WO2007139811A1 - Methods and kits for linking polymorphic sequences to expanded repeat mutations - Google Patents

Methods and kits for linking polymorphic sequences to expanded repeat mutations Download PDF

Info

Publication number
WO2007139811A1
WO2007139811A1 PCT/US2007/012259 US2007012259W WO2007139811A1 WO 2007139811 A1 WO2007139811 A1 WO 2007139811A1 US 2007012259 W US2007012259 W US 2007012259W WO 2007139811 A1 WO2007139811 A1 WO 2007139811A1
Authority
WO
WIPO (PCT)
Prior art keywords
allele
specific
seq
single nucleotide
nucleotide polymorphism
Prior art date
Application number
PCT/US2007/012259
Other languages
French (fr)
Inventor
Paul Van Bilsen
William F. Kaemmerer
Eric Burright
Original Assignee
Medtronic, Inc.
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 Medtronic, Inc. filed Critical Medtronic, Inc.
Priority to EP07777234A priority Critical patent/EP2029773A4/en
Publication of WO2007139811A1 publication Critical patent/WO2007139811A1/en
Priority to US12/560,178 priority patent/US9133517B2/en

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
    • 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
    • C12Q1/6813Hybridisation assays
    • C12Q1/6827Hybridisation assays for detection of mutation or polymorphism
    • 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
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • 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 generally to compositions and methods for diagnosing diseases which have an allele- spe ⁇ ific therapy and a disease-causing mutation that is sufficiently distant from the molecular site of the therapy to require a diagnostic linking method.
  • CAG repeats CAG trinucleotide repeats
  • polyQ polyglutamine
  • Spinocerebellar ataxia type 1 Spinocerebellar ataxia type 2, Spinocerebellar ataxia type 3, Spinocerebellar ataxia, type 6, Spinocerebellar ataxia type 7, Spinocerebellar ataxia type 8, Spinocerebellar ataxia type 17, Huntington disease-like 2, Spinal and bulbar muscular atrophy, Huntington disease, Dentatorubral - pallidoluysian atrophy, Oculopharyngeal dystrophy.
  • HD Huntington's disease
  • the HD gene (IT15 gene) , which encodes huntingtin, a 350 kDa protein of unknown function, is located on the human chromosome 4 and consists of 67 exons .
  • the disease-causing mutation is a CAG repeat expansion located within exon 1 of the HD gene (HD exonl) .
  • the CAG repeat is translated into a polyQ stretch. The disease manifests itself when the polyQ stretch exceeds the critical length of 37 glutamines
  • huntingtin fragments with polyQ tracts in the pathological range (more than 37 glutamines) , but not in the normal range (20-32 glutamines) , form high molecular weight protein aggregates with a fibrillar morphology in vitro and in cell culture model systems (Scherzinger et al . (1999) Proc. Natl Acad. Sci. USA, 96:4604-4509; and Waelter et al . , (2001) MoI. Biol. Cell, 12:1393-1407).
  • 2003100101 describes isolation of one sequence in a mixture by hybridization markers and single-strand specific nucleases for use in single-molecule analysis.
  • U.S. Patent Application Publication No. 20030039964 describes a method for isolation of one sequence in a mixture by hybridization to a fixed probe, but does not disclose the use of reverse transcription.
  • U.S. Patent No. 6,013,431 describes a method for analysis of bases adjacent to a hybridized, immobilized oligo, but does not disclose enrichment of one allele over the other.
  • WO Patent Application No. 9820166 describes a method for specific selection of one allele over the other, followed by mass spectroscopic analysis of the selected molecule, but does not ⁇ disclose the use of reverse transcription. None of these references disclose methods and diagnostic kits for linking polymorphic sequences to expanded repeat mutations for improved allele-specific diagnosis .
  • U.S. Patent Publication No. 20040241854 discloses allele-specific inhibition of specific single nucleotide polymorphism variants, and presents data showing that "expanded CAG repeats and adjacent sequences, while accessible to RNAi, may not be preferential targets for silencing" thus describing the problem that our invention addresses (determining what SNP variant at a remote molecular position is linked to the expanded CAG repeat in a particular patient) , but does not teach the use of reverse transcription using an allele-specific primer to solve this problem, nor otherwise disclose a method for how to solve this problem.
  • 20060270623 discloses multiple siRNA sequences, including those comprising SNP variants, but does not provide any working examples regarding allele-specific RNA interference using these disclosed siRNA sequences, nor disclose how to determine which allele-specific siRNA to administer to a particular Huntington' s disease human patient in order to effectively treat that patient's disease by suppression of only the expanded Huntington allele in that patient.
  • the invention provides a method for determining which single nucleotide polymorphism variant of an allele from a gene isolated from a heterozygous patient is on the same mRNA transcript as a disease-causing mutation at a remote region of the gene's mRNA comprising: a) an allele-specific reverse transcription reaction using an allele-specific primer which recognizes one single nucleotide polymorphism variant, and b) analysis of an allele-specific cDNA product from the allele-specific reverse transcription reaction at the remote region of the gene to determine the presence or absence of the mutation on the allele-specific cDNA product, wherein the allele- specific primer is shorter than about 20 nucleotides.
  • the 3' end of the allele-specific primer is positioned at the single nucleotide polymorphism nucleotide position.
  • the invention provides a method of treating a patient comprising determining which SNP variant is on the same mRNA transcript as a disease-causing mutation according to the method recited above, and applying an allele-specific therapy to the SNP variant, wherein the allele-specific therapy comprises an siRNA comprising a double-stranded portion, wherein the single nucleotide polymorphism site is located within seven nucleotides from an end of the double stranded portion.
  • the allele-specific therapy of the present invention includes by way of example allele-specific RNA interference using siRNA or shRNA.
  • the invention provides a kit for determining which single nucleotide polymorphism variant of an allele of a heterozygous patient is on the same mRNA transcript as a disease-causing mutation located at a remote region of the gene' s mRNA comprising a) an allele-specific primer which recognizes one single nucleotide polymorphism variant, and b) a set of instructions, wherein the allele- specific primer is shorter than about 20 nucleotides.
  • the 3' end of the allele-specific primer is positioned at the single nucleotide polymorphism nucleotide position.
  • FIG. 1 is a schematic illustration of allele-specific reverse transcription for the "A" allele.
  • FIG. 2 is a photograph of a gel showing results of allele-specific reverse transcription.
  • FIG. 3 is an illustration of primers and molecular beacons .
  • FIG. 4 is an illustration of allele specificity of the molecular beacons used for the allele-specific quantification of real-time PCR product
  • FIG. 5 illustrates that a siRNA molecule designated as 363125_C-16 inhibits the mutant, but not the wild-type huntingtin mRNA.
  • the present invention relates to methods and kits for performing allele-specific reverse transcription from an SNP site and analysis of a cDNA at a region of gene mutation.
  • the methods, systems and reagents of the present invention are applicable to any disease which contains an SNP variant of an allele in a heterozygous subject that is on the same mRNA transcript as a disease-causing mutation that is at a remote region of the gene's mRNA.
  • the term "gene” refers to a DNA sequence that comprises control and coding sequences necessary for the production of a polypeptide or its precursor.
  • the polypeptide can be encoded by a full length coding sequence (either genomic DNA or cDNA) or by any portion of the coding sequence so long as the desired activity is retained.
  • the term “gene” also refers to an mRNA sequence or a portion thereof that directly codes for a polypeptide or its precursor.
  • transfection refers to the uptake of foreign DNA by a cell.
  • a cell has been "transfected” when exogenous (i.e., foreign) DNA has been introduced inside the cell membrane.
  • Transfection can be either transient (i.e., the introduced DNA remains extra ⁇ hromosomal and is diluted out during cell division) or stable (i.e., the introduced DNA integrates into the cell genome or is maintained as a stable episomal element) .
  • Cotransfection refers to the simultaneous or sequential transfection of two or more vectors into a given cell.
  • promoter element refers to a DNA regulatory region capable of being bound by an RNA polymerase in a cell (e.g., directly or through other promoter-bound proteins or substances) and initiating transcription of a coding sequence .
  • a promoter sequence is, in general, bounded at its 3 1 terminus by the transcription initiation site and extends upstream (5 1 direction) to include the minimum number of bases or elements necessary to initiate transcription at any level .
  • Within the promoter sequence may be found a transcription initiation site (conveniently defined, for example, by mapping with nuclease Sl) , as well as protein binding domains (consensus sequences) responsible for the binding of KNA polymerase.
  • the promoter may be operably associated with other expression control sequences, including enhancer and repressor sequences.
  • operable combination refers to the linkage of nucleic acid sequences in such a manner that a nucleic acid molecule capable of directing the transcription of a given gene and/or the synthesis of a desired protein molecule is produced.
  • operable order refers to the linkage of amino acid sequences in such a manner so that a functional protein is produced.
  • vector refers to a nucleic acid assembly capable of transferring gene sequences to target cells- (e.g., viral vectors, non-viral vectors, particulate carriers, and liposomes).
  • expression vector refers to a nucleic acid assembly containing a promoter which is capable of directing the expression of a sequence or gene of interest in a cell. Vectors typically contain nucleic acid sequences encoding selectable markers for selection of cells that have been transfected by the vector.
  • vector construct refer to any nucleic acid construct capable of directing the expression of a gene of interest and which can transfer gene sequences to target cells.
  • the term includes cloning and expression vehicles, as well as viral vectors.
  • antibody refers to a whole antibody, both polyclonal and monoclonal, or a fragment thereof, for example a F(ab) 2 , Fab, FV, . VH or VK fragment, a single chain antibody, a multimeric monospecific antibody or fragment thereof, or a bi- or multi-specific antibody or fragment thereof.
  • the term also includes humanized and chimeric antibodies.
  • treating refers to executing a protocol, which may include administering one or more drugs to a patient (human or otherwise) , in an effort to alleviate signs or symptoms of the disease. Alleviation can occur prior to signs or symptoms of the disease appearing, as well as after their appearance. Thus, “treating” or “treatment” includes “preventing” or “prevention” of disease. In addition, “treating” or “treatment” does not require complete alleviation of signs or symptoms, does not require a cure, and specifically includes protocols which have only a marginal effect on the patient .
  • patient refers to a biological system to which a treatment can be administered.
  • a biological system can include, for example, an individual cell, a set of cells
  • a patient can refer to a human patient or a non-human patient.
  • remote region or “remote location” indicate a distance of at least 100 base pairs from the SNP site to the site of the disease-causing mutation, such as, for example, at least 0.5 kb, or at least 1 kb, or at least 2 kb or at least 3 kb, or at least 4 kb or at least 5 kb, or at least 6 kb or more .
  • the term "practitioner” refers to a person who uses methods, kits and compositions of the current invention on the patient.
  • the term includes, without limitations, doctors, nurses, scientists, and other medical or scientific personnel .
  • RNA molecule siRNA molecule
  • shRNA molecule RNA molecule
  • DNA molecule DNA molecule
  • cDNA molecule DNA molecule
  • nucleic acid molecule each intended to cover a single molecule, a plurality of molecules of a single species, and a plurality of molecules of different species.
  • siRNA refers to a double-stranded RNA molecule wherein each strand is between about 15 and about 30 bases of ribonucleic acid in length, and the two strands have a region of complementarity such that the two strands hybridize or "base pair" together through the annealing of complementary bases (Adenosine to Uracil, and Guanine to Cytosine) .
  • complementary bases Addenosine to Uracil, and Guanine to Cytosine
  • the two strands hybridize together in a manner such that there is an overhang of non-annealed bases at the 5' or 3 ' ends of the strand.
  • the two strands hybridize together such that each base of one strand is paired with a base of the other strand.
  • the two strands may not be 100% complementary, but may have some bases that do not hybridize due to a mismatch.
  • the RNA bases may be chemically modified, or additional chemical moieties may be conjugated to one or more ends of one or more of the strands .
  • RNA refers to a "short, hairpin” RNA molecule comprised of a single strand of RNA bases that self-hybridizes in a hairpin structure.
  • the RNA molecule is comprised of a stem region of RNA bases that hybridize together to form a double-stranded region, and a loop region of RNA bases that form the bend of the hairpin.
  • shRNA also refers to a DNA molecule from which a short, hairpin RNA molecule may be transcribed in vitro or in vivo.
  • the methods of the present invention utilize routine techniques in the field of molecular biology.
  • Basic texts disclosing general molecular biology methods include Sambrook et al . , Molecular Cloning, A Laboratory Manual (3d ed. 2001) and Ausubel et al . , Current Protocols in Molecular Biology (1994) .
  • the present invention relates generally to compositions and methods for diagnosing diseases which have an allele- specific therapy and a disease-causing mutation that is sufficiently distant from the molecular site of the therapy.
  • Table 1 depicts certain diseases applicable to the present invention. Table 1 was derived from information previously published (DiProspero (2005)). Table 1 describes in part examples of triplet repeat expansion diseases and the mutant gene associated with each disease. Table 1 Triplet Repeat: Expansion Disorders
  • ⁇ Polyalanine expansions have also been reported among mutations in other genes, including RUNX2 (runt -related transcription factor2) in cleidocranial dysplasia , ZIC2 (Zic family member 2 ) in holoprosencephaly HOXA13 (homeobox A13 ) in hand- foot -genital syndrome/ and FOXL2 (forkhead box L2 ) in type II blepharophimosis , ptosis , and epicanthus inversus syndrome .
  • Small aspartic acid repeat expansions have been reported among other mutations in the COMP (cartilage oligomeric mat4rix protein) gene in patients with multiple epiphyseal dysplasia .
  • the present invention is not limited to the diseases described above . There may be situations where a disease is caused by many different mutations in a single gene (thus designing many different gene -target ing therapies may not be practical from a commercial perspective ) . However , if one or two expressed SNPs are present in the disease -associated gene , then the SNPs may actually serve as the molecular target for the therapy (and thus determination of linkage of the SNP to the disease-causing mutation would be essential ) .
  • HD will be discus sed herein as an example of a triplet repeat expansion di sease and example of the applicability of the present invention in providing methods and kits for determining allele - speci f ic reverse transcript ion from an SNP site and analysi s of a cDNA at a region of mutation .
  • the coding region of the IT15 gene is about 13 , 000 bases long .
  • the HD disease- causing mutation is the expansion of the CAG repeat region .
  • the CAG repeat region starts at about nucleotide position 15. If the CAG triplets repeat for about 25 or 30 times, the patient is not at risk of the disease. If however, more than 37 CAG repeats occur in a row on the nucleotide sequence then the patient is going to get Huntington' s disease.
  • SNP Single Nucleotide Polymorphism
  • the practitioner also knows that one of the patient's parents has HD and would like to know if the patient will also get HD.
  • the practitioner can actually determine whether the patient is going to get HD or not, by looking at both of the patient's IT15 alleles, and determining how many CAG repeats the gene contains. If one of the CAG repeats is longer than 37, then the patient will get HD. Further, the practitioner can determine whether the patient is heterozygous (i.e., one allele has a normal number of repeats, e.g., 20, while the other allele has expanded repeats, e.g., 37) .
  • the practitioner may find that the patient received 'a "C residue” from one parent and an "A residue” from the other parent.
  • the crucial issue for the allele-specific diagnosis is which SNP is on the same mRNA transcript as the expanded number of repeats in the patient's IT15 gene. Isolating the genetic information from the patient's parents may not help because it is possible that one or both parents are also heterozygous ⁇ e.g., each parent has two SNP variants of the gene (i.e., an A residue and a C residue variants) .
  • This disclosure provides a method of determining which SNP allele of the gene co- segregates with the disease-causing mutation.
  • the test comprises a method for determining which single nucleotide polymorphism variant of an allele from a gene isolated from a heterozygous patient is on the same mRNA transcript as a disease-causing mutation at a remote region of the gene's mRNA comprising: a) an allele-specific reverse transcription reaction using an allele-specific primer which recognizes one single nucleotide polymorphism variant , wherein the 3 ' end of the allele-specific primer is positioned at the single nucleotide polymorphism nucleotide position, and b) analysis of an allele-specific cDNA product from the allele-specific reverse transcription reaction at the remote region of the gene to determine the presence or absence of the mutation on the allele-specific cDNA product.
  • the primer should preferably be shorter than about 20 nucleotides, e.g., about 15 nucleotides, long, because of a possibility that primers which are longer than about 20 nucleotides will not discriminate between the targeted SNP variants.
  • the practitioner takes RNA from the patient and applies a reverse transcription primer that recognizes just the "A allele.”
  • the "A allele” specific primer will have at its 3' position a complement to the SNP variant of interest.
  • the "A allele” specific primer will have the T at the 3' end, and so when this "A allele” specific primer anneals to the mRNA, it will base-pair with the 3' end and allow the reverse transcriptase to proceed to synthesize the cDNA from the "A allele.” Conversely, the l ⁇ A allele” primer will not base- pair at the 3' end of the primer with the "C allele” (since T is not complementary to C) . Thus, the reverse transcription polymerase will not be able to produce cDNA from the C allele.
  • the practitioner will obtain a pool of the cDNAs that corresponds to the "A allele.”
  • the reaction can be repeated in a separate tube with a "C allele” specific primer and no "A allele” primer.
  • the U C allele” specific primer will have a G on its 3' end.
  • the practitioner will perform at least one allele-specific reverse transcription reaction, but preferably two allele- specific reverse transcriptions reactions (each with its own allele-specific primer), on the mRNA from the patient.
  • the practitioner will have two sub-populations of cDNA, wherein each subpopulation is allele-specific, and the practitioner knows which pool corresponds to which variant.
  • the practitioner will be able to use any number of possible methods, the simplest being PCR to analyze the upstream portion of the cDNA containing the CAG repeat region and quantify the number of the repeats from the cDNA products that came specifically from the "C reaction” or specifically from the "A reaction.”
  • the embodiment of the invention described above employs the notion that a mismatch on the 3' position of the allele- specific primer will not allow reverse transcriptase to produce cDNA from the allele with a mismatched SNP variant .
  • a person of ordinary skill in the art will undoubtedly recognize that the 3' end of the allele- specific primer does not have to be positioned at the single nucleotide polymorphism nucleotide position.
  • a skilled artisan may design primers and conditions of the reverse transcription reaction in such a way that the allele- specific primer will not bind altogether and thus lead to the same end result: absence of cDNA the allele with a mismatched SNP variant.
  • CAG repeat length determination is based on polymerase chain reaction (PCR) amplification of genomic DNA using primers flanking the CAG repeat region in the IT15 gene, and subsequent electrophoretic separation of the products in denaturing polyacrylamide gels (Williams efc al. , (1999) Comparative semi -automated analysis of (CAG) repeats in the HD gene: use of internal standards. MoI. Cell Probes, 13:283-289).
  • PCR polymerase chain reaction
  • Examples include the Superscript Double Strand cDNA
  • RNA molecules e.g., mRNA, hnRNA, rRNA, tRNA, miRNA, snoRNA, non-coding RNAs
  • the RNA may be obtained from a mammalian or more preferably human tissue or cell source.
  • the methods of the present invention are particularly suited for amplification of RNA from small numbers of cells, including single cells, which can be purified from complex cellular samples using, e.g., micromanipulation, fluorescence-activated cell sorting (FACS) and laser microdissection techniques (see Player et al . , Expert Rev. MoI. Diagn. 4:831 (2004)).
  • Any reverse transcriptase can be used in the initial reverse transcription reaction, including thermostable, RNAse H + and RNase H ⁇ reverse transcriptases.
  • an RNase H ' reverse trancriptase is used-
  • Primers for first strand cDNA synthesis can be obtained commercially or synthesized and purified using techniques well known in the art. As disclosed above, the inventors discovered that primers which are about 15 nucleotides long provide the best results in terms of discriminating between the SNP variants and efficiency in producing the allele- specific cDNA.
  • PCR amplifications of the CAG repeat region have primarily been performed by incorporating [ ⁇ - 32 P] dNTPs, or using 32 P or fluorescently end- labeled primers. Sizing of fluorescently end-labeled amplification products was performed in various Applied Biosysterns DNA sequencers
  • kits may be prepared according to present invention.
  • a kit may include a single stranded promoter template comprising at least one RNA polymerase recognition sequence; and instructional materials for synthesizing cDNA molecules using said promoter template. While the instructional materials typically comprise written or printed materials, they are not limited to such. Any medium capable of storing such instructions and communicating them to an end user is contemplated by this invention. Such media include, but are not limited to, electronic storage media (e.g., magnetic discs, tapes, cartridges, chips), optical media (e.g., CD ROM), and the like. Such media may include addresses to internet sites that provide such instructional materials.
  • kits of the present invention may further include one or more of the following components or reagents: a reverse transcriptase (preferably with DNA-dependent DNA polymerase activity) ; an RNase inhibitor; an enzyme for attaching a 3' oligodeoxynucleotide tail onto DNA molecules
  • kits may include buffers, primers (e.g., oligodT primers, random primers), nucleotides, labeled nucleotides, an RNase inhibitor, polyA polymerase, RNase-free water, containers, vials, reaction tubes, and the like compatible with the synthesis of sRNA molecules according to the methods of the present invention.
  • the components and reagents may be provided in containers with suitable storage. media.
  • another aspect of the present invention provides a method of treating a patient susceptible to Huntington's disease comprising: a) determining which single nucleotide polymorphism variant is on the same mRNA transcript as a disease-causing mutation according to an allele-specific reverse transcription reaction using an allele-specific primer which recognizes one single nucleotide polymorphism variant, wherein further the 3' end of the primer is positioned at the single nucleotide polymorphism nucleotide position, and b) analysis of the resulting cDNA product from the reverse transcription reaction at the region of the mutation to determine ' the presence or absence of the mutation on this allele-specific cDNA product, and c) applying an allele-specific therapy to the SNP variant.
  • the allele-specific therapy comprises an RNA molecule comprising a double- stranded portion, wherein the single nucleotide polymorphism site is located within seven nucleotides from an end of the double stranded portion.
  • the double stranded portion is between 15 and 23 nucleotides long, e.g., about 19 nucleotides long.
  • the siRNA molecule may contain a loop (e.g., shRNA) , a 3' overhand or a 5' overhang which are outside of the double-stranded portion.
  • the instant invention also provides a method of allele-specific therapy, wherein the double stranded portion does not contain a mismatch in a position adjacent to the single nucleotide polymorphism site.
  • the siRNA molecule does not contain any mismatches and one strand of the double-stranded portion is 100% identical to the portion of the targeted mRNA transcript.
  • the disease treated by the allele-specific therapy is Huntington's disease
  • the non-limiting example of a single nucleotide polymorphism site suitable for the allele- specific therapy is rs262125.
  • the allele-specific therapy could itself operate at a different SNP site than the SNP site used to make the determination about which allele contains the mutation, so long as the SNP site of the therapy target and the SNP site used to identify the mutation-containing allele are already determined, before the therapy is administered to the patient, to be linked; that is, on the same mRNA transcript.
  • the allele-specific therapy comprises allele-specific RNA interference using siRNA or shRNA.
  • the allele-specific therapy destroys the "A allele" of the patient.
  • the siRNA targets the "A allele" upon introduction into the subject's brain by any method known to those of skill in the art (See for example, U.S. Application No. 11/253,393, U.S. Application No. 10/852,997, U.S. Application No. 10/721,693, U.S. Application No. 11/157,608, and PCT Patent Application No. US05/022156, which are incorporated herein in their entirety) .
  • siRNA When the siRNA is delivered into a cell it is used by proteins in the cell (known as the RISC complex) to find and destroy the tnRNA from the Huntington' s gene that has the "A allele." Thus, the messenger RNA is destroyed before it can be used to make protein. Conversely, the allele that came from the healthy parent does not get destroyed and so its messenger RNA still survives to be used to make functional biologically active protein.
  • the design and use of small interfering RNA complementary to mRNA targets that produce particular proteins is a recent tool employed by molecular biologists to prevent translation of specific mRNAs.
  • Various groups have been recently studying the effectiveness of siRNAs as biologically active agents for suppressing the expression of specific proteins involved in neurological disorders. Caplen et al .
  • siRNA molecule corresponding to at least a portion of a gene containing an SNP variant of an allele in a heterozygous subject that is on the same mRNA transcript as a disease- causing mutation located at a remote region of the gene's mRNA, wherein such siRNA nucleic acid sequence is capable of inhibiting expression of the mRNA transcript containing the disease-causing mutation in a cell.
  • siRNAs are typically short (19-29 nucleotides) , double-stranded RNA molecules that cause sequence- specific degradation of complementary target mRNA known as RNA interference (RNAi) . Bass, Nature 411:428 (2001) .
  • the siRNA molecules comprise a double-stranded structure comprising a sense strand and an antisense strand, wherein the antisense strand comprises a nucleotide sequence that is complementary to at least a portion of a desired nucleic acid sequence and the sense strand comprises a nucleotide sequence that is complementary to at least a portion of the nucleotide sequence of said antisense region, and wherein the sense strand and the antisense strand each comprise about 19-29 nucleotides .
  • nucleic acid sequence can be targeted by the siRNA molecules of the present invention.
  • Nucleic acid sequences encoding desired gene targets are publicly available from Genbank.
  • the siRNA molecules targeted to desired sequence can be designed based on criteria well known in the art (e.g., Elbashir et al. t EMBO J. 20:6877 (2001)).
  • the target segment of the target mRNA preferably should begin with AA (most preferred) , TA, GA, or CA; the GC ratio of the siRNA molecule preferably should be 45-55%; the siRNA molecule preferably should not contain three of the same nucleotides in a row; the siRNA molecule preferably should not contain seven mixed G/Cs in a row,- the siRNA molecule preferably should comprise two nucleotide overhangs
  • the target segment preferably should be in the ORF region of the target mRNA and preferably should be at least 75 bp after the initiation
  • siRNA molecules targeted to desired sequences can be designed by one of skill in the art using the aforementioned criteria or other known criteria (e.g., Gilmore et al . , J. Drug Targeting 12:315 (2004); Reynolds et al . , Nature Bi ⁇ technol . 22:326 (2004); Ui-Tei et al . , Nucleic Acids Res. 32:936 (2004)).
  • siRNA molecules targeted to desired sequences can be produced in vitro by annealing two complementary single- stranded RNA molecules together (one of which matches at least a portion of a desired nucleic acid sequence) (e.g.,
  • RNA molecules which folds back on itself to produce the requisite double-stranded portion (Yu et al . , Proc. Natl. Acad. Sci. USA 99:6047 (2002)).
  • shRNA short hairpin RNA
  • siRNA molecules targeted to desired target sequences can be introduced into cells to inhibit expression.
  • DNA molecules from which shRNA molecules targeted to desired target sequences can be introduced into cells to inhibit expression.
  • another aspect of the present invention provides for inhibiting expression of an mRNA sequence containing an SNP allele and a disease- causing mutation in a cell comprising introducing into a cell at least one siRNA molecule or shRNA molecule that corresponds to at least a portion of the mRNA nucleic acid sequence. Any cell can be targeted.
  • the siRNA or shRNA molecules are introduced into a heart cell or brain cell.
  • the brain cell is from a subject at risk for HD, i.e., the offspring of a HD patient.
  • the siRNA molecules produced herein can be introduced into cells in vitro or ex vivo using techniques well-known in the art, including electroporation, calcium phosphate co-precipitation, microinjection, lipofection, polyfection, and conjugation to cell penetrating peptides (CPPs) .
  • the siRNA molecules can also be introduced into cells in vivo by- direct delivery into specific organs such as the liver, brain, eye, lung and heart, or systemic delivery into the blood stream or nasal passage using naked siRNA molecules or siRNA molecules encapsulated in biodegradable polymer microspheres (Gilmore et al . , ⁇ 7. Drug Targeting 12:315 (2004) ) .
  • siRNA molecules targeted to specific mRNA sequences can be introduced into cells in vivo by endogenous production from an expression vector (s) encoding the sense and antisense siRNA sequences.
  • an expression vector comprising at least one DNA sequence encoding a siRNA molecule corresponding to at least a portion of a specific mRNA nucleic acid sequence capable of inhibiting expression of a specific mRNA in a cell operably linked to a genetic control element capable of directing expression of the siRNA molecule in a cell.
  • Expression vectors can be transfeeted into cells using any of the methods described above.
  • Genetic control elements include a transcriptional promoter, and may also include transcription enhancers to elevate the level of mRNA expression, a sequence that encodes a suitable ribosome binding site, and sequences that terminate transcription.
  • Suitable eukaryotic promoters include constitutive RNA polymerase II promoters (e.g., cytomegalovirus (CMV) promoter, the SV40 early promoter region, the promoter contained in the 3 ' long terminal repeat of Rous sarcoma virus (RSV) , the herpes thymidine kinase (TK) promoter, and the chicken beta-actin promoter) , cardia ⁇ -tissue-specific RNA polymerase II promoters (e.g., the ventricular myosin light chain 2 (MLC-2v) promoter, and the sodium-calcium exchanger gene Hl promoter (NCXlHl) ) , and RNA polymerase III promoters (e.g., U6, Hl, 7SK and
  • the sense and antisense strands of siRNA molecules are encoded by different expression vectors (i.e., cotransfected) (e.g., Yu et al . , Proc. Natl. Acad. Sci. USA 99:6047 (2002).
  • the sense and antisense strands of siRNA molecules are encoded by the same expression vector.
  • the sense and antisense strands can be expressed separately from a single expression vector, using either convergent or divergent transcription (e.g., Wang et al . , Proc. Natl. Acad. Sci. USA 100:5103 (2003); Tran et al., BMC Biotechnol. 3:21 (2003)).
  • the sense and antisense strands can be expressed together from a single expression vector in the form of a single hairpin RNA molecule, either as a short hairpin RNA (shRNA) molecule (e.g., Arts et al . , Genome Res. 13:2325 (2003)) or a long hairpin RNA molecule (e.g., Paddison et al . , Proc. Natl. Acad. Sex. USA 99:1443 (2002)).
  • shRNA short hairpin RNA
  • a long hairpin RNA molecule e.g., Paddison et al . , Proc. Natl. Acad. Sex. USA 99:1443 (2002).
  • viral expression vectors are preferred, particularly those that efficiently transduce heart cells (e.g., alphaviral, lentiviral, retroviral, adenoviral, adeno-associated viral (AAV)) (Williams and Koch, Annu. Rev. Physiol. 66:49 (2004); del Monte and Hajjar, J. Physiol. 546.1:49 (2003).
  • alphaviral, lentiviral, retroviral, adenoviral, adeno-associated viral (AAV) e.g., alphaviral, lentiviral, retroviral, adenoviral, adeno-associated viral (AAV)
  • transgenes including transgenes directed to deseases
  • cardiomycoytes e.g., Iwanaga et al., J. Clin. Invest. 113:727 (2004); Seth et al . , Proc.
  • AAV vectors for sustained gene expression in mouse and hamster myocardium and arteries for over one year (Li et al., Gene Ther. 10:1807 (2003); Vassalli et al . , Int. J. Cardiol. 90:229 (2003)) .
  • expression vectors based on AAV serotype 6 have been shown to efficiently transduce both skeletal and cardiac muscle (e.g., Blankinship et al . , MoI . Ther. 10:671 (2004)).
  • the present invention also provides for the use of coxsackie viral vectors for delivery of desired siRNA sequences.
  • Desired gene products include, for example, desired mRNA and desired polypeptide, and both can be measured using methods well-known to those skilled in the art.
  • desired mRNA can be directly detected and quantified using, e.g., Northern hybridization, in situ hybridization, dot and slot blots, or oligonucleotide arrays, or can be amplified before detection and quantitation using, e.g., polymerase chain reaction (PCR), reverse-transcription-PCR (RT-PCR) , PCR-enzyme-linked immunosorbent assay (PCR-ELISA) , or ligase chain reaction (LCR) .
  • PCR polymerase chain reaction
  • RT-PCR reverse-transcription-PCR
  • PCR-ELISA PCR-enzyme-linked immunosorbent assay
  • LCR ligase chain reaction
  • Desired polypeptide can be detected and quantified using various well-known immunological assays, such as, e.g., enzyme-linked immunosorbent assay (ELISA) , radioimmunoassay (RIA) , immunoprecipitation, immunofluorescence, and Western blotting.
  • immunological assays such as, e.g., enzyme-linked immunosorbent assay (ELISA) , radioimmunoassay (RIA) , immunoprecipitation, immunofluorescence, and Western blotting.
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • immunoprecipitation immunofluorescence
  • Western blotting Western blotting.
  • Anti-deaired antibodies preferably anti-human desired for use in immunological assays are commercially available from, e.g., EMD Biosciences (San Diego, CA),
  • RNA-isolation and R ⁇ vers ⁇ transcription reaction Applicants analyzed . the CAG-repeat sequences in the Huntington's disease gene using the following allele- specific reverse transcription reaction. Table 2 defines the sequences for various allele-specific reverse transcription primers for use in determining which allele of a heterozygous patient's Huntington's disease gene contains the disease-causing allele, in accordance with the subject invention.
  • Fibroblasts purchased from Coriell Cell Repositories
  • the growth medium was minimum essential medium containing 20% FBS, 1% PSN antibiotics, 1% fungizone, 2% non-essential amino acids, 2% amino acids and 2% vitamins
  • Genomic DNA was isolated using the DNeasy kit (Qiagen) and nucleotide identity for HD alleles at various SNP positions was determined by pyrosequencing conducted by Isogen Lifescience (The
  • Fibroblasts from 21 individuals from different kindreds, diagnosed with HD or known to be at risk for HD were purchased from Coriell Cell Repositories (Camden, NJ) . DNA was harvested from each fibroblast culture and pyrosequenced to determine the nucleotide identity at eleven known single nucleotide polymorphism (SNP) sites within the protein coding region of the HD sequence. Heterozygosity was found at four of the eleven SNP sites tested. Ten of the 21 genotyped fibroblast cultures (see Table 3) were heterozygous for at least 1 SNP site.
  • SNP single nucleotide polymorphism
  • GM04022 fibroblasts is heterozygous at SNP position rs363125 (NCBI) , harboring both an A-allele (adenine) and a C-allele (cytosine) at nucleotide position
  • donor-1 The fibroblasts from this donor (here called donor-1) were selected for further study. These fibroblasts are from a female Caucasian donor who was 28 years old at the time of cell collection. Her mother, who was 58 years old at the time of the cell collection, was reportedly diagnosed with HD at age 49.
  • donor-1 was determined to be heterozygous (adenine versus cytosine) at SNP site rs363125.
  • SNP site rs363125 In order to design an allele-specific RNA interference-based therapy for donor-1, it needs to be determined which of these SNP sequences is associated with the expanded CAG repeat mutation that is located approximately 5000 nucleotides upstream from the SNP position.
  • Appropriate siRNA or shRNA targeting SNP site rs363125 should only reduce protein expression from the allele that contains the expanded CAG repeat and should therefore only be specific for the associated SNP.
  • Correspondence between the SNP identity and the expanded allele cannot be readily determined by cDNA sequencing or by comparing the lengths of PCR products spanning the SNP position and the CAG repeat region.
  • RT reverse transcription
  • the primers each contained either guanine or thymine at the 3' terminal position, corresponding to SNP site rs363125, as shown in FIG. 1. Primers of various lengths were tested.
  • the 3 '-guanine and 3' -thymine primer were each designed to specifically facilitate reverse transcription (also known as first strand synthesis) of only the mRNA containing cytosine or adenine at the SNP position,- respectively.
  • RNA isolated from the fibroblasts was reverse transcribed using the Superscript III RT kit (Invitrogen) in the presence of 100 nM of one of the following DNA primers: the 20-mer 5'-GTGTTCTTCTAGCGTTGAAT-S', SEQ ID NO: 52 (or a shorter, corresponding 15-mer or 10-mer ending in T-3', corresponding to SEQ ID NO: 47 and SEQ ID NO: 42, respectively) or the 20-mer primer ⁇ '-GTGTTCTTCTAGCGTTGAAG- 3' SEQ ID NO: 64 (or a shorter, corresponding 15-mer or 10- mer primer ending in G-3', corresponding to SEQ ID NO: 59 and SEQ ID NO: 54, respectively) at 100 nM .
  • the 20-mer 5'-GTGTTCTTCTAGCGTTGAAT-S' SEQ ID NO: 52 (or a shorter, corresponding 15-mer or 10-mer ending in T-3', corresponding to SEQ ID NO: 47 and SEQ ID NO: 42,
  • the CAG repeat sequence on either RT product was then amplified by PCR (Bio-Rad iCycler) using Accuprime GC-Rxch DNA-polymerase (invitrogen) , and forward primer 5'-GCCTTCGAGTCCCTCAAGT -3' and reverse primer 5'-GACAATGATTCACACGGTCT-S' at 0.2 ⁇ M each (SEQ ID NO: 85 and SEQ ID NO: 86, respectively) .
  • the resulting PCR products contain the complete CAG repeat sequence of one of the two alleles of the GM04022 cells.
  • CAG-repeat size for each allelic RT products was determined using standard 1.5% agarose gel electrophoresis with ethidium bromide staining, and also by sequencing of the products of the PCR amplification of the CAG repeat region.
  • beacons are synthetic oligonucleotide probes that have a fluorophore and a quencher covalently linked to the respective ends of the ⁇ ligo. In solution, the beacon adopts a hairpin conformation, causing the fluorophore to be quenched. However, upon hybridization with complementary DNA in a PCR reaction, the hairpin conformation is lost and fluorescence from the fluorophore can be detected. Beacons can be constructed such that as little as a single nucleotide mismatch between the beacon and the complementary
  • DNA is sufficient for the probe to be more stable in its self-annealed state than in the probe -cDNA hybrid.
  • the inventors designed two such beacons corresponding to the two allelic variants of SNP rs363125 (Fig. 3) . Absence of genomic DNA carry over in the RNA isolates was confirmed by PCR using forward primer S'-CTCGCCTCCAGCATGAAAGT-S' and reverse primer S'-GTGACTGGGGCATTGATT-B', (SEQ ID NOS: 87 and 88, respectively) which amplify monocyte chemo-attractant protein-1 (MCP-I) genomic DNA.
  • MCP-I monocyte chemo-attractant protein-1
  • Genomic DNA from HeLa cells served as a positive control for the PCR reaction, and only total RNA samples from fibroblasts verified to be without detectable genomic DNA were used in further analyses . Concentration and integrity of the isolated RNA were checked by Experion automated electrophoresis (Bio-Rad, StdSens analysis kit) of the 28S/18S ribosomal RNA. RNA was reverse transcribed into cDNA using the iScript cDNA synthesis kit (BioRad) , after which allele-specific real time PCR was conducted using the molecular beacon method.
  • the sequences of the molecular beacons are described in Figure 3 and correspond to SEQ ID NOs: 89 (3 '-BHQI- tcacgcaGATCGCAACTTAATGACAGGGtgcgtga-FAM-5 1 ) and 90 (3 '-BHQI- gcgctagAGATCGCAACTTCATGACAGGGGTAGctagcgc- FAM-5'), for the "A” and the "C” alleles, respectively.
  • the "loop sequence” which binds to a portion of the targeted portion of the PCR product is in capital letters, while the "stem” portions, which form the double strand of the beacon if the beacon is not bound to the targeted portion of the PCR product.
  • sequences of the primers for the "A" allele correspond to SEQ ID NOs: 91 ⁇ 5' CCTTCTCTCCGTATTTAATCTCCTGTA-3 ' ) and 92 (5 1 TCATTTCCACCTTCAGCTGTTTTGTAA-3 •) for the forward and the reverse primers, respectively.
  • sequences of the primers for the "C” allele correspond to SEQ ID NOs 93 (5 1 AGATATTGTTCTTTCTCGTATTCAGG-S ') and 94 (5 1 TGCTCACTCATTTCCACCTTC-3 ' ) for the forward and the reverse primers, respectively.
  • Detection of the PCR product was conducted using molecular beacons (Proligo, France) each of which detects only a PCR product containing a complementary nucleotide at the SNP position.
  • the two alleles were quantified in separate PCR reactions, each containing 0.3 ⁇ M of the appropriate molecular beacon. During elongation, increase of the FAM fluorescence signal was recorded. The data were quantified against a standard curve generated using a serial dilution of a plasmid containing the PCR product (Baseclear genesynthesis , The Netherlands).
  • Results were normalized against GAPDH, amplified using IQ SYBR Green Supermix (Bio-Rad) with forward primer 5'- ACTCCTCCACCTTTGACGC-3' (SEQ ID NO: 95) and reverse primer 5' -GTTGCTGTAGCCAAATTCGTT-3 ' (SEQ ID NO : 96).
  • beacon MB363125_A and MB363125_C were found to yield a PCR amplification signal only from their corresponding DNA template .
  • siRNA has a guanine located at position 16 relative to the 3' end of the complementary region of the target huntingtin mRNA, providing specificity for the allele containing cytosine at the SNP position.
  • a parallel cell culture was transfected with a fluorescently labeled Block-It siRNA (Invitrogen) to verify efficiency of Lipofectamine 2000 transfection .
  • the cells were incubated overnight at 37°c, 5% CO 2 and maximum humidity. The cultures were then washed with PBS and fluorescence microscopy (Leica DM-IRB) was used to confirm transfection in the Block-It transfected cultures, which were considered representative for all transfection conditions.
  • the cells were cultured for another day before RNA isolation as described, approximately 48 hours post- transfection.
  • Fibroblasts from donor-1 were transfected with siRNA designed to specifically target the mRNA containing cytosine at SNP site rs363125.
  • This siRNA molecule (siRNA 363125_C- 16) was designed such that the cytosine nucleotide of the SNP is located at position 16 relative to the 5' end of the sense strand of the siRNA molecule.
  • the amount of mRNA from both endogenous alleles was separately quantified using the molecular beacons developed to be specific for the allelic variants at this SNP site.

Abstract

Methods and kits are provided for determining which single nucleotide polymorphism ('SNP') variant of an allele of a heterozygous patient is on the same allele as a disease- causing mutation that is at a remote region of the gene's mRNA comprising a) an allele specific reverse transcription reaction using an allele specific primer which recognizes one SNP variant, wherein further the 3' end of the primer is positioned at the SNP nucleotide position, and b) analysis of the resulting cDNA product from the reverse transcription reaction at the region of the mutation to determine the presence or absence of the mutation on this allele specific cDNA product, wherein the allele specific primer is shorter than about 20 nucleotides.

Description

METHODS AND KITS FOR LINKING POLYMORPHIC SEQUENCES TO EXPANDED REPEAT MUTATIONS
CROSS REFERENCE TO RELATED APPLICATION (S ) This application claims the benefit .of U.S. Patent Application No. 11/439,858, filed on May 24, 2006, the entire teachings of which are incorporated herein by reference.
FIELD OF THE INVENTION The present invention relates generally to compositions and methods for diagnosing diseases which have an allele- speσific therapy and a disease-causing mutation that is sufficiently distant from the molecular site of the therapy to require a diagnostic linking method. BACKGROUND OF THE INVENTION
Expansions of CAG trinucleotide repeats (CAG repeats) in coding regions of human genes cause numerous disorders by generating proteins with elongated polyglutamine (polyQ) stretches. This group of disorders includes by way of example Dystophia myotonica. Spinocerebellar ataxia type 1, Spinocerebellar ataxia type 2, Spinocerebellar ataxia type 3, Spinocerebellar ataxia, type 6, Spinocerebellar ataxia type 7, Spinocerebellar ataxia type 8, Spinocerebellar ataxia type 17, Huntington disease-like 2, Spinal and bulbar muscular atrophy, Huntington disease, Dentatorubral - pallidoluysian atrophy, Oculopharyngeal dystrophy. Congenital central hypoventilation syndrome, Infantile spasms, Synpolydactyly, Cleidocranial dysplasia, Holoprosencephaly, Hand-foot -genital syndrome, Type II blephorophimosis , ptosis, and epicanthus inversus syndrome. (Wanker E. E. (2000) Biol. Chem. , 381.-937-942; Gusella J.F. and MacDonald, M. E. (2000) Nature Rev. Neurosc±. , 1:109-115; and Usdin K. and Grabczyk,E. (2000) Cell. MoI. Life Sex., 57:914-931) .
For purposes of illustration only Huntington's disease (HD) will be discussed herein. HD is a chronic neurodegenerative disorder which is inherited as an autosomal dominant trait and is characterized by chorea, dementia and personality disorder. Martin, J. B. and Gusella,J.F. (1986) N. Engl. J. Med. 315:1267-1276. The gene responsible for HD contains an expanded and unstable CAG trinucleotide repeat. Huntington's Disease Collaborative Research Group (1993) Cell 72:971-983.
The HD gene (IT15 gene) , which encodes huntingtin, a 350 kDa protein of unknown function, is located on the human chromosome 4 and consists of 67 exons . The disease-causing mutation is a CAG repeat expansion located within exon 1 of the HD gene (HD exonl) . The CAG repeat is translated into a polyQ stretch. The disease manifests itself when the polyQ stretch exceeds the critical length of 37 glutamines
(pathological threshold) , whereas 8-35 glutamine residues in huntingtin are tolerated by neuronal cells. Experimental evidence has been presented that huntingtin fragments with polyQ tracts in the pathological range (more than 37 glutamines) , but not in the normal range (20-32 glutamines) , form high molecular weight protein aggregates with a fibrillar morphology in vitro and in cell culture model systems (Scherzinger et al . (1999) Proc. Natl Acad. Sci. USA, 96:4604-4509; and Waelter et al . , (2001) MoI. Biol. Cell, 12:1393-1407). In addition, inclusions with aggregated N-terminally truncated huntingtin protein were detected in HD transgenic mice carrying a CAG repeat expansion of 115-156 units and in HD patient brains (Davies et al . , (1997) Cell, 90:537-548; and DiFiglia et al . , (1997) Science, 277:1990-1993), suggesting that the process of aggregate formation may be important for the progression of HD. The mechanisms, however, by which the elongated polyQ sequences in huntingtin cause dysfunction and neurodegeneration are not yet understood (Scherzinger et al., (1999); Tobin A.J. and Signer, E.R. (2000) Trends Cell Biol., 10 : 531-536; and Perutz M. F. (1999) Glutamine repeats and neurodegenerative diseases: molecular aspects. Trends Biochem. Sci . , 24:58-63). Unaffected individuals have repeat numbers of up to 30, while individuals at a high risk of developing HD carry more than 37 CAG repeats. Individuals with 30-37 repeats have a high risk of passing on repeats in the affected size range to their offspring (Andrew et al . , (1997) Hum. MoI. Genet., 6:2005-2010; Duyao et al . , (1993) Nature Genet., 4:387-392; and Snell et al., (1993) Nature Genet., 4:393-397).
It is known that patients are able to survive and live healthy lives with only one functioning copy of the IT15 gene. Thus, selective inactivation of the allele with a disease-causing mutation should diminish or even eliminate the disease while improving the possibilities of survival in heterozygous patients.
The combination of emotional, cognitive and motor symptoms in HD contributes to an unusually high cost of care. People with Huntington's Disease require care from health professionals of many stripes, including general practitioners, neurologists, social workers, home health aides, psychologists, physical therapists, and speech/language pathologists. Currently, there are a few diagnostic approaches for nucleic acid sequence identification. U.S. Patent Application Publication No. 20040048301 describes allele- specific primer extension in the presence of labeled nucleotides for sequence identification, but does not include allele-specific primer extension for enrichment of one allele over the other for further analysis of the allele of interest as part of the kit. WO Patent Application No. 2003100101 describes isolation of one sequence in a mixture by hybridization markers and single-strand specific nucleases for use in single-molecule analysis. U.S. Patent Application Publication No. 20030039964 describes a method for isolation of one sequence in a mixture by hybridization to a fixed probe, but does not disclose the use of reverse transcription. U.S. Patent No. 6,013,431 describes a method for analysis of bases adjacent to a hybridized, immobilized oligo, but does not disclose enrichment of one allele over the other. WO Patent Application No. 9820166 describes a method for specific selection of one allele over the other, followed by mass spectroscopic analysis of the selected molecule, but does not ■ disclose the use of reverse transcription. None of these references disclose methods and diagnostic kits for linking polymorphic sequences to expanded repeat mutations for improved allele-specific diagnosis .
U.S. Patent Publication No. 20040241854 (Davidson) discloses allele-specific inhibition of specific single nucleotide polymorphism variants, and presents data showing that "expanded CAG repeats and adjacent sequences, while accessible to RNAi, may not be preferential targets for silencing" thus describing the problem that our invention addresses (determining what SNP variant at a remote molecular position is linked to the expanded CAG repeat in a particular patient) , but does not teach the use of reverse transcription using an allele-specific primer to solve this problem, nor otherwise disclose a method for how to solve this problem. U.S. Patent Publication No. 20060270623 (McSwiggen) discloses multiple siRNA sequences, including those comprising SNP variants, but does not provide any working examples regarding allele-specific RNA interference using these disclosed siRNA sequences, nor disclose how to determine which allele-specific siRNA to administer to a particular Huntington' s disease human patient in order to effectively treat that patient's disease by suppression of only the expanded Huntington allele in that patient.
Accordingly, there is need in the art for novel compounds, methods, and kits for allele-specific diagnostics and therapies. SUMMARY OF THE INVENTION Applicants have invented methods and kits for determining which variant allele of a single nucleotide polymorphism ("SNP") located at a distance from a disease- causing mutation co-segregates with the disease-causing mutation. In other words, the invention will allow for the determination of which SNP allele is located on the same mRNA transcript as the transcribed disease-causing mutation.
In one aspect, the invention provides a method for determining which single nucleotide polymorphism variant of an allele from a gene isolated from a heterozygous patient is on the same mRNA transcript as a disease-causing mutation at a remote region of the gene's mRNA comprising: a) an allele-specific reverse transcription reaction using an allele-specific primer which recognizes one single nucleotide polymorphism variant, and b) analysis of an allele-specific cDNA product from the allele-specific reverse transcription reaction at the remote region of the gene to determine the presence or absence of the mutation on the allele-specific cDNA product, wherein the allele- specific primer is shorter than about 20 nucleotides. Xn one embodiment, the 3' end of the allele-specific primer is positioned at the single nucleotide polymorphism nucleotide position.
In another aspect, the invention provides a method of treating a patient comprising determining which SNP variant is on the same mRNA transcript as a disease-causing mutation according to the method recited above, and applying an allele-specific therapy to the SNP variant, wherein the allele-specific therapy comprises an siRNA comprising a double-stranded portion, wherein the single nucleotide polymorphism site is located within seven nucleotides from an end of the double stranded portion. The allele-specific therapy of the present invention includes by way of example allele-specific RNA interference using siRNA or shRNA.
In yet another aspect, the invention provides a kit for determining which single nucleotide polymorphism variant of an allele of a heterozygous patient is on the same mRNA transcript as a disease-causing mutation located at a remote region of the gene' s mRNA comprising a) an allele-specific primer which recognizes one single nucleotide polymorphism variant, and b) a set of instructions, wherein the allele- specific primer is shorter than about 20 nucleotides. In one embodiment, the 3' end of the allele-specific primer is positioned at the single nucleotide polymorphism nucleotide position.
BRIEF DESCRIPTION OF FIGURES
FIG. 1 is a schematic illustration of allele-specific reverse transcription for the "A" allele.
FIG. 2 is a photograph of a gel showing results of allele-specific reverse transcription. FIG. 3 is an illustration of primers and molecular beacons .
FIG. 4 is an illustration of allele specificity of the molecular beacons used for the allele-specific quantification of real-time PCR product
FIG. 5 illustrates that a siRNA molecule designated as 363125_C-16 inhibits the mutant, but not the wild-type huntingtin mRNA.
DETAILED DESCRIPTION OF THE INVENTION The present invention relates to methods and kits for performing allele-specific reverse transcription from an SNP site and analysis of a cDNA at a region of gene mutation. The methods, systems and reagents of the present invention are applicable to any disease which contains an SNP variant of an allele in a heterozygous subject that is on the same mRNA transcript as a disease-causing mutation that is at a remote region of the gene's mRNA.
To aid in the understanding of the invention, the following non-limiting definitions are provided: The term "gene" refers to a DNA sequence that comprises control and coding sequences necessary for the production of a polypeptide or its precursor. The polypeptide can be encoded by a full length coding sequence (either genomic DNA or cDNA) or by any portion of the coding sequence so long as the desired activity is retained. In some aspects, the term "gene" also refers to an mRNA sequence or a portion thereof that directly codes for a polypeptide or its precursor.
The term "transfection" refers to the uptake of foreign DNA by a cell. A cell has been "transfected" when exogenous (i.e., foreign) DNA has been introduced inside the cell membrane. Transfection can be either transient (i.e., the introduced DNA remains extraσhromosomal and is diluted out during cell division) or stable (i.e., the introduced DNA integrates into the cell genome or is maintained as a stable episomal element) .
"Cotransfection" refers to the simultaneous or sequential transfection of two or more vectors into a given cell.
The term "promoter element" or "promoter" refers to a DNA regulatory region capable of being bound by an RNA polymerase in a cell (e.g., directly or through other promoter-bound proteins or substances) and initiating transcription of a coding sequence . A promoter sequence is, in general, bounded at its 31 terminus by the transcription initiation site and extends upstream (51 direction) to include the minimum number of bases or elements necessary to initiate transcription at any level . Within the promoter sequence may be found a transcription initiation site (conveniently defined, for example, by mapping with nuclease Sl) , as well as protein binding domains (consensus sequences) responsible for the binding of KNA polymerase. The promoter may be operably associated with other expression control sequences, including enhancer and repressor sequences.
The term "in operable combination", "in operable order" or "operably linked" refers to the linkage of nucleic acid sequences in such a manner that a nucleic acid molecule capable of directing the transcription of a given gene and/or the synthesis of a desired protein molecule is produced. The term also refers to the linkage of amino acid sequences in such a manner so that a functional protein is produced.
The term "vector" refers to a nucleic acid assembly capable of transferring gene sequences to target cells- (e.g., viral vectors, non-viral vectors, particulate carriers, and liposomes). The term "expression vector" refers to a nucleic acid assembly containing a promoter which is capable of directing the expression of a sequence or gene of interest in a cell. Vectors typically contain nucleic acid sequences encoding selectable markers for selection of cells that have been transfected by the vector. Generally, "vector construct," "expression vector," and "gene transfer vector," refer to any nucleic acid construct capable of directing the expression of a gene of interest and which can transfer gene sequences to target cells. Thus, the term includes cloning and expression vehicles, as well as viral vectors.
The term "antibody" refers to a whole antibody, both polyclonal and monoclonal, or a fragment thereof, for example a F(ab)2, Fab, FV, . VH or VK fragment, a single chain antibody, a multimeric monospecific antibody or fragment thereof, or a bi- or multi-specific antibody or fragment thereof. The term also includes humanized and chimeric antibodies.
The term "treating" or "treatment" of a disease refers to executing a protocol, which may include administering one or more drugs to a patient (human or otherwise) , in an effort to alleviate signs or symptoms of the disease. Alleviation can occur prior to signs or symptoms of the disease appearing, as well as after their appearance. Thus, "treating" or "treatment" includes "preventing" or "prevention" of disease. In addition, "treating" or "treatment" does not require complete alleviation of signs or symptoms, does not require a cure, and specifically includes protocols which have only a marginal effect on the patient . The term "patient" refers to a biological system to which a treatment can be administered. A biological system can include, for example, an individual cell, a set of cells
(e.g., a cell culture) , an organ, a tissue, or a multi- cellular organism. A patient can refer to a human patient or a non-human patient.
The terms "remote region" or "remote location" indicate a distance of at least 100 base pairs from the SNP site to the site of the disease-causing mutation, such as, for example, at least 0.5 kb, or at least 1 kb, or at least 2 kb or at least 3 kb, or at least 4 kb or at least 5 kb, or at least 6 kb or more .
The term "practitioner" refers to a person who uses methods, kits and compositions of the current invention on the patient. The term includes, without limitations, doctors, nurses, scientists, and other medical or scientific personnel .
The terms "siRNA molecule," "shRNA molecule," "RNA molecule," "DNA molecule," "cDNA molecule" and "nucleic acid molecule" are each intended to cover a single molecule, a plurality of molecules of a single species, and a plurality of molecules of different species.
The term "siRNA" refers to a double-stranded RNA molecule wherein each strand is between about 15 and about 30 bases of ribonucleic acid in length, and the two strands have a region of complementarity such that the two strands hybridize or "base pair" together through the annealing of complementary bases (Adenosine to Uracil, and Guanine to Cytosine) . For some siRNA molecules, the two strands hybridize together in a manner such that there is an overhang of non-annealed bases at the 5' or 3 ' ends of the strand. For other siRNA molecules, the two strands hybridize together such that each base of one strand is paired with a base of the other strand. For some siRNA molecules, the two strands may not be 100% complementary, but may have some bases that do not hybridize due to a mismatch. For some siRNA molecules, the RNA bases may be chemically modified, or additional chemical moieties may be conjugated to one or more ends of one or more of the strands .
The term "shRNA" refers to a "short, hairpin" RNA molecule comprised of a single strand of RNA bases that self-hybridizes in a hairpin structure. The RNA molecule is comprised of a stem region of RNA bases that hybridize together to form a double-stranded region, and a loop region of RNA bases that form the bend of the hairpin. The term "shRNA" also refers to a DNA molecule from which a short, hairpin RNA molecule may be transcribed in vitro or in vivo.
The methods of the present invention utilize routine techniques in the field of molecular biology. Basic texts disclosing general molecular biology methods include Sambrook et al . , Molecular Cloning, A Laboratory Manual (3d ed. 2001) and Ausubel et al . , Current Protocols in Molecular Biology (1994) .
The present invention relates generally to compositions and methods for diagnosing diseases which have an allele- specific therapy and a disease-causing mutation that is sufficiently distant from the molecular site of the therapy. Table 1 depicts certain diseases applicable to the present invention. Table 1 was derived from information previously published (DiProspero (2005)). Table 1 describes in part examples of triplet repeat expansion diseases and the mutant gene associated with each disease. Table 1 Triplet Repeat: Expansion Disorders
Figure imgf000013_0001
Figure imgf000014_0001
♦Polyalanine expansions have also been reported among mutations in other genes, including RUNX2 (runt -related transcription factor2) in cleidocranial dysplasia , ZIC2 (Zic family member 2 ) in holoprosencephaly HOXA13 (homeobox A13 ) in hand- foot -genital syndrome/ and FOXL2 (forkhead box L2 ) in type II blepharophimosis , ptosis , and epicanthus inversus syndrome . Small aspartic acid repeat expansions have been reported among other mutations in the COMP (cartilage oligomeric mat4rix protein) gene in patients with multiple epiphyseal dysplasia .
The present invention is not limited to the diseases described above . There may be situations where a disease is caused by many different mutations in a single gene (thus designing many different gene -target ing therapies may not be practical from a commercial perspective ) . However , if one or two expressed SNPs are present in the disease -associated gene , then the SNPs may actually serve as the molecular target for the therapy (and thus determination of linkage of the SNP to the disease-causing mutation would be essential ) .
For purposes of illustration, only HD will be discus sed herein as an example of a triplet repeat expansion di sease and example of the applicability of the present invention in providing methods and kits for determining allele - speci f ic reverse transcript ion from an SNP site and analysi s of a cDNA at a region of mutation .
The coding region of the IT15 gene is about 13 , 000 bases long . The HD disease- causing mutation is the expansion of the CAG repeat region . The CAG repeat region starts at about nucleotide position 15. If the CAG triplets repeat for about 25 or 30 times, the patient is not at risk of the disease. If however, more than 37 CAG repeats occur in a row on the nucleotide sequence then the patient is going to get Huntington' s disease.
About ten thousand bases downstream from the CAG repeat sequence, there is a natural variation (Single Nucleotide Polymorphism, or SNP) of the ITl5 gene in the human population, where for many people it might be an "A residue" and for many others it is a "C residue" . That is just a normal variation, as it does not cause any disease. The information about the SNP can be used to determine that a child of a Huntington' s disease patient has inherited an allele with the "A residue" from one parent and an allele with the "C residue" from the other parent.
The practitioner also knows that one of the patient's parents has HD and would like to know if the patient will also get HD. The practitioner can actually determine whether the patient is going to get HD or not, by looking at both of the patient's IT15 alleles, and determining how many CAG repeats the gene contains. If one of the CAG repeats is longer than 37, then the patient will get HD. Further, the practitioner can determine whether the patient is heterozygous (i.e., one allele has a normal number of repeats, e.g., 20, while the other allele has expanded repeats, e.g., 37) . Analyzing the IT15 gene downstream of the CAG repeats, the practitioner may find that the patient received 'a "C residue" from one parent and an "A residue" from the other parent. Thus, the crucial issue for the allele-specific diagnosis is which SNP is on the same mRNA transcript as the expanded number of repeats in the patient's IT15 gene. Isolating the genetic information from the patient's parents may not help because it is possible that one or both parents are also heterozygous {e.g., each parent has two SNP variants of the gene (i.e., an A residue and a C residue variants) . This disclosure provides a method of determining which SNP allele of the gene co- segregates with the disease-causing mutation.
One aspect of the present invention provides a diagnostic test, allowing the practitioner to determine which allele, classified by the nucleotide at the SNP position, co-segregates with the disease-causing mutation. In one embodiment, the test comprises a method for determining which single nucleotide polymorphism variant of an allele from a gene isolated from a heterozygous patient is on the same mRNA transcript as a disease-causing mutation at a remote region of the gene's mRNA comprising: a) an allele-specific reverse transcription reaction using an allele-specific primer which recognizes one single nucleotide polymorphism variant , wherein the 3 ' end of the allele-specific primer is positioned at the single nucleotide polymorphism nucleotide position, and b) analysis of an allele-specific cDNA product from the allele-specific reverse transcription reaction at the remote region of the gene to determine the presence or absence of the mutation on the allele-specific cDNA product. The inventors have discovered that the primer should preferably be shorter than about 20 nucleotides, e.g., about 15 nucleotides, long, because of a possibility that primers which are longer than about 20 nucleotides will not discriminate between the targeted SNP variants. In a layman's terms, the practitioner takes RNA from the patient and applies a reverse transcription primer that recognizes just the "A allele." The "A allele" specific primer will have at its 3' position a complement to the SNP variant of interest. In case of the A-variant , the "A allele" specific primer will have the T at the 3' end, and so when this "A allele" specific primer anneals to the mRNA, it will base-pair with the 3' end and allow the reverse transcriptase to proceed to synthesize the cDNA from the "A allele." Conversely, the A allele" primer will not base- pair at the 3' end of the primer with the "C allele" (since T is not complementary to C) . Thus, the reverse transcription polymerase will not be able to produce cDNA from the C allele. On the other hand, in the "A" portion of a reaction, the practitioner will obtain a pool of the cDNAs that corresponds to the "A allele." The reaction can be repeated in a separate tube with a "C allele" specific primer and no "A allele" primer. A person of ordinary skill in the art will understand that the UC allele" specific primer will have a G on its 3' end. Essentially the practitioner will perform at least one allele-specific reverse transcription reaction, but preferably two allele- specific reverse transcriptions reactions (each with its own allele-specific primer), on the mRNA from the patient. As a result, the practitioner will have two sub-populations of cDNA, wherein each subpopulation is allele-specific, and the practitioner knows which pool corresponds to which variant. Thus, the practitioner will be able to use any number of possible methods, the simplest being PCR to analyze the upstream portion of the cDNA containing the CAG repeat region and quantify the number of the repeats from the cDNA products that came specifically from the "C reaction" or specifically from the "A reaction."
The embodiment of the invention described above employs the notion that a mismatch on the 3' position of the allele- specific primer will not allow reverse transcriptase to produce cDNA from the allele with a mismatched SNP variant . A person of ordinary skill in the art will undoubtedly recognize that the 3' end of the allele- specific primer does not have to be positioned at the single nucleotide polymorphism nucleotide position. For example, a skilled artisan may design primers and conditions of the reverse transcription reaction in such a way that the allele- specific primer will not bind altogether and thus lead to the same end result: absence of cDNA the allele with a mismatched SNP variant.
The accurate determination of the number of CAG repeats is required for the DNA-based predictive testing of at-risk individuals. To date, CAG repeat length determination is based on polymerase chain reaction (PCR) amplification of genomic DNA using primers flanking the CAG repeat region in the IT15 gene, and subsequent electrophoretic separation of the products in denaturing polyacrylamide gels (Williams efc al. , (1999) Comparative semi -automated analysis of (CAG) repeats in the HD gene: use of internal standards. MoI. Cell Probes, 13:283-289).
Numerous methods and commercial kits for the synthesis of first strand cDNA molecules are well known in the art.
TM
Examples include the Superscript Double Strand cDNA
TM Synthesis Kit (Invitrogen, Carlsbad, CA) , the Array 50 ,
TM TM
Array 350 and Array 900 Detectxon Kits (Genxsphere,
Hatfield, PA) , and the CyScribe Post-Labelling Kit
(Amersham, Piscataway, NJ). RNA molecules (e.g., mRNA, hnRNA, rRNA, tRNA, miRNA, snoRNA, non-coding RNAs) from a source of interest are used as templates in a reverse transcription reaction. The RNA may be obtained from a mammalian or more preferably human tissue or cell source. The methods of the present invention are particularly suited for amplification of RNA from small numbers of cells, including single cells, which can be purified from complex cellular samples using, e.g., micromanipulation, fluorescence-activated cell sorting (FACS) and laser microdissection techniques (see Player et al . , Expert Rev. MoI. Diagn. 4:831 (2004)).
Any reverse transcriptase can be used in the initial reverse transcription reaction, including thermostable, RNAse H+ and RNase H~ reverse transcriptases. Preferably, an RNase H' reverse trancriptase is used- Primers for first strand cDNA synthesis can be obtained commercially or synthesized and purified using techniques well known in the art. As disclosed above, the inventors discovered that primers which are about 15 nucleotides long provide the best results in terms of discriminating between the SNP variants and efficiency in producing the allele- specific cDNA.
PCR amplifications of the CAG repeat region have primarily been performed by incorporating [α-32P] dNTPs, or using 32P or fluorescently end- labeled primers. Sizing of fluorescently end-labeled amplification products was performed in various Applied Biosysterns DNA sequencers
(Andrew et al . , (1993) Nature Genet., 4:398-403; Choudhry et al., (2001) Hum. MoI. Genet., 10:2437-2446; Ishii et al . ,
(2001) J. Clin. Endocrinol. Metab. , 86:5372-5378; Le et al . ,
(1997) MoI. Pathol., 50:261-265; Mangiarini et al . , (1997)
Nature Genet., 15:197-200; Pelotti et al . , (2001) Am. J.
Forensic Med. Pathol., 22:55-57; Wallerand et al . , (2001) Fertil. Steril., 76:769-774; Warner et al . , (1993) MoI. Cell
Probes, 1 -.233-239 ; and Warner et al . , (1996) J. Med. Genet.,
33 :1022-1026) . High-resolution method can be used for the exact length determination of CAG repeats in HD genes as well as in genes affected in related CAG repeat disorders (Elisabeth Mόncke- Buchner et al. , Nucleic Acids Res. 2002 August 15; 30(16)). A wide variety of kits may be prepared according to present invention. For example, a kit may include a single stranded promoter template comprising at least one RNA polymerase recognition sequence; and instructional materials for synthesizing cDNA molecules using said promoter template. While the instructional materials typically comprise written or printed materials, they are not limited to such. Any medium capable of storing such instructions and communicating them to an end user is contemplated by this invention. Such media include, but are not limited to, electronic storage media (e.g., magnetic discs, tapes, cartridges, chips), optical media (e.g., CD ROM), and the like. Such media may include addresses to internet sites that provide such instructional materials.
The kits of the present invention may further include one or more of the following components or reagents: a reverse transcriptase (preferably with DNA-dependent DNA polymerase activity) ; an RNase inhibitor; an enzyme for attaching a 3' oligodeoxynucleotide tail onto DNA molecules
(e.g., terminal deoxynuσleotidyl transferase); an enzyme for degrading RNA in RNA/DNA duplexes (e.g., RNase H); and one or more RNA polymerases (e.g., T7, T3 or SP6 RNA polymerase). Additionally, the kits may include buffers, primers (e.g., oligodT primers, random primers), nucleotides, labeled nucleotides, an RNase inhibitor, polyA polymerase, RNase-free water, containers, vials, reaction tubes, and the like compatible with the synthesis of sRNA molecules according to the methods of the present invention. The components and reagents may be provided in containers with suitable storage. media.
A person of ordinary skill in the art will appreciate that such allele-specific diagnosis empowers a practitioner to devise and implement an allele-specific treatment which generally comprises inactivation of the mutated copy of the gene. It is known that patients are able to survive and live healthy lives with only one functioning copy of the HD gene. It is known that the expression of the mutant gene is causing the trouble for the HD patient. Applicants' therapeutic model provides for selectively shutting off mutant gene expression without affecting expression of the normal gene, and is applicable to any disease which contains an SNP variant of an allele in a heterozygous subject that is on the same mRNA transcript as a disease-causing mutation that is at a remote region of the gene's mRNA.
Accordingly, another aspect of the present invention provides a method of treating a patient susceptible to Huntington's disease comprising: a) determining which single nucleotide polymorphism variant is on the same mRNA transcript as a disease-causing mutation according to an allele-specific reverse transcription reaction using an allele-specific primer which recognizes one single nucleotide polymorphism variant, wherein further the 3' end of the primer is positioned at the single nucleotide polymorphism nucleotide position, and b) analysis of the resulting cDNA product from the reverse transcription reaction at the region of the mutation to determine ' the presence or absence of the mutation on this allele-specific cDNA product, and c) applying an allele-specific therapy to the SNP variant. In one embodiment, the allele-specific therapy comprises an RNA molecule comprising a double- stranded portion, wherein the single nucleotide polymorphism site is located within seven nucleotides from an end of the double stranded portion. In one embodiment, the double stranded portion is between 15 and 23 nucleotides long, e.g., about 19 nucleotides long. Further, as discussed above, the siRNA molecule may contain a loop (e.g., shRNA) , a 3' overhand or a 5' overhang which are outside of the double-stranded portion. The instant invention also provides a method of allele-specific therapy, wherein the double stranded portion does not contain a mismatch in a position adjacent to the single nucleotide polymorphism site. Thus, in one embodiment of the invention, the siRNA molecule does not contain any mismatches and one strand of the double-stranded portion is 100% identical to the portion of the targeted mRNA transcript. In one embodiment, wherein the disease treated by the allele-specific therapy is Huntington's disease, the non-limiting example of a single nucleotide polymorphism site suitable for the allele- specific therapy is rs262125. It should be noted that the allele-specific therapy could itself operate at a different SNP site than the SNP site used to make the determination about which allele contains the mutation, so long as the SNP site of the therapy target and the SNP site used to identify the mutation-containing allele are already determined, before the therapy is administered to the patient, to be linked; that is, on the same mRNA transcript.
In some embodiments of the present invention the allele-specific therapy comprises allele-specific RNA interference using siRNA or shRNA. In this embodiment of the invention, the allele-specific therapy destroys the "A allele" of the patient. In this embodiment the siRNA targets the "A allele" upon introduction into the subject's brain by any method known to those of skill in the art (See for example, U.S. Application No. 11/253,393, U.S. Application No. 10/852,997, U.S. Application No. 10/721,693, U.S. Application No. 11/157,608, and PCT Patent Application No. US05/022156, which are incorporated herein in their entirety) . When the siRNA is delivered into a cell it is used by proteins in the cell (known as the RISC complex) to find and destroy the tnRNA from the Huntington' s gene that has the "A allele." Thus, the messenger RNA is destroyed before it can be used to make protein. Conversely, the allele that came from the healthy parent does not get destroyed and so its messenger RNA still survives to be used to make functional biologically active protein. The design and use of small interfering RNA complementary to mRNA targets that produce particular proteins is a recent tool employed by molecular biologists to prevent translation of specific mRNAs. Various groups have been recently studying the effectiveness of siRNAs as biologically active agents for suppressing the expression of specific proteins involved in neurological disorders. Caplen et al . Human Molecular Genetics, 11(2): 175-184 (2002) assessed a variety of different double stranded RNAs for their ability to inhibit cell expression of mRNA transcripts of the human androgen receptor gene containing different CAG repeats. Their work found gene-specific inhibition occurred with double stranded RNAs containing CAG repeats only when flanking sequences to the CAG repeats were present in the double stranded RNAs. They were also able to show that constructed double stranded RNAs were able to rescue caspase-3 activation induced by expression of a protein with an expanded polyglutamine region. Xia, Mao, efc al., Nature Biotechnology, 20: 1006-1010 (2002) demonstrated the inhibition of polyglutamine (CAG) expression in engineered neural PC12 clonal cell lines that express a fused polyglutamine- fluorescent protein using constructed recombinant adenovirus expressing siRNAs targeting the tnRNA encoding green fluorescent protein.
One aspect of the present invention provides an siRNA molecule corresponding to at least a portion of a gene containing an SNP variant of an allele in a heterozygous subject that is on the same mRNA transcript as a disease- causing mutation located at a remote region of the gene's mRNA, wherein such siRNA nucleic acid sequence is capable of inhibiting expression of the mRNA transcript containing the disease-causing mutation in a cell. siRNAs are typically short (19-29 nucleotides) , double-stranded RNA molecules that cause sequence- specific degradation of complementary target mRNA known as RNA interference (RNAi) . Bass, Nature 411:428 (2001) .
Accordingly, in some embodiments, the siRNA molecules comprise a double-stranded structure comprising a sense strand and an antisense strand, wherein the antisense strand comprises a nucleotide sequence that is complementary to at least a portion of a desired nucleic acid sequence and the sense strand comprises a nucleotide sequence that is complementary to at least a portion of the nucleotide sequence of said antisense region, and wherein the sense strand and the antisense strand each comprise about 19-29 nucleotides .
Any desired nucleic acid sequence can be targeted by the siRNA molecules of the present invention. Nucleic acid sequences encoding desired gene targets are publicly available from Genbank. The siRNA molecules targeted to desired sequence can be designed based on criteria well known in the art (e.g., Elbashir et al.t EMBO J. 20:6877 (2001)). For example, the target segment of the target mRNA preferably should begin with AA (most preferred) , TA, GA, or CA; the GC ratio of the siRNA molecule preferably should be 45-55%; the siRNA molecule preferably should not contain three of the same nucleotides in a row; the siRNA molecule preferably should not contain seven mixed G/Cs in a row,- the siRNA molecule preferably should comprise two nucleotide overhangs
(preferably TT) at each 3' terminus; the target segment preferably should be in the ORF region of the target mRNA and preferably should be at least 75 bp after the initiation
ATG and at least 75 bp before the stop codon; and the target segment preferably should not contain more than 16-17 contiguous base pairs of homology to other coding sequences.
Based on some or all of these criteria, siRNA molecules targeted to desired sequences can be designed by one of skill in the art using the aforementioned criteria or other known criteria (e.g., Gilmore et al . , J. Drug Targeting 12:315 (2004); Reynolds et al . , Nature Biσtechnol . 22:326 (2004); Ui-Tei et al . , Nucleic Acids Res. 32:936 (2004)). Such criteria are available in various web-based program formats useful for designing and optimizing siRNA molecules (e.g., siDESIGN Center at Dharmacon; BLOCK-iT RNAi Designer at Invitrogen; siRNA Selector at Wistar Insitute; siRNA Selection Program at Whitehead Institute; siRNA Design at Integrated DNA Technologies; siRNA Target Finder at Ambion; and siRNA Target Finder at Genscript) . siRNA molecules targeted to desired sequences can be produced in vitro by annealing two complementary single- stranded RNA molecules together (one of which matches at least a portion of a desired nucleic acid sequence) (e.g.,
U.S. Pat. No. 6,506,559) or through the use of a short hairpin RNA (shRNA) molecule which folds back on itself to produce the requisite double-stranded portion (Yu et al . , Proc. Natl. Acad. Sci. USA 99:6047 (2002)). Such single-stranded RNA molecules can be chemically synthesized
(e.g., Elbashir et al . , Nature 411:494 (2001)) or produced by in vitro transcription using DNA templates (e.g., Yu et al., Proc. Natl. Acad. Sci. USA 99:6047 (2002)). When chemically synthesized, chemical modifications can be introduced into the siRNA molecules to improve biological stability. Such modifications include phosphorothioate linkages, fluorine-derivatized nucleotides, deoxynucleotide overhangs, 2 ' -O-methylation, 2 ' -ø-allylation, and locked nucleic acid (LNA) substitutions (Dorset and Tuschl, Nat. Rev. Drug Discov. 3:318 (2004); Gilmσre et al . , J. Drug Targeting 12:315 (2004)). siRNA molecules targeted to desired target sequences can be introduced into cells to inhibit expression. Alternatively, DNA molecules from which shRNA molecules targeted to desired target sequences can be introduced into cells to inhibit expression. Accordingly, another aspect of the present invention provides for inhibiting expression of an mRNA sequence containing an SNP allele and a disease- causing mutation in a cell comprising introducing into a cell at least one siRNA molecule or shRNA molecule that corresponds to at least a portion of the mRNA nucleic acid sequence. Any cell can be targeted. For example, the siRNA or shRNA molecules are introduced into a heart cell or brain cell. In some embodiments, the brain cell is from a subject at risk for HD, i.e., the offspring of a HD patient. The siRNA molecules produced herein can be introduced into cells in vitro or ex vivo using techniques well-known in the art, including electroporation, calcium phosphate co-precipitation, microinjection, lipofection, polyfection, and conjugation to cell penetrating peptides (CPPs) . The siRNA molecules can also be introduced into cells in vivo by- direct delivery into specific organs such as the liver, brain, eye, lung and heart, or systemic delivery into the blood stream or nasal passage using naked siRNA molecules or siRNA molecules encapsulated in biodegradable polymer microspheres (Gilmore et al . , ι7. Drug Targeting 12:315 (2004) ) .
Alternatively, siRNA molecules targeted to specific mRNA sequences can be introduced into cells in vivo by endogenous production from an expression vector (s) encoding the sense and antisense siRNA sequences. Accordingly, another aspect of the present invention provides an expression vector comprising at least one DNA sequence encoding a siRNA molecule corresponding to at least a portion of a specific mRNA nucleic acid sequence capable of inhibiting expression of a specific mRNA in a cell operably linked to a genetic control element capable of directing expression of the siRNA molecule in a cell. Expression vectors can be transfeeted into cells using any of the methods described above.
Genetic control elements include a transcriptional promoter, and may also include transcription enhancers to elevate the level of mRNA expression, a sequence that encodes a suitable ribosome binding site, and sequences that terminate transcription. Suitable eukaryotic promoters include constitutive RNA polymerase II promoters (e.g., cytomegalovirus (CMV) promoter, the SV40 early promoter region, the promoter contained in the 3 ' long terminal repeat of Rous sarcoma virus (RSV) , the herpes thymidine kinase (TK) promoter, and the chicken beta-actin promoter) , cardiaσ-tissue-specific RNA polymerase II promoters (e.g., the ventricular myosin light chain 2 (MLC-2v) promoter, and the sodium-calcium exchanger gene Hl promoter (NCXlHl) ) , and RNA polymerase III promoters (e.g., U6, Hl, 7SK and 7SL) .
In some embodiments, the sense and antisense strands of siRNA molecules are encoded by different expression vectors (i.e., cotransfected) (e.g., Yu et al . , Proc. Natl. Acad. Sci. USA 99:6047 (2002). In other embodiments, the sense and antisense strands of siRNA molecules are encoded by the same expression vector. The sense and antisense strands can be expressed separately from a single expression vector, using either convergent or divergent transcription (e.g., Wang et al . , Proc. Natl. Acad. Sci. USA 100:5103 (2003); Tran et al., BMC Biotechnol. 3:21 (2003)). Alternatively, the sense and antisense strands can be expressed together from a single expression vector in the form of a single hairpin RNA molecule, either as a short hairpin RNA (shRNA) molecule (e.g., Arts et al . , Genome Res. 13:2325 (2003)) or a long hairpin RNA molecule (e.g., Paddison et al . , Proc. Natl. Acad. Sex. USA 99:1443 (2002)).
Although numerous expression vectors can be used to express siRNA molecules in cells (Dorsett and Tuschl, Nat. Rev. Drug Discov.3:318 (2004)), viral expression vectors are preferred, particularly those that efficiently transduce heart cells (e.g., alphaviral, lentiviral, retroviral, adenoviral, adeno-associated viral (AAV)) (Williams and Koch, Annu. Rev. Physiol. 66:49 (2004); del Monte and Hajjar, J. Physiol. 546.1:49 (2003). Both adenoviral and AAV vectors have been shown to be effective at delivering transgenes (including transgenes directed to deseases) into heart, including failing cardiomycoytes (e.g., Iwanaga et al., J. Clin. Invest. 113:727 (2004); