4A / 3A no 5/5 id id
10 Tunisia 1 5 4A yes 5/5 id id
11 Tunisia 2 5 / 3 3A / 2A yes 5/5 id id
12 Tunisia 2 6 / 9 3A / 2A no 5/5 id id
13 Spain 1 ND 3A no 5/5 id id
14 Portugal 1 30 4A yes 5/5 1522C→T (R478X) exon 16 (nonsense)
ND: not determined. 4A: Achalasia, Alacrima, Adrenal insufficiency, Autonomous neuropathy and others neurologic manifestations; 3A: the first three only; 2A: the first two only. Allele at the D12S1604 locus: 5 = 265 bp, 2 = 272 bp.
The present invention relates to methods of in vitro diagnosis wherein the nucleic acid sequences of the invention or probes or primers derived thereof are used to detect aberrant synthesis or genetic abnormalities such as genetic rearrangement at the ALADIN gene level.
The present invention is more particularly directed to a method of in vitro diagnosis comprising the steps of :
- contacting a biological sample containing DNA with specific oligonucleotides (primers) permitting the amplification of all or part of the ALADIN gene, the DNA contained in the sample having being rendered accessible, where appropriate, to hybridization, and under conditions permitting a hybridization of the primers with the DNA contained in the biological sample ;
- amplifying said DNA ; - detecting the amplification products ;
- comparing the amplified products as obtained to the amplified products obtained with a normal control biological sample, and thereby detecting a possible abnormality in the ALADIN gene.
The method of the invention can also be applied to the detection of an abnormality in the transcript of the ALADIN gene, by amplifying the mRNAs contained in a biological sample, for example by RT-PCR.
So another subject of the present invention is a method of in vitro diagnosis comprising the steps of :
- producing cDNA from mRNA contained in a biological sample ; - contacting said cDNA with specific oligonucleotides (primers) permitting the amplification of all or part of the transcript of the ALADIN gene, under conditions permitting a hybridization of the primers with said cDNA ;
- amplifying said cDNA ;
- detecting the amplification products ; - comparing the amplified products as obtained to the amplified products obtained with a normal control biological sample, and thereby detecting a possible abnormality in the transcript of the ALADIN gene. This comparison of the amplified products obtained from the biological sample with the amplified products obtained with a normal biological sample can be carried out for example by specific probe hybridization, by sequencing or by restriction site analysis. One skilled in the art knows very well the standard methods for analysing the DNA contained in a biological sample and for diagnosing a genetic disorder. Many strategies for genotypic analysis are available (Antonarakis et al., 1989, Cooper et al., 1991 ).
Preferably, one can use the DGGE method (Denaturing Gradient Gel Electrophoresis), the SSCP method (Single Strand Conformation Polymorphism) and DHPLC Method (Denaturing high-performance liquid chromatography) for detecting an abnormality in the ALADIN gene. Such methods are preferably followed by direct sequencing. The RT-PCR method may be advantageously used for detecting abnormalities in the ALADIN transcript, as it allows to visualize the consequences of a splicing mutation such as exon skipping or aberrant splicing due to the activation of a cryptic site. This method is preferably followed by direct sequencing as well. The more recently developped technique using DNA chip can also be advantageously implemented for detecting an abnormality in the ALADIN gene (Bellis et al., 1997).
The cloning of the ALADIN gene, as well as the identification of various mutations responsible for various disorders according to the invention, allow direct or semi-direct diagnosis. The specificity and reliability of such diagnosis methods are more particularly appreciable for prenatal diagnosis. The nucleic acid sequences of the present invention represent a highly interesting tool for genetic counseling.
