CA2248016C - Direct molecular diagnosis of friedreich ataxia - Google Patents

Direct molecular diagnosis of friedreich ataxia Download PDF

Info

Publication number
CA2248016C
CA2248016C CA2248016A CA2248016A CA2248016C CA 2248016 C CA2248016 C CA 2248016C CA 2248016 A CA2248016 A CA 2248016A CA 2248016 A CA2248016 A CA 2248016A CA 2248016 C CA2248016 C CA 2248016C
Authority
CA
Canada
Prior art keywords
seq
composition
matter
molecule consisting
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA2248016A
Other languages
French (fr)
Other versions
CA2248016A1 (en
Inventor
Massimo Pandolfo
Laura Montermini
Maria Molto
Michael Koenig
Victoria Campuzano
Mireille Cossee
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.)
Institut National de la Sante et de la Recherche Medicale INSERM
Baylor College of Medicine
Original Assignee
Institut National de la Sante et de la Recherche Medicale INSERM
Baylor College of Medicine
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, Baylor College of Medicine filed Critical Institut National de la Sante et de la Recherche Medicale INSERM
Publication of CA2248016A1 publication Critical patent/CA2248016A1/en
Application granted granted Critical
Publication of CA2248016C publication Critical patent/CA2248016C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/81Packaged device or kit

Abstract

The invention relates generally to methods for the diagnosis and therapeutic treatment of Friedreich Ataxia. Friedreich ataxia (FRDA) is an autosomal recessive, degenerative disease that involves the central and peripheral nervous system and the heart. A gene, X25, was identified in the critical region for the FRDA locus on chromosome 9q13. The gene encodes a 210 amino acid protein, frataxin, that has homologues in distant species such as C.
elegans and yeast. A few FRDA patients have been found to have point mutations in X25, but the vast majority are homozygous for a variable, unstable GAA
trinucleotide expansion in the first X25 intron. Mature X25 mRNA was severely reduced in abundance in individuals with FRDA. Carriers and individuals at risk for developing FRDA can be ascertained by the methods of the present invention. Further, the methods of the present invention provide treatment to those individuals having FRDA.

Description

DIRECT MOLECULAR DIAGNOSIS OF FRIEDREICH ATAXIA
This invention was supported in part by a grant from the United ES States Government through federal funds (NINDS NS34192). The U.S.
government has certain rights to this invention.

FIELD OF THE INVENTION

This invention relates generally to methods for the diagnosis, screening and therapeutic treatment of Friedreich ataxia. Friedreich ataxia (FRDA) is an autosomal recessive. degenerative disease that involves the central and peripheral nervous system and the heart. A
gene, X25, was identified in the critical region for the FRDA locus on chromosome 9q13. The X25 gene encodes a 210 amino acid protein, frataxin, that has homologues in distant species such as C. elegans and yeast. A few FRDA patients have been found to have point mutations in X25, but the vast majority are homozygous for a variable. unstable GAA
trinucleotide expansion in the first X25 intron. Mature X25 mRNA was severely reduced in abundance in individuals with FRDA.

SUBSTITUTE SHEET (RULE 26)
2 BACKGROUND OF THE INVENTION

Friedreich ataxia (FRDA) is the most common hereditary ataxia, with an estimated prevalence of 1 in 50,000 and a deduced carrier frequency of 1/120 in the European population. FRDA is an autosomal recessive degenerative disease characterized by progressive gait and limb ataxia, a lack of tendon reflexes in the legs, loss of position sense, dysarthria, and pyramidal weakness of the legs. Hypertrophic cardiomyopathy is io found in almost all patients. Diabetes mellitus is seen in about 10% of the cases. carbohydrate intolerance in an additional 20%, and a reduced insulin response to arginine stimulation in all cases. The age of onset is usually around puberty, and almost always before age twenty-five. Most patients are wheelchair bound by their late twenties and currently there is no treatment to slow progression of the disease.
The first pathologic changes are thought to occur in the dorsal root ganglia with loss of large sensory neurons, followed by deterioration of the sensory posterior columns, spinocerebellar tracts and corticospinal motor tracts of the spinal cord, and atrophy of large sensory fibers in peripheral nerves. Only occasional mild degenerative changes are seen in the cerebellum, pons and medulla. While most symptoms are a consequence of neuronal degeneration, cardiomyopathy and diabetes are thought to reflect independent sites of primary degeneration.
Overall. the pathology of FRDA is very different from that of other hereditary ataxias, particularly the dominant forms and ataxia-telangiectasia, where the cerebellum is the primary site of degeneration.
The mutated gene in FRDA has been mapped to chromosome 9q13-q21.1. S. Chamberlain, et al., Nature, 334:248 (1988); and the FRDA
candidate region has been narrowed to a 150 kb segment flanked by the ~o Z0-2 gene (distal) and the marker F8101 (proximal), L. Montermini et al., SUBSTITUTE SHEET (RULE 26)
3 Am. J. Hum. Genet.. 57:1061 (1995). Previously proposed candidate genes are excluded: the X104/CSFA1/Z0-2 gene on the basis of the absence of deleterious mutation in patients. and the STM7 and PRKACG
genes because they lie in entirety on the centromeric side of F8101 (Figure 1A).

SUMMARY OF THE INVENTION

It is a particular object of the present invention to provide a method of screening individuals for a mutation that leads to Friedreich's ataxia, comprising determining the number of GAA repeats in an intron of the X25 gene.
It is a further object of the present invention to provide a method of screening individuals for a mutation that leads to Friedreich's ataxia, comprising the steps of measuring expression of the X25 gene at the mRNA or protein levels.
It is another object of the present invention to provide a method of screening individuals for a mutation that leads to Friedreich's ataxia, comprising the step of detecting a variation in a size of a (GAA)õ repeat in a first intron of a X25 gene by measuring the length of said repeat.
wherein n for normal individuals ranges from 1-22 and n for affected individuals is more than about 120-900.
It is another object of the present invention to provide a method of screening individuals for a mutation that leads to Friedreich's ataxia, comprising the steps of sequencing DNA from an individual, and comparing said sequence from said individual to SEQ ID NOS 1-12 to determine what differences, if any, there are between the two sequences.
It is yet a further object of the present invention to provide a method of treating Friedreich's ataxia in an individual, comprising the step of SUBSTITUTE SHEET (RULE 26) administering an effective pharmacologic dose of a protein having an amino acid sequence substantially similar to SEQ ID NO 4 to said individual.
It is an additional object of the present invention to provide a method of treating Friedreich's ataxia in an individual, comprising administration of a nucleic acid vector containing an X25 gene capable of expression in a pharmacologically acceptable carrier to said individual.
It is a further object of the present invention to provide compositions of matter having SEQ ID NOS 1-32.
iu Other and further objects. features and advantages will be apparent and the invention more readily understood from a reading of the following specification and by reference to the accompanying drawings forming a part thereof, wherein the examples of the presently preferred embodiments of the invention are given for the purposes of disclosure.

DESCRIPTION OF THE DRAWINGS

Figure 1(A): Transcription map of the FRDA critical interval.
Distances are in kilobase pairs from the first Not I site upstream to the ZO-2 gene. The critical FRDA region is between the F8101 marker and the ZO-2 gene. M. MIu I site; N, Not I site; E, Eag I site; S. Sac II site; B, BssH I site. Figure 1(B): Alignment of the exon 5a-containing isoform of frataxin with translated ORFs contained within a C. elegans cosmid (CELT59G1) and a S. cerevisiae EST (T38910). Identical amino acids are boxed. The putative signal peptide is underlined. Amino acids involved by point mutation (L106X and 1154F) are indicated by vertical arrows. The exon 5b-containing isoform diverges at position 161, and its 11 COOH-terminal amino acids are RLTWLLWLFHP.
Figure 2: Northern blot analysis of X25 transcripts. A 32P-labeled 5'-;0 RACE product containing exons 1-5b was hybridized to a multiple tissue SUBSTITUTE SHEET (RULE 26) Northern blot (Clontech), containing 2 pg of poly-A + RNA in each lane.
The membrane was washed at 50 with 0.1 x SSC, 0.1% SDS, then exposed to x-ray film at -70 for 7 days. The lower panel shows a successive hybridization of the same blot with an actin probe (provided 5 by the blot manufacturer).
Figure 3: Southern blot analysis showing FRDA-associated expanded restriction fragments. Lanes I and 12, normal controls; lanes 2-7, individuals from a Saudi Arabian FRDA family; lanes 8-11, individuals from a Louisiana Acadian (Cajun) FRDA family. Affected 1o subjects are in lanes 3-5 and 9-10, heterozygous carriers in lanes 2, 6-8, and 11. The position of molecular weight markers is indicated on the side. The constant bands correspond to exons 2 and 3 (15 kb), and to a related sequence outside of the FRDA region (5 kb). Ten pg of genomic DNA from each individual were digested with Eco RI, run in a 0.6%
agarose gel, and blotted onto a nylon membrane (Hybond+). The blot was hybridized with a 32P-labeled X25 cDNA probe. After a highest stringency wash with 0.1 x SSC, 0.1% SDS for 5' at 65 , the blot was exposed to x-ray film at -70 for two days.
Figure 4: An automated sequence of the FRDA-associated expanded region from a cosmid subclone. The CTT strand was sequenced.
Figure 5: Automated sequence of the FRDA-associated expanded region containing the expanded repeat in a FA patient. -The CTT strand was sequenced. It is interesting to note the presence of two imperfect repeats in the patient (the 7th and 8th in the sequenced strand) that are not present on the normal sequence and which could indicate a polymorphic variant present on the chromosome in which the original expansion occurred.

* Trade-mark Figure 6(A): Example of PCR analysis of normal alleles of the GAA
repeat. Lane 1 is the 1 kb ladder DAN size marker, lanes 2-6 are normal controls previously identified to be heterozygous at the repeat. The GAA-F/GAA-R primers were used for amplification. Fragments vary in size in the 480-520 bp range.
Figure 6(B): PCR amplification of the expanded GAA repeat in a FRDA carrier (lane 8) and in a patient (lane 4). Lane 1 is the 1 kb ladder DNA marker, lane 2 is a normal control. The Bam/2500 primers were used for FOR. Expanded alleles have a slightly fuzzy appearance.
Instability of the repeat is indicated by the presence of two distinct bands in the patient lane, although the patient is an offspring of consanguineous parents. Also, the carrier in lane 3 is the patient's mother, but the corresponding expanded allele does not exactly match in size any of her offspring bands.
Figure 7: Segregation of the L106X mutation and of the GAA
expansion in a FRDA family. The SSCP pattern shown in A indicates the paternal origin of the point mutation, while Southern blot analysis, shown in B, indicates the maternal origin of the expansion. NR indicates an unrelated normal control.
Figure 8 : RT-PCR analysis of X25 mRNA in FRDA subjects, obligate carriers and normal controls. Reactions were performed on total RNA extracted from lymphoblastoid cell lines. The serine hydroxymethyltransferase (SHMT) transcript (encoded by a gene on chromosome 17) was used as a control for RNA amount. Mock reactions without reverse transcriptase (-RT) were also performed as a negative control. In the case of SHMT, the PCR following the -RT
reactions generated a product of larger size than the product expected from the cDNA because a fragment of genomic DNA (contaminating the RNA preparation) containing a small intron was amplified. In all three SUBSTITUTE SHEET (RULE 26) panels the lane marked with r.t. is a negative control (water), lane 9 corresponds to a normal control individual, lanes 1 and 4 to obligate carriers of FRDA, lanes 2, 3, and 5 to 8 to individuals with FRDA. To generate cDNA from the X25 transcript, the RT reaction was primed with the oligonucleotide E2R (SEQ ID NO 13), then PCR was performed between this primer and the nF primer (SEQ ID NO 14).

DETAILED DESCRIPTION OF THE INVENTION
It will be apparent to one skilled in the art that various substitutions to and modifications may be made to the invention disclosed herein without departing from the scope of the invention.
As used herein, "FRDA" refers to Friedreich ataxia. an autosomal recessive, degenerative disease that involves the central and peripheral nervous system as well as the heart.
As used herein, "GAA expansion" refers to multiple (GAA)õ repeats located 1.4 kb downstream from exon 1 in an intron of the X25 gene.
As used herein, the "X25" gene refers to the gene identified on chromosome 9g13 that is in the critical region of the FRDA-determinative locus .
As used herein the term "polymerise chain reaction" or "PCR" refers to the PCR procedure described in the patents to Mullis, et al., U.S.
Patent Nos. 4,683,195 and 4,683,202. The procedure basically involves: (1) treating extracted DNA to form single-stranded complementary strands: (2) adding a pair of oligonucleotide primers, wherein one primer of the pair is substantially complementary to part of the sequence in the sense strand and the other primer of each pair is substantially complementary to a different part of the same sequence in the complementary antisense strand; (3) annealing the paired primers to the complementary sequence; (4) simultaneously extending the annealed primers from a 3' terminus of each primer to synthesize an SUBSTITUTE SHEET (RULE 26) extension product complementary to the strands annealed to each primer wherein said extension products after separation from the complement serve as templates for the synthesis of an extension product for the other primer of each pair; (5) separating said extension products from said templates to produce single-stranded molecules: and (6) amplifying said single-stranded molecules by repeating at least once said annealing, extending and separating steps.
As used herein. the term "pulsed field gel electrophoresis" or "PFGE"
refers to a procedure described by Schwartz, et at., Cold Springs Harbor io Symposium, Quantitative Biology, 47:189-195 (1982). The procedure basically comprises running a standard electrophoresis gel (agarose, polyacrylamide or other gel known to those skilled in the art) under pulsing conditions. One skilled in the art recognizes that the strength of the field as well as the direction of the field is pulsed and rotated in order to separate megabase DNA molecules. Current commercial systems are computer controlled and select the strength, direction and time of pulse depending on the molecular weight of DNA to be separated.
As used herein, the phrase "gene transcript" shall mean the RNA
product that results from transcribing a genetic (DNA) template. "Gene"
shall mean a hereditary unit: in molecular terms. a sequence of chromosomal DNA that is required for production of a functional product.
As used herein, the phrase "messenger RNA" or "mRNA" shall mean an RNA transcribed from the DNA of a gene, that directs the sequence of amino acids of the encoded polypeptide.
As used herein. the phrase "copy DNA" or "cDNA" shall mean DNA
synthesized from a primer hybridized to a messenger RNA template.
As used herein, the phrase "oligonucleotide" shall mean a short nucleic acid molecule (usually 8 to 50 base paris), synthesized for use as a probe or primer.

SUBSTITUTE SHEET (RULE 26) As used herein, the phrase "primer'' shall mean a short DNA or RNA
molecule that is paired with a complementary DNA or RNA template, wherein the short DNA or RNA molecule provides a free 3'-0H terminus at which a DNA polymerase starts synthesis of a nucleotide chain.
It is a particular object of the present invention to provide a method of screening individuals for a mutation that leads to Friedreich's ataxia, comprising the steps of digesting DNA from an individual to be tested with a restriction endonuclease; and measuring the length of a restriction fragment length polymorphism (RFLP) by hybridization to probes that recognize a region encompassing a GAA repeat in a first intron of an X25 gene and performing Southern Blot analysis, wherein an RFLP
corresponding to a GAA repeat longer than a normal range of 7-22 triplets, usually more than about 120, is an indication of said mutation that leads to Friedreich's ataxia.
It is a further object of the present invention to provide a method of screening individuals for a mutation that leads to Friedreich's ataxia, comprising the steps of measuring expression of an X25 gene by determining an amount of mRNA expressed from the X25 gene and from known controls, and comparing the amount of mRNA from the X25 gene to the amount of mRNA from the known controls, wherein a reduced amount of mRNA from the X25 gene indicates individuals having said mutation that leads to Friedreich's ataxia.
It is an additional object of the present invention to provide a method of screening individuals for a mutation that leads to Friedreich's ataxia, wherein the amounts of mRNA is determined by the steps of extracting mRNA from individuals to be tested; preparing cDNA from said mRNA, amplifying said cDNA to produce amplification products; and comparing relative amounts of X25 and control cDNA present, wherein a reduced amount of mRNA from the X25 gene indicates individuals having said mutation that leads to Friedreich's ataxia.

SUBSTITUTE SHEET (RULE 26) (0 It is an additional object of the present invention to provide a method of screening individuals for a mutation that leads to Friedreich's ataxia.
by detecting the amount of specific proteins encoded by X25 in cells from patients using antibodies specific for the X25 proteins, It is another object of the present invention to provide a method of screening individuals for a mutation that leads to Friedreich's ataxia, comprising the step of detecting a variation in a size of a (GAA), repeat in a first intron of a X25 gene by measuring the length of said repeat, wherein n for normal individuals ranges from 1-22 and n for affected individuals is more than about 120-900.
It is an additional object of the present invention to provide a method for detecting a GAA poiymorphism in a first intron of an X25 gene comprising the steps of performing a PCR assay to produce amplified products of said first intron of said X25 gene and measuring the length of said amplified products with molecular techniques known in the art.
It is another object of the present invention to provide a method of screening individuals for a mutation that leads to Friedreich's ataxia, comprising the steps of sequencing DNA from an individual, and comparing said sequence from said individual to SEQ ID NOS 1-12 to determine what differences, if any, there are between said sequence from said individual and said SEQ ID NOS 1-12.
It is yet a further object of the present invention to provide a method of treating Friedreich's ataxia in an individual, comprising the step of administering a pharmacologic effective dose of a protein having an amino acid sequence substantially similar to SEQ ID NO 4 to said individual.
It is an additional object of the present invention to provide a method of treating Friedreich's ataxia in an individual, comprising administration of a nucleic acid vector containing an X25 gene capable of expression ;0 and a pharmacologically acceptable carrier to said individual.

SUBSTITUTE SHEET (RULE 26) I I
It is a further object of the present invention to provide -compositions of matter having SEQ ID NOS 1-32.
The therapeutic compositions of the present invention can be formulated according to known methods to prepare pharmacologically useful compositions. The compositions of the present invention or their functional derivatives are combined in admixture with a pharmacologically acceptable carrier vehicle. Suitable vehicles and their formulations are well known in the art. In order to form a pharmacologically acceptable composition suitable for effective therapeutic administration, such compositions will contain an effective amount of the X25 gene or its equivalent or the functional derivative thereof. or the frataxin protein or its equivalent or the functional derivative thereof, together with the suitable amount of carrier vehicle.
The nucleic acid therapeutic composition of the present invention will usually be formulated in a vector. The frataxin protein therapeutic composition will usually be administered as a purified protein in a pharmacologically suitable carrier.The compositions can be administered by a variety of methods including parenterally, by injection, rapid infusion, nasopharyngeal absorption, dermal absorption or orally.
The compositions may alternatively be administered intramuscularly or intravenously. In addition. the compositions for parenteral administration can further include sterile aqueous or nonaqueous solutions, suspensions and emulsions. Examples of known nonaqueous solvents include propylene glycol. polyethylene glycol, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Carriers, adjuncts or occlusive dressings can be used to increase tissue permeability and enhance absorption. Liquid dosage forms for oral administration may generally comprise a liposome solution. Suitable forms for suspension include emulsions, solutions, syrups and elixirs ?0 containing inert diluents commonly used in the art, such as purified SUBSTITUTE SHEET (RULE 26) water. Besides the inert dilutants. such compositions can also include wetting agents. emulsifying and suspending agents or sweetening, flavoring, coloring or perfuming agents.
Additionally, pharmaceutical methods may be employed to control the duration of action. These are well known in the art and include control release preparations and can include appropriate macromolecules, for example polymers, polyesters, polyamino acids, polyvinyl, pyrolidone, ethylenevinylactate, methylcellulose, carboxymethylcelIulose, or protamine sulfate. The concentration of macromolecules, as well as the to methods of incorporation. can be adjusted in order to control release.
Additionally. the vector could be incorporated into particles of polymeric materials such as polyesters, polyamino acids, hydrogells, poly (lactic acid) or ethylene vinylacetate copolymers. In addition to being incorporated, these agents can also be used to trap the vectors in microcapsules. These techniques are well known in the art.
A composition is said to be "pharmacologically acceptable" if its administration can be tolerated by a recipient patient. Such an agent is said to be administered in a "therapeutically effective amount" if the amount administered is physiologically significant. An agent is physiologically significant if its presence results in detectable change in the physiology of a recipient patient.
Generally, the dosage needed to provide an effective amount of composition will vary depending on such factors as the recipient's age, condition, sex and extent of disease, if any, and other variables which can be adjusted by one of ordinary skill in the art.
One skilled in the art will appreciate readily that the present invention is well adapted to carrying out the ends and advantages mentioned as well as those inherent herein. The probes, primers, methods, procedures and techniques described are presently representative of the _ 0 preferred embodiments. are intended to be exemplary, and are not SUBSTITUTE SHEET (RULE 26) intended as limitations on the scope of the invention. Changes therein and other uses will occur to those skilled in the art, and are encompassed within the spirit of the invention or defined by the scope of the appended claims.

The following examples are offered by way of illustration and are not intended to limit the invention in any manner:

Example 1: Localizing and Sequencing the FRDA Critical Region.
Potential exons were identified in the FRDA critical region by direct cDNA selection, exon amplification, and computer prediction from random sequences. Twelve cosmids spanning 120 kb of the critical FRDA interval, plus 80 kb immediately proximal to the interval were 1s subcloned individually as Bam HI - Bgl 11 fragments into pSPLI and pSPL3 exon-trapping vectors and transfected into COS-7 (A6) cells for splicing of potential exons. See D.M. Church et al., Nature Genet. 6:98 (1994). The same cosmids were used for hybridization-selection from uncloned cDNAs synthesized from human cerebellum and spinal cord poly-A+RNA. See J.G. Morgan et al., Nucl. Acids Res. 20:5173 (1992).
Finally, seven of the cosmids were subcloned as Sau 3AI, Apo 1, and Hae III fragments and about 1500 random single pass sequences were generated. These sequences were analyzed using the GRAILI a and GRAIL2 (E.C. Uberbacher and R.J. Mural, Proc. Natl. Acad. Sci. USA
88:11261 (1991), and FEXH (V.V. Solovyev et al., Nucl. Acids Res. 22, 5156 (1994)) programs.
These analyses yielded 19, 5, and 17 potential coding sequences, respectively, including two that matched known genes, namely the protein kinase A gamma catalytic subunit gene (obtained by cDNA
selection and random sequencing), and a mitochondrial adenylate kinase 3 pseudogene (obtained by random sequencing). One exon, called d26, was identified independently by two approaches. Nested primers based on the d26 sequence, when used in a rapid amplification of cDNA 5' end (5'-RACE) experiment on a heart cDNA template yielded two independent but overlapping products. The 5'-RACE was performed using the Clontech RACE-ready cDNA kit according to the manufacturer's instructions. Sequence from these clones matched another amplified exon and an expressed sequence tag (EST) from a human liver+spleen cDNA library (Homosapiens cDNA clone 126314, 5' 1o sequence (GenBank accession number R06470)). This gene, called X25, apparently had alternate transcripts, because the sequences at the 3' end of the EST and RACE products were different.
The gene structure of X25 (Figure IA) was resolved by obtaining intronic sequences flanking the identified exons, by inverse PCR, and by direct sequencing of cosmids. The EST clone contained 4 exons, and the longer RACE product contained one additional 5' exon. This exon mapped with the CpG island at position 100 on the genomic map. A
transcription start site was predicted 388 bp upstream of the exon I
donor splice site, and a TATA box was found 28 bp further upstream by the TSSG program. Five exons (1 to 5a, where exon 5a corresponds to the 3' end of the EST) were found to be spread over 40 kb. They contain an open reading frame (ORF) encoding a 210 amino-acid protein, which was named frataxin (Figure 1 B). An alternative exon (5b), corresponding to d26, was localized at about 40 kb from exon 5a in the telomeric direction. Exon 5b also has an in-frame stop codon, so that the alternative transcript encodes a shorter, 171 amino acid protein, whose 11 COOH- terminal residues differ from the main isoform.
Nucleotide sequences of the X25 exons have been deposited in the GenBank database under the accession numbers U43748 to U43753.
3o The 3' end of the transcript encoding the alternative form was * Trade-mark Is investigated by 3' RACE (see Froman and Martin. Technique 1:165 (1989)), 2 pg total RNA from Hela cells was used with nested primers in exon 5b3 and showed that, depending on the alternate usage of the 3' donor splice site in exon 5b, either a transcript ending with this exon, or a longer transcript including an additional non-coding exon 6 could be generated. This longer 3' RACE product ended with the poly-A tail of a downstream Alu sequence. Genomic sequence of exon 6 showed that it contains 3 Alu sequences in tandem, followed by a polyadenylation signal 1050 bp away form the acceptor splice site. Exon 6 was mapped 13 kb telomeric to exon 5b (Fig. 1A). Splice sites of all 7 exons (1 to 4, 5a, 5b. and 6) conform to the canonical consensus.

Example 2: Expression of the X25 Transcript.

13 Poly A+ Northern blots of different human tissues revealed the highest expression of X25 in heart, intermediate levels in liver, skeletal muscle, and pancreas, and minimal expression in other tissues, including whole brain (Figure 2). A 1.3 kb major transcript was identified, in agreement with the predicted size of an exon 5a-containing mRNA. Fainter bands of 1.05, 2Ø 2.8, and 7.3 kb were also detected.
Further hybridizations of the northern blot with exon 5a- and 5b-specific probes revealed that the 1.05 and 2.0 kb bands contained exon 5b, while sequences matching exon 5a were found in the 1.8 and 7.3 kb bands in addition to the major 1.3 kb band. A northern blot of total RNA from selected parts of the central nervous system (CNS) revealed high expression of the 1.3 kb transcript in the spinal cord, with less expression in cerebellum, and very little in cerebral cortex (not shown).
Overall, expression of X25 appeared to be highest in the primary sites of degeneration in FRDA, both within and outside the CNS.

SUBSTITUTE SHEET (RULE 26) !6 To investigate the nature of the larger transcripts, a fetal brain cDNA
library was screened with the EST clone (exons 2-5a). Among nine positives, four clones were isolated whose sequence extended beyond the limits of the previously identified X25 mRNAs. Sequence analysis of these clones indicated that they originated from a related gene, differing from X25 at several positions, and with stop codons in the sequence corresponding to X25 exon 1. Three of the cDNAs, which are identical in the portion that has been sequenced, extend respectively for 0.5, 1 and 2 kb upstream of exon 1. Their sequence presents numerous divergences from X25 in the part corresponding to exon 1, mostly CpG
dinucleotides changed in TG or CA, then being almost identical in the part corresponding to exons 2 to 4.
An additional 1.6 kb cDNA begins with a sequence closely matching exon 5a, even in its UTR, with only occasional single base changes and short insertions/deletions. The X25 related gene was excluded from the critical FRDA region, and at least one intronless copy exists in the genome, as indicated by Southern blot and PCR analysis. Southern blot analysis with an X25 exon 1-5a cDNA probe revealed a prominent 5 kb Eco RI band in genomic DNA that did not correspond to any exon and was absent in YAC and cosmid DNA from the critical FRDA region.
Several additional bands, also absent from cloned DNA from the FRDA
region, appeared when blots were washed at lower stringency (1 X SSC
at room temperature). The primers nF2 (5'-TCCCGCGGCCGGCAGAGTT-3') [SEQ ID NO 14] and E2R (5'-CCAAAGTTCCAGATTTCCTCA-3') [SEQ ID NO 13], which can amplify a 173 bp fragment spanning exons 1 and 2 of the X25 cDNA, generated a PCR product of corresponding size from genomic DNA, but not from cloned DNA from FRDA region, indicating the presence of sequences with high similarity to a processed X25 transcript elsewhere in the genome.

SUBSTITUTE SHEET (RULE 26) Example 3: Computer Database Search.

A BLASTN DNA database search with the X25 DNA sequence and a BLASTP search with the translated sequence did not reveal any significant match. However, a TBLASTN search in which the protein sequence was compared to the six frame translation of the DNA
databases yielded highly significant matches with an ORF contained in a C. elegans cosmid (P = 7.6 x 10'13) and with a S. cerevisiae EST (P=2.0 1o x 10'10) (Fig. 1 B). In both cases, the closest match involved a 27-aa segment of the protein (positions 141-167) encoded in exons 4 and 5a, showing 25/28 and 22/27 amin-oacid identity with the C. elegans and S.
cerevisiae sequences, respectively, and 65% identity at the DNA level.
Secondary structure predictions for the X25-encoded protein suggested an a-helical structure for the NH2-terminal 30 amino acids and the regions between residues 90-110 and 185-195, with possible interspersed (3-sheet regions around residues 125-145 and 175-180.
Secondary structure prediction was performed with the SSP and NNSSP
programs, which are designed to locate secondary structure elements (V.V. Solovyev and A.A. Salamov, CABIOS 10:661 (1994)). The TMpred program was used to predict putative transmembrane domains (K.
Hoffmann and W. Stoffel, Biol. Chem. Hoppe-Seyler 374:166 (1993)).
PSORT was used to predict possible protein sorting signals (K. Nakai and M. Kanehisa, Proteins: Structure, Function, and Genetics 11:95 (1991)). No transmembrane domain was identified. As computer analysis of the amino acid sequence suggests that the frataxin protein contains an N-terminal hydrophobic signal, it may be a precursor for a secreted protein with a growth factor or hormone-like action, making frataxin an ideal protein for expression in bacteria, yeast and mammalian cells.

* Trade-mark Example 4: Determining the Nature of the Mutation Leading to FRDA.

All six coding exons of X25 in 184 FRDA patients were amplified with flanking primers and screened for mutations. The following intronic primers were used to amplify the X25 exons: exon 1 (240 bp), F: 5'-AGCACCCAGCGCTGGAGG-3'[SEQ ID N015], R: 5'-CCGCGGCTGTTCCCGG-3' [SEQ ID NO 16]; exon 2 (168 bp), F: 5'-1u AGTAACGTACTTCTTAACTTTGGC-3' [SEQ ID NO 17]; R: 5'-AGAGGAAGATACCTATCACGTG'-3' [SEQ ID NO 18], exon 3 (227 bp), F: 5- AAAATGGAAGCATTTGGTAATCA-3' [SEQ ID NO 19], R: 5'-AGTGAACTAAAATTCTTAGAGGG-3' [SEQ ID NO 20]; exon 4 (250 bp), F: 5'-AAGCAATGATGACAAAGTGCTAAC-3' [SEQ ID NO 21]; R: 5'-TGGTCCACAATGTCACATTTCGG-3' [SEQ ID NO 221; exon 5a (223 bp), F: 5'- CTGAAGGGCTGTGCTGTGGA-3' [SEQ ID NO 23], R: 5'-TGTCCTTACAAACGGGGCT-3' [SEQ ID NO 24], exon 5b (224 bp), F:
5'-CCCATGCTCAAGACATACTCC-3' [SEQ ID NO 25], R: 5'-ACAGTAAGGAAAAAACAAACAGCC-3' [SEQ ID NO 26]. Amplifications for exons 2. 3, 4. 5a. and 5b consisted of 30 cycles using the following conditions: 1 min. at 94 , 2 min. at 55 , 1 min. at 72 . To amplify the highly GC-rich exon 1, the annealing temperature was raised to 68 and 10% DMSO was added to the reaction. The search for mutations was conducted using single-strand conformation polymorphism (SSCP) analysis (see M. Orita et al., Genomics 5:874 (1989)) in 168 FRDA
patients, and chemical clevage (see J. A. Saleeba et al., Hum. Mutat.
1:63 (1992)) in 16. Three point mutations that introduce changes in the X25 gene product were identified.

SUBSTITUTE SHEET (RULE 26) Point Mutations. The first change, in a French family with two affected siblings, consisted of a T-+ G transversion in exon 3 that changed a leucine codon (TTA) into a stop codon (TGA)(L106X). The second case, in a Spanish family with one affected member. was an A-*G transition that disrupted the acceptor splice site at the end of the third intron, changing the invariant AG into a GG. Finally, a change from isoleucine to phenylalanine (1154F) was found in exon 4 in five patients from three Southern Italian families. This conservative change of an hydrophobic amino acid affects an invariant position within the highly conserved domain shared between human, worm and yeast. In all three cases. affected individuals were heterozygous for the point mutation.
The 1154F mutation was also found in 1 out of 417 chromosomes from 210 control individuals from the same Southern Italian population, which is compatible with the possibility that this is a disease-causing mutation.
is (Assuming a FRDA carrier frequency in Italy of 1/120 individuals and a frequency of 1154F of 1/40 FRDA chromosomes in Southern Italians, one individual in 3.300 in that population is expected to be a carrier of 1154F. Finding such an individual in a random sample of 210 subjects can occur with >6% probability.) Intron 1 Expansion. Southern blot analysis did not reveal any difference between FRDA patients and normal controls, when DNAs digested with Msp I. Taq 1, or Bst XI were hybridized with an X25 cDNA
probe, thereby excluding major rearrangements. Hybridization of Eco RI-digested DNAs from FRDA patients, however, revealed that the fragment containing exon 1 was on average 2.5 kb larger than in normal controls, with no detectable normal band. FRDA carriers were heterozygous for an enlarged- and a normal-sized fragment. The size of the enlarged fragment was clearly variable, even among FRDA carriers who were related (Figure 3). The enlarged region was localized further SUBSTITUTE SHEET (RULE 26) to a 5.2 kb Eco RI/Not I fragment within the first intron of X25, which was subcloned from a cosmid and sequenced.
Oligonucleotide primers were designed to amplify this fragment using a long-range PCR technique, and its increased in size in FRDA patients 5 was confirmed. The Perkin-Elmer XL long-PCR reagent kit was used to set up the reactions, utilizing standard conditions as suggested by the manufacturer and primers 5200Eco (5'- GGGCTGGCAGATTCCTCCAG-3') [SEQ ID NO 27] and 5200Not (5'-GTAAGTATCCGCGCCGGGAAC-3') [SEQ ID NO 28]. Amplifications were performed in a Perkin-Elmer 10 9600 machine, and consisted of 20 cycles of the following steps: 94 for 20 sec., 68 for 8 min., followed by further 17 cycles in which the length of the 68 increased by 15 sec./cycle. The generated amplification product is 5 kb from normal chromosomes, and about 7.5 kb from FRDA
chromosomes.
15 Cosmid sequence analysis revealed a (GAA)9 repeat apparently derived from a poly-A expansion of the canonical A5TACA6 sequence linking the two halves of an Alu repeat (Figure 4 showing the reverse complementary sequence). The (GAA)9 repeat is located 1.4 kb downstream from exon 1, and restriction analysis of long-range PCR
20 fragments from FRDA patients located the abnormal size increase within 100 bp from this triplet repeat. Digestion of the same fragments with Mbo ii, whose recognition site is GAAGA, suppressed size difference between patients and controls, indicating that the GAA repeat may be involved. Direct sequencing proved that the mutation consists of an almost pure GAA repeat expansion (Figure 5). PCR primers were then designed to evaluate the presence and size of the GAA expanded repeat FRDA patients, and any variability of the repeat in normal individuals (Figure 6).
The primers GAA-F (5'-GGGATTGGTTGCCAGTGCTTAAAAGTTAG-3')[SEQ ID N029] and GAA-R (5-Trade-mark ATCTAAGGACCATCATGGCCACACTTGCC-3') [SEQ ID NO 30] flank the GAA repeat and generate a PCR product of 457 + 3n bp (n = number of GAA triplets). With these primers, efficient amplification of normal alleles could be obtained by using the traditional PCR procedure with Taq polymerise, after 30 cycles consisting of the following steps: 940 for 45 sec., 68 for 30 sec., 72 for 2 min. Enlarged alleles were much less efficiently amplified, particularly when present together with a normal allele; therefore, use of these primers is not indicated for FRDA
carrier detection. A more efficient amplification of expanded alleles, also in FRDA carriers, is obtained using the primers Bam (5'-GGAGGGATCCGTCTGGGCAAAGG-3') [SEQ IDNO 31] and 2500F (5'-CAATCCAGGACAGTCAGGGCTTT-3') [SEQ ID NO 32]. These primers generated a -1.5 kb (1398 bp) normal fragment. Amplification was conducted using the long PCR protocol, in 20 cycles composed of the following steps: 94 for 20 sec., 68 for 2 min. and 30 sec., followed by further 17 cycles in which the length of the 68 step was increased by 15 sec/cycle.
Seventy-nine unrelated FRDA patients with typical disease, including five patients known to carry X25 point mutations, were tested for the GAA expansion by Southern analysis and/or by PCR. The patients previously known to carry point mutations were all heterozygous for the expansion. Segregation analysis within families indicated that the point mutation and the GAA expansion had different parental origin (Figure 7), demonstrating that the point mutations--including the conservative missense mutation 1154F--are disease causing.
Homozygosity for expanded alleles was demonstrated in 71 of the 74 patients without previously-detected X25 point mutations, and heterozygosity was demonstrated in three.
Overall, according to these data the GAA expansion accounted for about 98% of the FRDA phenotype. The sizes of the enlarged alleles SUBSTITUTE SHEET (RULE 26) were found to vary between 200 and more than 900 GAA units, with most alleles containing 700-800 repeats. Instability of expanded repeats during parent-offspring transmission was clearly demonstrated, both directly by analysis of parent-offspring pairs. and indirectly by the detection of two distinct alleles in affected children of consanguineous parents, who are expected to be homozygous-by-descent at the FRDA
locus. PCR products corresponding to expanded repeats appeared as slightly blurred bands, suggesting the occurrence of only a limited degree of somatic mosaicism for different size repeats due to mitotic instability, at least in lymphocyte DNA (Figure 6B). Seventy-seven normal individuals who were tested by Southern analysis were homozygous for a normal allele. PCR analysis of additional 98 normal controls also did not show any expansion, and revealed that the GAA
repeat is polymorphic, its length varying from 7 to 22 units (Figure 6A).
Smaller alleles were more prevalent.
GAA repeats, up to 30-40 units, are common in many organisms and are sometimes polymorphic, as in the 3' UTR of the rat polymeric Ig receptor; however. they have not previously been associated with disease. A recently proposed theoretical model suggests that ability to form a hairpin structure is crucial for the susceptibility of trinucleotide repeats to give rise to large expansions (See A. M. Gray et al.. Cell 81:533 (1995)). According to this model, CAG/CTG or CGG/CCG
repeats are predicted to be expansion prone, while the GAA/CCT repeat has lowest propensity to expand, making the FRDA expansion an 2; unexpected finding. A striking linkage disequilibrium between FRDA and a polymorphism in a newly-identified exon of the Z0-2 gene (about 120 kb telomeric to the expanded triplet repeat) in French and Spanish families suggests a single origin for the FRDA expansion, but it is also compatible with a multistep or recurrent expansion on an allele at risk.
(See Imbert et al.. Nature Genet. 4:72 (1993) where the absolute linkage SUBSTITUTE SHEET (RULE 26) disequilibrium in myotonic dystrophy is expanded by recurrent mutations on such a risk allele.) The fact that RDA is autosomal recessive makes the natural history of the mutation at the population level strikingly different from any other known disease due to trinucleotide expansions.
In fragile X and myotonic dystrophy, where expansions of comparable size occur in non-coding sequences, carriers have severe early-onset disease and a strong reproductive disadvantage. Large expansions in these diseases are newly formed from unstable alleles of intermediate sizes. resulting in the phenomenon of anticipation. In FRDA, large expanded alleles are transmitted by asymptomatic carriers, and new expansion events in heterozygotes would go undetected at the phenotypic level. Absence of negative selection against heterozygotes plays the key role in maintaining the frequency of large FRDA expanded alleles as high as 1 per 250 chromosomes, at least one order of magnitude higher than any other characterized trinucleotide expansion.
Conversely, deletions of CTG repeats in myotonic dystrophy with reversion to normal size alleles have been observed (see Imbert et al., Nature Genet. 4:72 (1993)) wherein the absolute linkage disequilibrium in myotonic dystrophy is explained by recovered mutations in such a risk allele. In the sample of FRDA families in the study of the present invention. large expanded alleles were present in all tested symptomatic carriers. and, despite their size instability, neither new expansions deriving from an intermediate allele nor reversions to normality were detected. Although the occasional occurrence of such events cannot be excluded in the general population given the large number of heterozygous individuals, it appears that the frequency is low enough not to introduce detectable distortions in the pattern of FRDA
inheritance, particularly inconsistencies in linkage results.

SUBSTITUTE SHEET (RULE 26) Example 5: Quantification of the FRDA Transcript.

When the X25 transcript was amplified with primers connecting exons 1 and 2, FRDA patients showed either undetectable or extremely low mRNA levels when compared to carriers and unrelated controls.
RT-PCR. RT-PCR was done on lymphoblast RNA from two normal controls, two obligate carriers, and six patients, using the exon 2 reverse primer E2R (5'- CCAAAGTTCCAGATTTCCTGA-3') [SEQ ID NO 13] and the exon 1 forward primer 19F (5'-CAGGCCAGACCCTCAC-3') [SEQ ID
NO 14]. As a precaution to avoid amplification of X25-related sequences not deriving from the FRDA region transcript, the nF primer was chosen to have no match with the non-9q13 related gene. PCR
reactions were carried out for 25 cycles in order to maintain linearity is between starting and final concentrations of DNA fragments. PCR
products were blotted onto nylon membranes and hybridized with the 32P
end-labeled internal oligonucleotide nF2 ('5-TCCCGCGGCCGGCAGAGTT-3') [SEQ ID NO 15]. This observation suggests that either an abnormality in RNA processing, or an interference with the transcription machinery, occur as a consequence of the intronic GAA expansion.
Patients with deleterious point mutations affecting X25 clearly demonstrate that no other gene in the region, which could, in principle, be affected by a GAA expansion, is involved in the causation of FRDA.
The restricted expression of X25 in the sites of degeneration or malfunction distinguishes FRDA from the dominant ataxias and from ataxia telangiectasia, where expression of the causative gene is ubiquitous. A severely reduced X25 mature mRNA is expected to result in a similarly low level of frataxin. Reduced frataxin in spinal cord, heart SUBSTITUTE SHEET (RULE 26) and pancreas is likely the primary cause of neuronal degeneration, cardiomyopathy and increased risk of diabetes.
RNase Protection. In order to synthesize antisense riboprobes, two regions of the X25 cDNA were subcloned in a plasmid vector 5 containing the T7 RNA polymerase promoter. Two separate segments of then X25 cDNA, one containing exons 1 and 2 (partial) and the other containing exons 4 (partial) and 5b were subcloned accordingly. 1 lag of linearized plasmid was used as a template for in vitro transcription (using the Ambion Maxiscript kit) in a reaction containing 3 pM a- 32P UTP. The io reaction was carried out at 37 C for an hour, after which the DNA
template was completely digested by RNase-free DNase treatment.
Full-length labeled transcripts were then purified following proparative denaturing polyacrylamide gel electrophoresis. A human GAPDH
riborprobe (pTRI-GAPDH human, Ambion) was also generated as a 15 control.
The RNase protection assay was performed using the RPAII
Ribonuclease protection assay kit from Ambion following the manufacturer's recommendations. Briefly, 20 pg of total RNA extracted from patient and control lymphoblastoid cell lines was mixed with 8 x 104 20 cpm-labeled riboprobe in a 20 pl reaction, denatured and allowed to incubate at 45 C for 16 hours. 2 lag of RNA was used for the control GAPDH reaction. For each riboprobe, yeast RNA control hybridizations were performed as well. RNase (RNase A/RNase TI mixture) treatment was carried out for 30 minutes at 37 C. The reaction products were 25 ethanol precipitated and resuspended in formamide loading dye. These products were denatured and electrophoresed on a pre-heated 5%
polyacrylamide/8 M urea gel in Ix Tris-borate buffer at 35 watts constant power.The gel was dried and exposed to an X-ray film for 6 days at -=70 C using intensifying screens. The sizes of the protected fragments * Trade-mark were estimated accurately using a sequence ladder that had been co-electrophoresed with the sample.
Example 6: Therapeutics.

FRDA is caused by abnormalities in the X25 gene leading to a deficiency of its protein product, frataxin, occasionally due to point mutations that generate a truncated protein but, most commonly, to a GAA expansion in the first intron that causes supression of gene expression. Therapeutic administration of frataxin to FRDA patients is to therefore an aspect of the present invention. Large amounts of recombinant frataxin is produced by cloning X25 cDNA into an expression vector that is tranformed into a suitable organism.
Expression vectors that lead to production of high amounts of recombinant protein can be purified by several techniques, and prepared 15 for systemic or local administration to patients. Computer analysis of the frataxin sequence suggests that frataxin is not a membrane protein, and is likely secreted. Both characteristics make frataxin an ideal protein for administration.
Another approach is examining the function of the frataxin protein and identifying compounds that can induce a cellular response or modification in the cell metabolism in cells that produce and/or respond to frataxin. Such compounds overcome the consequences of the lack of frataxin protein in FRDA.
Additionally, one can inactivate the murine X25 homolog via homologous recombination to provide an animal model for Friedreich's ataxia in order to test various therapeutic strategies.
Finally, the coding sequence for frataxin is inserted into a suitable expression vector that is administered to FRDA patients. The coding sequence of frataxin is inserted in the genome of a modified RNA or DNA virus, which is administered systemically or locally to patients, or SUBSTITUTE SHEET (RULE 26) used to transduce cultured cells from patients that are then re-implanted into the patient body. Alternatively, non-viral vectors are utilized and administered directly to the patients or to patient's cultured cells that are re-implanted into the patient.

One skilled in the art will appreciate readily that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The nucleotides, proteins, peptides, methods, procedures and techniques l0 described herein are presently representative of the preferred embodiments, are intended to be exemplary and are not intended as limitations on the scope. Changes therein and other uses will occur to those skilled in the art which are encompassed within the spirit of the invention or defined by the scope of attached claims.

SUBSTITUTE SHEET (RULE 26) SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT:
(A) NAME: INSTITUT NATIONAL DE LA SANTE ET DE LA
RECHERCHE MEDICALE(INSERM) (B) STREET: 101 RUE DE TOLBIAC
(C) CITY: PARIS
(E) COUNTRY: FRANCE
(F) POSTAL CODE (ZIP): 75654 (A) NAME: BAYLOR COLLEGE OF MEDICINE
(B) STREET: ONE BAYLOR PLAZA
(C) CITY: HOUSTON
(D) STATE: TEXAS
(E) COUNTRY: USA
(F) POSTAL CODE (ZIP): 77030 (ii) TITLE OF INVENTION: DIRECT MOLECULAR DIAGNOSIS OF FRIEDREICH
ATAXIA

(iii) NUMBER OF SEQUENCES: 33 (iv) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk (B) COMPUTER: IBM PC compatible (C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: Patentln Release #1.0, Version #1.30 (EPO) (v) CURRENT APPLICATION DATA:
APPLICATION NUMBER: 2,248,016 FILING DATE: MARCH 4, 1997 (2) INFORMATION FOR SEQ ID NO: 1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 8353 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1:

CCCGTCTCTA CTAACAAATA AA?AAAAATA CAAAAAATTA GCCAGGCATG GTGGCGGGCG 2220 (2) INFORMATION FOR SEQ ID NO: 2:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1490 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2:

(2) INFORMATION FOR SEQ ID NO: 3:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 642 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3:

(2) INFORMATION FOR SEQ ID NO: 4:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 210 amino acids (B) TYPE: amino acid (C) STRANDEDNESS:
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4:
Met Trp Thr Leu Gly Arg Arg Ala Val Ala Gly Leu Leu Ala Ser Pro Ser Pro Ala Gln Ala Gln Thr Leu Thr Arg Val Pro Arg Pro Ala Glu Leu Ala Pro Leu Cys Gly Arg Arg Gly Leu Arg Thr Asp Ile Asp Ala Thr Cys Thr Pro Arg Arg Ala Ser Ser Asn Gln Arg Gly Leu Asn Gln Ile Trp Asn Val Lys Lys Gln Ser Val Tyr Leu Met Asn Leu Arg Lys Ser Gly Thr Leu Gly His Pro Gly Ser Leu Asp Glu Thr Thr Tyr Glu Arg Leu Ala Glu Glu Thr Leu Asp Ser Leu Ala Glu Phe Phe Glu Asp Leu Ala Asp Lys Pro Tyr Thr Phe Glu Asp Tyr Asp Val Ser Phe Gly Ser Gly Val Leu Thr Val Lys Leu Gly Gly Asp Leu Gly Thr Tyr Val Ile Asn Lys Gln Thr Pro Asn Lys Gln Ile Trp Leu Ser Ser Pro Ser Ser Gly Pro Lys Arg Tyr Asp Trp Thr Gly Lys Asn Trp Val Phe Ser His Asp Gly Val Ser Leu His Glu Leu Leu Ala Ala Glu Leu Thr Lys Ala Leu Lys Thr Lys Leu Asp Leu Ser Trp Leu Ala Tyr Ser Gly Lys Asp Ala (2) INFORMATION FOR SEQ ID NO: 5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 171 amino acids (B) TYPE: amino acid (C) STRANDEDNESS:
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5:

Met Trp Thr Leu Gly Arg Arg Ala Val Ala Gly Leu Leu Ala Ser Pro Ser Pro Ala Gln Ala Gln Thr Leu Thr Arg Val Pro Arg Pro Ala Glu Leu Ala Pro Leu Cys Gly Arg Arg Gly Leu Arg Thr Asp Ile Asp Ala Thr Cys Thr Pro Arg Arg Ala Ser Ser Asn Gln Arg Gly Leu Asn Gln Ile Trp Asn Val Lys Lys Gln Ser Val Tyr Leu Met Asn Leu Arg Lys 35 Ser Gly Thr Leu Gly His Pro Gly Ser Leu Asp Glu Thr Thr Tyr Glu Arg Leu Ala Glu Glu Thr Leu Asp Ser Leu Ala Glu Phe Phe Glu Asp Leu Ala Asp Lys Pro Tyr Thr Phe Glu Asp Tyr Asp Val Ser Phe Gly Ser Gly Val Leu Thr Val Lys Leu Gly Gly Asp Leu Gly Thr Tyr Val Ile Asn Lys Gin Thr Pro Asn Lys Gln Ile Trp Leu Ser Ser Pro Ser Arg Leu Thr Trp Leu Leu Trp Leu Phe His Pro (2) INFORMATION FOR SEQ ID NO: 6:

(i) SEQUENCE CHARACTERISTICS:

(A) LENGTH: 2458 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6:

TCCGGAGTTC AAGACTAACC TGGCCAACAT GGTGAAACCC AGTATCTACT AAAA.AATACA 2160 (2) INFORMATION FOR SEQ ID NO: 7:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1177 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 7:

(2) INFORMATION FOR SEQ ID NO: 8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 227 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8:

(2) INFORMATION FOR SEQ ID NO: 9:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 241 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9:

(2) INFORMATION FOR SEQ ID NO: 10:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 538 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10:

(2) INFORMATION FOR SEQ ID NO: 11:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 429 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11:

(2) INFORMATION FOR SEQ ID NO: 12:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1501 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 12:

(2) INFORMATION FOR SEQ ID NO: 13:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 13:

(2) INFORMATION FOR SEQ ID NO: 14:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"

(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 14:

(2) INFORMATION FOR SEQ ID NO: 15:

(i) SEQUENCE CHARACTERISTICS:
15 (A) LENGTH: 18 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear 20 (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

25 (iv) ANTI-SENSE: NO

30 (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15:

(2) INFORMATION FOR SEQ ID NO: 16:
=`w (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 16 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 16:

(2) INFORMATION FOR SEQ ID NO: 17:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 17:

(2) INFORMATION FOR SEQ ID NO: 18:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 22 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 18:

(2) INFORMATION FOR SEQ ID NO: 19:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"

(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 19:

(2) INFORMATION FOR SEQ ID NO: 20:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 20:

(2) INFORMATION FOR SEQ ID NO: 21:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 21:

(2) INFORMATION FOR SEQ ID NO: 22:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 22:

(2) INFORMATION FOR SEQ ID NO: 23:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 23:

(2) INFORMATION FOR SEQ ID NO: 24:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"

(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 24:

(2) INFORMATION FOR SEQ ID NO: 25:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 25:

(2) INFORMATION FOR SEQ ID NO: 26:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 26:

(2) INFORMATION FOR SEQ ID NO: 27:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 base pairs 5 (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid 10 (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

.~4 (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 27:

(2) INFORMATION FOR SEQ ID NO: 28:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 28:

(2) INFORMATION FOR SEQ ID NO: 29:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 29 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"

(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 29:

(2) INFORMATION FOR SEQ ID NO: 30:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 30:

(2) INFORMATION FOR SEQ ID NO: 31:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 31:

(2) INFORMATION FOR SEQ ID NO: 32:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 32:

(2) INFORMATION FOR SEQ ID NO: 33:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 19 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (A) DESCRIPTION: /desc = "oligonucleotide"
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 33:

Claims (54)

Claims
1. A method of screening individuals for a mutation that leads to Friedreich's ataxia, comprising the step of detecting a variation in a size of a(GAA)n, repeat located between nucleotides corresponding respectively to nucleotide at position 4451 and nucleotide at position 4501 of sequence SEQ ID N°1 by measuring a length of said repeat, wherein n for affected individuals Is more than about 120.
2. The method of claim 1, wherein said size of said repeat is measured by restriction endonuclease digestion of sample DNA and Southern Blot analysis.
3. The method according to any one of claims 1 and 2, wherein the length of said repeat is measured by hybridisation to probes that recognize a, region encompassing said (GAA) repeat and performing Southern Blot analysis.
4. The method of claim 2, wherein the restriction endonuclease is EcoRl.
S. The method of claim 3, wherein the probe used for performing said Southern Blot is SEQ ID N°2.
6. The method of claim 3, wherein the probe used for performing said Southern Blot is an amplification product obtained by performing PCR on said DNA with SEQ ID N°18 and SEQ
ID N°15.
7. The method of claim 1, wherein said size of said repeat Is determined by pulsed field gel electmphoresis.
8. The method of claim 1, wherein the oligonucleotide primers of SEQ ID
NO° 29 and SEQ ID NO° 30 are used in said detecting step.
9. The method of claim 1, wherein the oligonucleotide primers of SEQ ID
NO° 31 and SEQ ID NO° 32 are used in said detecting step.
10. A method of screening individuals for a mutation that leads to Friedreich's ataxia, by determining an amount of expressed mRNA corresponding to the cDNA defined by sequence SEQ ID N°2 and from known controls, and comparing the amount of mRNA corresponding to the cDNA defined by sequence SEQ
ID-N°2 to the amount of mRNA from the known controls wherein a reduced amount of mRNA corresponding to the cDNA defined by se-quence SEQ ID N°2 Indicates individuals having said mutation that-leads to Friedreich's ataxia.
11. The method of claim 10, wherein the mRNA is determined by the steps of :
extracting mRNA from individuals to be tested;
preparing cDNA from mRNA, amplifying said cDNA to produce amplification products; and comparing relative amounts of the cDNA defined by sequence SEQ ID N°2 and control amplification products present, wherein a reduced amount of mRNA corresponding to the cDNA defined by sequence SEQ ID NO°2 Indicates Individuals having said mutation that leads to Friedreich's ataxia.
12. The method of claim 10, wherein the comparing step includes electrophoresis of said amplification products; trans-ferring said amplification products to a solid support; hybridizing said amplification products to a probe; and quan-tifying amplification products corresponding to the cDNA defined by sequence SEQ ID N°2 versus control gene amplification products.
13. The method of claim 12, wherein said probe is SEQ ID N°14.
14. The method of claim 12, wherein said control gene is serine hydroxymethyltransfenase (SHMT).
15. A method for detecting a GAA polymorphism located between nucleotides corresponding respectively to nucleotide at position 4451 and nucleotide at position 4501 of SEQ ID No1 comprising the steps of performing a PCR assay to produce amplified products of said GAA polymorphism and measuring the length of said amplified products, wherein the measuring of a number of GAA repeats of more than about 120 is indicative of a GAA polymorphism that leads to Friederich's Ataxia.
16. The method of claim 15, wherein SEQ ID No29 and SEQ ID No30 are used in said PCR
assay.
17. The method of claim 15, wherein SEQ ID No31 and SEQ ID No32 are used in said PCR
assay.
18. A composition of matter, comprising the molecule consisting of SEQ ID No1.
19. A composition of matter, comprising the molecule consisting of SEQ ID No2.
20. A composition of matter, comprising the molecule consisting of SEQ ID No3.
21. A composition of matter, comprising the molecule consisting of SEQ ID No4.
22. A composition of matter, comprising the molecule consisting of SEQ ID No5.
23. A composition of matter, comprising the molecule consisting of SEQ ID No6.
24. A composition of matter, comprising the molecule consisting of SEQ ID No7.
25. A composition of matter, comprising the molecule consisting of SEQ ID No8.
26. A composition of matter, comprising the molecule consisting of SEQ ID No9.
27. A composition of matter, comprising the molecule consisting of SEQ ID
No10.
28. A composition of matter, comprising the molecule consisting of SEQ ID No11
29. A composition of matter, comprising the molecule consisting of SEQ ID
No12.
30. A composition of matter, comprising the molecule consisting of SEQ ID
No13.
31. A composition of matter, comprising the molecule consisting of SEQ ID
No14.
32. A composition of matter, comprising the molecule consisting of SEQ ID
No15.
33. A composition of matter, comprising the molecule consisting of SEQ ID
No16.
34. A composition of matter, comprising the molecule consisting of SEQ ID
No17.
35. A composition of matter, comprising the molecule consisting of SEQ ID
No18.
36. A composition of matter, comprising the molecule consisting of SEQ ID
No19.
37. A composition of matter, comprising the molecule consisting of SEQ ID
No20.
38. A composition of matter, comprising the molecule consisting of SEQ ID
No21.
39. A composition of matter, comprising the molecule consisting of SEQ ID
No22.
40. A composition of matter, comprising the molecule consisting of SEQ ID
No23.
41. A composition of matter, comprising the molecule consisting of SEQ ID
No24.
42. A composition of matter, comprising the molecule consisting of SEQ ID
No25.
43. A composition of matter, comprising the molecule consisting of SEQ ID
No26.
44. A composition of matter, comprising the molecule consisting of SEQ ID
No27.
45. A composition of matter, comprising the molecule consisting of SEQ ID
No28.
46. A composition of matter, comprising the molecule consisting of SEQ ID
No29.
47. A composition of matter, comprising the molecule consisting of SEQ ID
No30.
48. A composition of matter, comprising the molecule consisting of SEQ ID
No31.
49. A composition of matter, comprising the molecule consisting of SEQ ID
No32.
50. A therapeutic composition for treating Friedreich's Ataxia comprising a pharmacologic dose of a protein having the amino acid sequence of SEQ ID No 4 and a pharmacologically acceptable carrier.
51. A therapeutic composition for treating Friedreich's Ataxia comprising a nucleic acid vector containing sequence SEQ ID No2 and capable of expression of this sequence and a pharmacologically acceptable carrier.
52. The use of a protein having the amino acid sequence of SEQ ID No 4 for treating Friedreich's Ataxia.
53. The use of a nucleic acid vector containing sequence SEQ ID No 2 and capable of expression of this sequence for manufacturing a therapeutic composition for treating Friedreich's ataxia.
54. A method of screening individuals for a mutation that leads to Friedreich's Ataxia, comprising the steps of:

(a) sequencing DNA from an individual and comparing said sequence from said individual to SEQ ID No1;

(b) determining what differences there are between said sequences from said individual to SEQ ID No 1 and (c) determining the presence of a mutation that leads to Friedreich's Ataxia if differences are determined at step b) that are selected from the group consisting of:

- a T .fwdarw. G transversion in exon 3 causing a L106X amino acid change;

- a A-G transition at the end of intron 3 causing a change of the invariant AG
into a GG;

- a nucleic acid change in exon 4 causing a 1154F amino acid change; and - an intron 1 expansion containing a number of GAA repeats of more than about 120.
CA2248016A 1996-03-06 1997-03-04 Direct molecular diagnosis of friedreich ataxia Expired - Lifetime CA2248016C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/611,587 1996-03-06
US08/611,587 US6150091A (en) 1996-03-06 1996-03-06 Direct molecular diagnosis of Friedreich ataxia
PCT/EP1997/001070 WO1997032996A1 (en) 1996-03-06 1997-03-04 Direct molecular diagnosis of friedreich ataxia

Publications (2)

Publication Number Publication Date
CA2248016A1 CA2248016A1 (en) 1997-09-12
CA2248016C true CA2248016C (en) 2011-05-10

Family

ID=24449614

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2248016A Expired - Lifetime CA2248016C (en) 1996-03-06 1997-03-04 Direct molecular diagnosis of friedreich ataxia

Country Status (8)

Country Link
US (1) US6150091A (en)
EP (1) EP0885309B1 (en)
JP (1) JP4602481B2 (en)
AT (1) ATE265544T1 (en)
AU (1) AU2095097A (en)
CA (1) CA2248016C (en)
DE (1) DE69728864T2 (en)
WO (1) WO1997032996A1 (en)

Families Citing this family (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010154858A (en) * 1996-03-06 2010-07-15 Inst National De La Sante & De La Recherche Medicale Direct molecular diagnosis of friedreich ataxia
US6623938B2 (en) 1997-01-21 2003-09-23 Human Genome Sciences, Inc. I-flice, a novel inhibitor of tumor necrosis factor receptor-1 and CD-95 induced apoptosis
US6787523B1 (en) 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6761888B1 (en) * 2000-05-26 2004-07-13 Neuralab Limited Passive immunization treatment of Alzheimer's disease
US20080050367A1 (en) 1998-04-07 2008-02-28 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
US6710226B1 (en) 1997-12-02 2004-03-23 Neuralab Limited Transgenic mouse assay to determine the effect of Aβ antibodies and Aβ Fragments on alzheimer's disease characteristics
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US7179892B2 (en) 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US6905686B1 (en) 1997-12-02 2005-06-14 Neuralab Limited Active immunization for treatment of alzheimer's disease
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US6923964B1 (en) 1997-12-02 2005-08-02 Neuralab Limited Active immunization of AScr for prion disorders
US6913745B1 (en) 1997-12-02 2005-07-05 Neuralab Limited Passive immunization of Alzheimer's disease
US6750324B1 (en) 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
DE19820201A1 (en) * 1998-05-06 1999-11-11 Wilhelm Krone Diagnostic test for cardiovascular risk factors and their complications
US20030147882A1 (en) * 1998-05-21 2003-08-07 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US6787637B1 (en) 1999-05-28 2004-09-07 Neuralab Limited N-Terminal amyloid-β antibodies
UA81216C2 (en) * 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
AU1222501A (en) * 1999-10-20 2001-04-30 Hopital Sainte-Justine Identification of arsacs mutations and methods of use therefor
US6812333B1 (en) 1999-10-20 2004-11-02 Hopital Sainte-Justine Identification of arsacs mutations and methods of use therefor
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
PE20020574A1 (en) 2000-12-06 2002-07-02 Wyeth Corp HUMANIZED ANTIBODIES THAT RECOGNIZE THE AMYLOID PEPTIDE BETA
MY139983A (en) 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
TWI374893B (en) 2003-05-30 2012-10-21 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
TW200635607A (en) 2004-12-15 2006-10-16 Elan Pharm Inc Humanized Aβ antibodies for use in improving cognition
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
WO2008085898A2 (en) * 2007-01-04 2008-07-17 President And Fellows Of Harvard College Methods for identifying essential proteins and therapeutic agents
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
DK2182983T3 (en) 2007-07-27 2014-07-14 Janssen Alzheimer Immunotherap TREATMENT OF AMYLOIDOGENIC DISEASES WITH HUMANIZED ANTI-ABETA ANTIBODIES
JO3076B1 (en) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap Immunotherapy regimes dependent on apoe status
FI20085450A0 (en) * 2008-05-14 2008-05-14 Arto Orpana Method for synthesizing a DNA strand
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
US9476043B2 (en) 2011-04-08 2016-10-25 Rula Zain-Luqman Diagnosis and treatment of friedreich's ataxia
NZ752941A (en) 2016-11-09 2023-04-28 Intrexon Corp Frataxin expression constructs
US20200040390A1 (en) * 2018-04-14 2020-02-06 Centrillion Technologies, Inc. Methods for Sequencing Repetitive Genomic Regions

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997005234A2 (en) * 1995-07-26 1997-02-13 Imperial College Of Science, Technology & Medicine Gene for friedreich's ataxia

Also Published As

Publication number Publication date
US6150091A (en) 2000-11-21
DE69728864T2 (en) 2005-07-28
ATE265544T1 (en) 2004-05-15
AU2095097A (en) 1997-09-22
EP0885309B1 (en) 2004-04-28
WO1997032996A1 (en) 1997-09-12
DE69728864D1 (en) 2004-06-03
JP4602481B2 (en) 2010-12-22
EP0885309A1 (en) 1998-12-23
JP2000507093A (en) 2000-06-13
CA2248016A1 (en) 1997-09-12

Similar Documents

Publication Publication Date Title
CA2248016C (en) Direct molecular diagnosis of friedreich ataxia
Campuzano et al. Friedreich's ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion
Ozelius et al. Human monoamine oxidase gene (MAOA): chromosome position (Xp21-p11) and DNA polymorphism
US8962269B2 (en) Spinal muscular atrophy diagnostic methods
Levy et al. Mutation in gelsolin gene in Finnish hereditary amyloidosis.
Campbell et al. Genomic variation and gene conversion in spinal muscular atrophy: implications for disease process and clinical phenotype
Satokata et al. Three nonsense mutations responsible for group A xeroderma pigmentosum
US5227292A (en) Neurofibromatosis type 1 gene
Ellis et al. HLA class II haplotype and sequence analysis support a role for DQ in narcolepsy
Duclos et al. The Friedreich ataxia region: characterization of two novel genes and reduction of the critical region to 300 kb
Satokata et al. Molecular basis of group A xeroderma pigmentosum: a missense mutation and two deletions located in a zinc finger consensus sequence of the XPAC gene
US6207811B1 (en) Nephrin gene and protein
Patrono et al. Missense and splice site mutations in SPG4 suggest loss-of-function in dominant spastic paraplegia
Humphries et al. Three keratin gene mutations account for the majority of dominant simplex epidermolysis bullosa cases within the population of Ireland
EP0711833B1 (en) Survival motor neuron (SMN) gene: a gene for spinal muscular atrophy
US5962230A (en) Diagnosis and treatment of glaucoma
US5534438A (en) Process for isolating genes and the gene causative of Huntington's disease and differential 3' polyadenylation in the gene
JPH11509730A (en) Early-onset Alzheimer's disease gene and gene product
CA2158791C (en) Dna sequences encoding the machado-joseph disease gene and uses thereof
JP2010154858A (en) Direct molecular diagnosis of friedreich ataxia
US5830661A (en) Diagnosis and treatment of glaucoma
GAO et al. Screening of the PDE6B gene in patients with autosomal dominant retinitis pigmentosa
EP1038015B1 (en) Mood disorder gene
US5811244A (en) In vitro method for identifying a clinical disorder associated with Nhe1 mutation
Aldrich et al. HLA class II haplotype and sequence analysis support a role for DQ in narcolepsy

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20170306