Defects in the ALADIN gene, or in the ALADIN gene product cause AUgrove syndrome involving cerebellar ataxia, mild dementia or neuropathy. More generally, this gene could be involved in neurodegenerative diseases, as well as in diseases such as familial glucocorticoϊd insufficiency or oesophagal achalasia. All these diseases involving the ALADIN gene product are here designated as disorders of AUgrove syndrome's family. The ALADIN gene of the present invention codes for polypeptide having the aminoacid sequence encoded shown in SEQ ID n° 19.
This polypeptide can be prepared by any of the standard methods of purification of soluble proteins, by peptide synthesis or by genetic engineering. Said techniques comprise the insertion of a nucleic acid sequence of SEQ ID n° 18 into an expression vector, such as a plasmid, and the transformation of host cells with the expression vector, by any of the methods available to the skilled person, like for instance electroporation. Said expression vector contains a promoter sequence, signals for initiation and termination of translation, as well as appropriate regions for regulation of translation. Its insertion into the host cell may be transient or stable.
These various control signals are selected according to the host cell which may be inserted into vectors which self-replicate in the selected host cell, or into vectors which integrate the genome of said host. Host cells may be prokaryotic or eukaryotic, including but not limiting to bacteria, yeasts, insect cells, mammalian cells, including cell lines which are commercially available.
This polypeptide can be used to produce specific monoclonal or polyclonal antibodies, or fragments thereof, or chimeric or immunoconjugate antibobies.
Polyclonal antibodies can be obtained from serum of an animal immunized against the ALADIN gene product, which can be produced by genetic engineering for example, as above described, according to standard methods well-known by one skilled in the art. Monoclonal antibodies can be obtained according to the standard method of hybridoma culture (Kohler and Milstein, 1975).
Said antibodies are particularly useful for detecting or purifying an ALADIN gene product polypeptide according to the invention in a biological sample.
They are more particularly useful for detecting an abnormal expression of the ALADIN gene product in connection with a disorder of AUgrove syndrome's family. A further subject of the present invention is thus a method of diagnosis comprising the steps of :
- contacting a biological sample from a person to be tested with at least one antibody specifically directed against an ALADIN polypeptide, said antibody being detectably labelled ;
- detecting the formation of immune complexes, and thereby assaying the quantity of ALADIN polypeptide in said sample ;
- comparing the quantity of ALADIN polypeptide in said sample with the quantity of ALADIN polypeptide in a control sample ; any differences being correlated with AUgrove syndrome's family diseases.
This immunoassay can be carried out according to any standard technique well-known by one skilled in the art, such as ELISA (Enzyme-Linked Immunosorbent Assays), E.I.A. (Enzyme-lmmunoAssays), R.I.A. (RadiolmmunoAssays) and the like. Sandwich-type immunoassays are particularly advantageous. For that purpose, the antibodies directed against the ALADIN gene product can be detectably labelled, either directly (for example by means of coupling with a radioisotope, an enzyme, biotine, ...) or indirectly (for example by means of a secondary labelled antibody).
Another subject of the present invention is a pharmaceutical composition comprising, as an active agent, the ALADIN polypeptide, in association with a pharmaceutically acceptable carrier.
A further subject of the present invention is a pharmaceutical composition comprising, as an active agent, a nucleic acid encoding said polypeptide and a pharmaceutically acceptable carrier. Said nucleic acid, preferably inserted in a vector, may be administered in a naked form or in association with transfection facilitating agents.
A further subject of the invention is a pharmaceutical composition comprising, as an active agent, an anti-sense sequence capable of specifically hybridizing with SEQ ID n° 18, in association with a pharmaceutically acceptable carrier.
A still further subject of the invention is a pharmaceutical composition comprising, as an active agent, an antibody directed against said the ALADIN gene product, in association with a pharmaceutically acceptable carrier.
The pharmaceutical compositions of the invention are useful for preventing and/or treating disorders, wherein the ALADIN gene or its expression product is implicated, as above described.
The pharmaceutical compositions of the invention may be administered to a mammal, preferably to a human, in need of a such treatment, according to a dosage which may vary widely as a function of the age, weight and state of health of the patient, the nature and severity of the complaint and the route of administration.
The appropriate unit forms of administration comprise oral forms such as tablets, gelatin capsules, powders, granules and oral suspensions or solutions, sublingual and buccai administration forms, subcutaneous, intramuscular, intravenous, intranasal or intraoccular administration forms and rectal administration forms.
A further subject of the present invention is a method of preventing and/or treating disorders, wherein the ALADIN gene or its expression product is implicated, which comprises administering to a subject in need of a such treatment an amount of a pharmaceutical composition as above defined effective to prevent and/or alleviate said disorders.
The below examples illustrate the invention without limiting its scope.
EXAMPLE 1 : Cloning of the ALADIN gene :
1. Experimental procedures Patients
A total of 21 patients and 58 relatives from 14 families were included in the study. Ten families originated from North Africa (Algeria : 7 families and Tunisia : 3 families). The other families originated from France,
Portugal, Spain and Turkey. Inclusion criteria were : i) laboratory evidence of glucocorticoid deficiency, ii) alacrima with positive Schirmer test, and iii) achalasia of the cardia with positive manometry or barium study, in at least one family member.
Genotyping Genomic DNA was extracted from blood samples with informed consent and linkage studies were performed using microsatellite DNA markers from the Genethon genetic map and the Genome Database (map available on http://www.cephb.fr/cqi-bin/wdb/ceph/systeme/form - http://www.cephb.fr/cgi- bin/wdb/ceph/systeme/form - http://research.marshfieldclinic.org/genetics/) (Dib et al, 1996). Marker informativity varied from 61 to 83 %. Genotyping with microsatellite markers was performed as described, in a 25-μl total reaction volume containing 100 ng genomic DNA (Belin et al, 1998).
Statistical and computer analyses Linkage studies were performed assuming that the disease is inherited as an autosomal recessive trait with full penetrance (normal allele frequency f = .999). Two-point lod scores were calculated using the Linkage Package (Version 5.1 , M-LINK program). For linkage disequilibrium, allele frequencies in mutant chromosomes were compared to 60 control chromosomes in 30 unrelated individuals originating from the same ethnic background. Significance of the test was estimated using the standard χ2 test. The common ancestral haplotype was reconstructed and the age of the Triple A mutation was estimated by measuring the number of generations elapsed since the appearance of the founder mutation in the North African population, based on the earliest common ancestor as proposed by Picollo et al, (1996) (ABEL program).
Comparison of the nucleotide ALADIN cDNA sequence to known genes and ESTs was performed using the BLAST program in a non-redundant compilation of the EMBL and GenBank databases. Amino-acid comparisons were carried out with the non-redundant Swiss-Prot database using the BLASTP program. Multiple alignments were performed using CLUSTAL W, protein-domain homologies and motifs predicted using the Prosite and bmerk databases, and membrane topology using the PSORT program. Physical mapping and BAC contig construction
For physical mapping of the Triple A locus, the YACs of the 12q13 region were selected from the CEPH-Genethon database (Chumakov et al, 1993 ; Burke et al, 1991) using polymorphic markers mapped to the critical region. A human genomic BAC library of the CNS was screened by PCR using microsatellites, STS and ESTs. The length of BAC and YAC clones was evaluated by field inversion gel electrophoresis (FIGE Mapper, Biorad, Richmond, CA, USA) under the following conditions: forward : 180V, reverse : 120 V, with a 0.1-2 s switch time for 16 h at 20°C. The authors of the invention designed STSs by directly sequencing the ends of BAC clones. The BAC DNA was isolated and directly sequenced (Osoegawa) and sequence data were integrated to the CNS contig.
2. Results :
Linkage analysis allowed the authors of the present invention to refine the mapping of the Triple A locus to the 3.9 cM interval defined by loci D12S368 and D12S312 (Zmax = 10.89 at θ = 0, at locus D12S1604, Fig. 1a). All affected individuals born to consanguineous parents were homozygotes for each of the markers comprised in this region. Moreover, each of the affected individuals in eight North African inbred families shared a 1 cM common ancestral haplotype (haplotype 5-2-5-4 at loci D12S1604, D12S1651 , D12S1586 and D12S325 respectively, Fig. 1a). Interestingly, all North African patients were homozygotes for an unfrequent allele (allele 5) at the D12S1604 locus (versus 9.1 % in control North African chromosomes, p<0.001 , Fig.1a). These data supported an ancient founder effect in Triple A families of North African ancestry and strongly suggested that the disease gene maps at (or close to) the D12S1604 locus. In order to construct a detailed physical map of the region, the authors of the invention screened YAC and BAC libraries with the microsatellites DNA markers of the D12S1618-D12S1651 interval (Fig. 1 b). Three YACs encompassed the entire Triple A interval (YACs 813h5, 927f5, and 903f1), while the region in full linkage disequilibrium was included into YAC 903f1 only (1.7 Mb, Fig. 1 b). Interestingly, BAC AC0ACA566F07 contained both the D12S1618 and D12S1604 loci that were in partial and complete linkage disequilibrium with the Triple A mutation respectively (Fig. 1b). This suggested that this BAC encompassed the centromeric boundary of the Triple A gene. Extensive sequencing revealed that five genes map to BAC AC0ACA566F07 (Fig. 1 b), namely the SP1 (UniGene Hs.2021), ZPK (Hs.211601 ), polyRC (Hs.63525), TAR (Hs.326), and AMHR (Hs.123014) genes, but none of them showed sequence variation in our patients and no other significant ORF could be detected in this BAC, suggesting that the Triple A locus was telomeric to D12S1604, most probably in the D12S1604- D12S1651 genetic interval.
The Centre National de Sequencage (CNS) BAC library was further screened using the most telomeric STS of BAC ACO566F07 (Fig. 1b). Two BAC clones (1035M9 and 541 P23) were identified that contained a novel EST (AK000833, Fig 1b). This EST (1,792 bp) derives from a human adipose tissue cDNA library and encodes an hitherto unknown protein (NEDO human cDNA sequencing project, submission 2-15-2000). Genomic sequence alignment and exon-prediction programs revealed that the AK000833 EST is transcribed from a 16 exon gene, mapping 30 kb telomeric to D12S1604 (Fig. 1b). This gene was regarded as a candidate in Triple A syndrome, based on fine linkage disequilibrium mapping.
EXAMPLE 2 : Identification of mutations in unrelated patients with AUgrove syndrome
1. Experimental procedure Mutation screening Intronic primers derived from the sequences of the 15 ALADIN gene introns were designed by aligning the cDNA sequence of EST AK000833 with the genomic sequence (http://www-shgc.stanford.edu). Primers and PCR conditions were selected using the Oligo 5.0 program (NBI): exon 1 , 5'- GTCCGCATACGAATCTAGCCC -3' (SEQ ID n° 20) and 5'- AGACTCTGTGACCCTGCCCCT-3' (SEQ ID n° 21) ; exon 2, 5'- ATCTCTTATACTTAGCCCAGC-3' (SEQ ID n° 22) and 5'- GAATAAAAGTCTTTTGAAGAACAC -3' (SEQ ID n° 23) ; exon 3, 5'- GTGCCCATATTTTTAATACAT-3' (SEQ ID n° 24) and 5'- GGCTAAAAGAGGCTGAGCCAA-3' (SEQ ID n° 25) ; exon 4, 5'- GGACCTGGGGAGTGCCTGTCC-3' (SEQ ID n° 26) and 5'- AAGTCATCACACCCAACCCTG-3' (SEQ ID n 27); exon 5, 5'- TGACTTCAGAGGGTAGGAGTG-3' (SEQ ID n° 28) and 5'- GAGCAATCAGAAAAGGAGTAG-3' (SEQ ID n° 29) ; exon 6, 5'- AAAAGTAGGAAAAAACCATGT-3' (SEQ ID n° 30) and 5'- CATGAATCAGGTTCAAGAACT-3' (SEQ ID n° 31) ; exon 7, 5'- ATCATCCTTCTTCTCTGTGGCTTT-3'(SEQ ID n° 32) and 5'- GTGAGGACAAAGAACTTCTCC-3' (SEQ ID n° 33) ; exon 8, 5'- CAGAGTGGGCCATCAGGATAG G -3' (SEQ ID n° 34) and 5'- CCGAGTGAGGAACACTTTTGC C-3" (SEQ ID n° 35) ; exon 9 5'-
TTTGTACATTAGAGAGGCCAG-3' (SEQ ID n° 36) and 5'- CTAAGTTCTAAAAGTTGGACC-3' (SEQ ID n° 37) exon 10, 5'- AGAAAGGCACTTAGCTCCTGG-3' (SEQ ID n° 38) and 5'- GAATGCAGGAGGGAAAGTAGA-3' (SEQ ID n° 39) ; exon 11 , 5'- CCAATGGGCCCCTGATGCTCCCAA-3' (SEQ ID n 40) and 5'- CTCCTAAGCTTCTATATTTCC-3' (SEQ ID n° 41 ) exon 12, 5'- CTTAGGAGA1 rCGAGGTGTT-3' (SEQ ID n° 42) and 5'- GCCTCTCCCCCAAGCCTGTGGGTA-3' (SEQ ID n° 43) ; exon 13, 5'- AGATGGTGAGGAGAGGTGAGCTGA-3' (SEQ ID n° 44) and 5'- CCCAGGACATGGGCAGAGGAG-3' (SEQ ID n° 45) ; exon 14, 5'- ATCTAATGAGGCCGTGCCCTGGCC-3' (SEQ ID n° 46) and 5'- AACCGGAGGCAAGAAGGGAGC-3' (SEQ ID n° 47) ; exon 15, 5'- GGTCTGGACTTCTCACCAGCCTTC-3' (SEQ ID n° 48) and 5'- CTGGGCCCAAAGAAGGGCCACCTG-3' (SEQ ID n° 49) ; exon 16, 5'- AGTTGGATGGAGAAGCTGAGG-3' (SEQ ID n° 50) and 5'- AGTGGCTAAAAAGTGACAGAA-3' (SEQ ID n° 51). PCR-amplified DNA was sequenced on both strands using the fluorochrome PRISM kit (PE Biosystems). Electrophoresis was carried out for 12 h on a 373 automated DNA sequencer (ABI).
2. Results The 16 coding exons and intron-exon junctions were sequenced in 14 unrelated families and a total of five homozygous mutations were detected in 14/14 index cases (Fig 2 and Table 1). Each of the five mutations resulted in either a premature stop codon or a frameshift, predicted to produce a truncated, presumably, nonfunctional protein. Indeed, two nonsense mutations occurred in CpG dinucleotides in exons 9 and 16 respectively (mutations R312X and R478X). The third mutation, a single nucleotide insertion at position 1071 in exon 10 (1071 insT), resulted in translation termination 38 codons downstream the coding sequence (Fig. 2 and Table 1 ). The other two mutations were located in canonical consensus splice sites, namely the acceptor site of intron 4 (IVS4 -2 A→G) and the donor site of intron 14 (IVS14 +1 G→A, Fig. 3 and Table 1). The IVS4 -2 A→G mutation resulted in the retention of intron 4 in the mutant mRNA and in a premature translation termination 47 codons downstream exon 4. On the other hand, the IVS14 +1 G→A mutation produced various abnormal transcripts, which resulted in premature translation termination upstream exon 16. This latter mutation was found in 9/10 families of North African ancestry carrying allele 5 at the D12S1604 locus, thus confirming our prediction of linkage disequilibrium. The base changes segregated with the disease in each of the affected families studied (Fig 2) and heterozygosity was confirmed in all parents. Finally, none of the sequence alterations could be found in 60 control chromosomes matched for their ethnic background. EXAMPLE 3 : Expression of the ALADIN gene
1. Experimental procedure Northern-blot analysis EST AKOOO833 was amplified using primers 5'-
CACCATAGTCCCCTCCCTGAA-3' (SEQ ID n° 52) and 5'-CAGGGAAGGAGC TCAAACACA -3' (SEQ ID n° 53) (from exon 7 to exon 14), labelled with [32P]-dCTP and used as a probe on a human fetal and adult multiple-tissue dot blots (Clontech) and on human adult Northern blots (control probe : UBIQUITINE).
2. Results
The gene product is expected to contain four putative WD- repeats (Fig. 3a). Northern blot and dot blot studies showed that the gene is widely expressed but much stronger signals were observed in endocrine and neuroendocrine derivatives (adrenal and pituitary), the cerebellum and the corpus callosum (Fig. 3b).
The presence of 4 WD-repeat modules suggest that this protein belongs to an ancient regulatory-protein family whose members may be involved in functions as diverse as cell division, cell-fate determination, transcription or transmembrane signaling (Neer et al, 1994 ; Smith et al, 1999). Along these lines, ACTH-resistant glucocorticoid deficiency in Triple A syndrome patients may suggest that the ALADIN gene product could be involved in the ACTH signaling pathway mediated by the melanocortin 2 receptor (MC2R) (Clark et al, 1998 ; Naville et al, 1996). In particular, it could play a role in addressing of the MC2R to the membrane as suggested by the involvement of several WD-repeat proteins in intracellular membrane trafficking (Smith et al, 1999). Furthermore, since the MC2R is expressed in the adrenal cortex only, the ALADIN gene product may be associated with other membrane receptors in other tissues such as the lacrymal glands, oesophageal cardia, and brain. Conclusion
Linkage and homozygosity mapping allowed the authors of the invention to reduce the Triple A gene interval to 3.9 cM only, while strong linkage disequilibrium further reduced this critical region to a 180 kb BAC contig, as all North African patients shared a rare genotype at the D12S1604 locus mapping to this BAC contig. Since EST AK00833 : i) maps 30 kb telomeric to D12S1604 locus on the contig, ii) is strongly expressed in organs and tissues targeted in the disease, and iii) is consistently mutated in the patients, it is now clear that this EST, now referred to as ALADIN (for ALacrima - Achalasia - ADrenal INsuffisiency - Neurologic disorders), corresponds to the disease causing gene.
REFERENCES
- Antonarakis et al, (1989) Diagnosis of genetic disorders at the DNA level. N Engl J. Med. 320 : 153-163.
- Belin et al, (1998) SHOX mutations in dyschondrosteosis (Leri- Weill syndrome). Nature Genet. 19, 67-69
- Bellis et al, (1997) medecine/sciences, 13 : 1317-24.
- Burke et al, (1991) The role of Yeast artificial chromosome clones in generating genome maps. Curr. Opin. Genet. Dev. 1, 69-74
- Chumakov et al, (1993) A YAC contig map of the human genome. Nature 377, 175-197
- Clark et al, (1998) Adrenocorticotropin insensitivity syndromes. Endocrine Reviews 19, 828-843 - Cooper et al, (1991) Diagnosis of genetic disease using recombinant DNA, 3rd Edition, Hum Genet., 87: 519-560.
- Dib et al, (1996) A comprehensive genetic map of the human genome based on 5,264 microsatellites. Nature 380, 152-154
- Kόhler and Milstein, (1975), Nature vol. 256, 495-497. - Naville et al, (1996) Demonstration by transfection studies that mutations in the adrenocorticotropin receptor gene are one cause of the hereditary syndrome of glucocorticoid deficiency. J Clin Endocr Metab 81, 1442-1448
- Neer et al, (1994) The ancient regulatory-protein of WD-repeat protein family. Nature 371 , 297-300
- Osoegawa et al, (1998) An Improved Approach for construction of Bacterial Artificial Chromosome Libraries. Genomics 52, 1-8
- Piccolo et al, (1996) A founder mutation in the γ-sarcoglycan gene of Gypsies possibly predating their migration out of India. Hum. Mol. Genet. 5, 2019-2022
- Sambrook et al, (1989) In Molecular cloning: a laboratory manual, (Cold spring Harbor Laboratory Press).
- Smith et al, (1999) The WD repeat: a common architecture for diverse functions. Trends Biochem. Sci. 24,181-185
- Stratakis et al, (1997) Segregation of AUgrove (Triple-A) syndrome in Puerto Rican kindreds with chromosome 12 (12q13) polymorphic markers. Proc. Assoc. Am. Physicians 109, 478-482
- Weber et al, (1996) Linkage of the gene for the Triple A syndrome to chromosome 12q13 near the type II keratin gene cluster. Hum. Mol. Genet. 5, 2061-2066

Claims

1. Isolated nucleic acid, the sequence of which is selected from the group consisting of SEQ ID n° 1 to SEQ ID n° 17.
2. Isolated nucleic acid, the sequence of which differs from said nucleic acid sequences of claim 1 by one or more mutation(s) selected from the mutations defined in table 1.
3. A method of in vitro diagnosis of a disorder of AUgrove syndrome's family, wherein one or more mutation(s) is detected in the ALADIN gene defined as comprising any sequences from SEQ ID n° 1 to n° 17, by means of at least one nucleotide probe or primer which specifically hybridizes with any of said sequences.
4. The method of in vitro diagnosis according to claim 3 comprising the steps of :
- contacting a biological sample containing DNA with specific oligonucleotide primers permitting the amplification of all or part of the ALADIN gene, the DNA contained in the sample having being rendered accessible, where appropriate, to hybridization, and under conditions permitting a hybridization of the primers with the DNA contained in the biological sample ;
- amplifying said DNA ;
- detecting the amplification products ;
- comparing the amplified products as obtained to the amplified products obtained with a normal control biological sample, and thereby detecting a possible abnormality in the ALADIN gene.
5. A method of in vitro diagnosis of a disorder of AUgrove syndrome's family, wherein one or more mutation's is detected in the ALADIN gene transcript defined as comprising sequence SEQ ID n° 18, by means of at least one nucleotide probe or primer which specifically hybridizes with said sequence.
6. The method of in vitro diagnosis according to claim 5 comprising the steps of :
- producing cDNA from mRNA contained in a biological sample ;
- contacting said cDNA with specific oligonucleotide primers permitting the amplification of all or part of the transcript of the ALADIN gene, under conditions permitting a hybridization of the primers with said cDNA ;
- amplifying said cDNA ;
- detecting the amplification products ;
- comparing the amplified products as obtained to the amplified products obtained with a normal control biological sample, and thereby detecting a possible abnormality in the transcript of the ALADIN gene.
7. A method of in vitro diagnosis of a disorder of AUgrove syndrome's family, wherein an abnormal expression of ALADIN gene product defined as comprising aminoacid sequence SEQ ID n° 19 is detected.
8. The method of in vitro diagnosis according to claim 7, comprising the steps of :
- contacting a biological sample from a person to be tested with at least one antibody specifically directed against an ALADIN polypeptide, said antibody being detectably labelled ;
- detecting the formation of immune complexes, and thereby assaying the quantity of ALADIN polypeptide in said sample ;
- comparing the quantity of ALADIN polypeptide in said sample with the quantity of ALADIN polypeptide in a control sample ; any differences being correlated with a disorder of AUgrove syndrome's family.
9. A method for preventing and/or treating a disorder of AUgrove syndrome's family, which comprises administering to a subject in need of a such treatment an amount of a pharmaceutical composition comprising an active agent selected from the group consisting of :
- a nucleic acid comprising sequence SEQ ID n° 18 ;
- a polypeptide comprising sequence SEQ ID n° 19 ;
- an antisense sequence capable of specifically hybridizing with sequence SEQ ID n° 18 ; and
- an antibody directed against a polypeptide comprising sequence SEQ ID n° 19 ; in association with a pharmaceutically acceptable carrier.
PCT/IB2001/001574 2000-08-30 2001-08-30 Gene called aladin, involved in allgrove syndrome, its expression product and their applications WO2002018640A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US22900000P 2000-08-30 2000-08-30
US60/229,000 2000-08-30

Publications (2)

Publication Number Publication Date
WO2002018640A2 true WO2002018640A2 (en) 2002-03-07
WO2002018640A3 WO2002018640A3 (en) 2003-03-13

Family

ID=22859416

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2001/001574 WO2002018640A2 (en) 2000-08-30 2001-08-30 Gene called aladin, involved in allgrove syndrome, its expression product and their applications

Country Status (1)

Country Link
WO (1) WO2002018640A2 (en)

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
DATABASE EBI [Online] EMBL; Homo sapiens chromosome 12, 5 July 2000 (2000-07-05) MUZNY, D.M. ET AL.: "Homo sapiens chromosome 12 clone RP11-680A11, Working draft sequence, 12 unordered pieces" retrieved from EBI, accession no. AC073611 Database accession no. AC073611 XP002219648 *
DATABASE GENBANK [Online] NCBI; 21 July 2000 (2000-07-21) LI, Y. ET AL.: "A novel gene expressed in human liver non-tumor-tissue" retrieved from GENBANK, accession no. AF226048 Database accession no. AF226048 XP002219646 *
DATABASE GENBANK [Online] NCBI; 22 February 2000 (2000-02-22) TANIGAMI ET AL.: "NEDO human cDNA sequencing project" retrieved from NCBI, accession no. AK000833 Database accession no. AK000833 XP002219645 cited in the application *
DATABASE GENBANK [Online] NCBI; unnamed protein product, 22 February 2000 (2000-02-22) SUGANO, S. ET AL.: "NEDO human cDNA sequencing project" retrieved from GENBANK, accession no. BAA91394 Database accession no. BAA91394 XP002219647 *
HADJ-RABIA SMAIL ET AL: "Linkage disequilibrium in inbred North African families allows fine genetic and physical mapping of triple A syndrome." EUROPEAN JOURNAL OF HUMAN GENETICS, vol. 8, no. 8, August 2000 (2000-08), pages 613-620, XP002219643 ISSN: 1018-4813 *
STRATAKIS CONSTANTINE ET AL: "Allgrove (triple-A) syndrome in Puerto Rican kindreds maps to chromosome 12 (12q13)." PEDIATRIC RESEARCH, vol. 41, no. 4 PART 2, 1997, page 107A XP008010131 Meeting of the American Pediatric Society and the Society for Pediatric Research;Washington, D.C., USA; May 2-6, 1997 ISSN: 0031-3998 *
WEBER ANGELA ET AL: "Linkage of the gene for the triple A syndrome to chromosome 12q13 near the type II keratin gene cluster." HUMAN MOLECULAR GENETICS, vol. 5, no. 12, 1996, pages 2061-2066, XP002219644 ISSN: 0964-6906 *

Also Published As

Publication number Publication date
WO2002018640A3 (en) 2003-03-13

Similar Documents

Publication Publication Date Title
US8962269B2 (en) Spinal muscular atrophy diagnostic methods
US5763168A (en) Method to determine predisposition to hypertension
Ozelius et al. Human monoamine oxidase gene (MAOA): chromosome position (Xp21-p11) and DNA polymorphism
EP1590485A2 (en) Genetic polymorphisms associated with rheumatoid arthritis, methods of detection and uses thereof
H. Yen et al. Isolation of a new gene from the distal short arm of the human X chromosome that escapes X-inactivation
Ris-Stalpers et al. A practical approach to the detection of androgen receptor gene mutations and pedigree analysis in families with x-linked androgen insensitivity
US20050059067A1 (en) Chemical compounds
WO2004083403A2 (en) Genetic polymorphisms associated with rheumatoid arthritis, methods of detection and uses thereof
US20080261231A1 (en) Diabetes gene
US6153386A (en) Method to determine predisposition to hypertension
EP0711833B1 (en) Survival motor neuron (SMN) gene: a gene for spinal muscular atrophy
JP2002510508A (en) Glaucoma treatment and diagnostic agents
JP2005528089A (en) Peripheral artery occlusion disease genes
WO1997003086A1 (en) Early onset alzheimer&#39;s disease gene and gene products
WO2002018640A2 (en) Gene called aladin, involved in allgrove syndrome, its expression product and their applications
WO2000071751A1 (en) Diabetes gene
CA2373466C (en) Application of aprataxin gene to diagnosis and treatment for early-onset spinocerebellar ataxia (eaoh)
EP1036195A1 (en) Short gcg expansions in the pab ii gene for oculopharyngeal muscular dystrophy and diagnostic thereof
EP2275575A2 (en) Coding sequence haplotypes of the human BRCA2 gene
US5811244A (en) In vitro method for identifying a clinical disorder associated with Nhe1 mutation
US6395488B1 (en) Cholecystokinin (CCK) gene as a risk factor for smoking in women
US20040018497A1 (en) Human obesity LIPIN3 polynucleotide and polypeptide sequences and methods of use thereof
WO2005012565A1 (en) Genetic marker for coronary artery disease
WO1997034625A9 (en) Cystatin b mutants
WO1997001573A2 (en) Early onset alzheimer&#39;s disease gene and gene products

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP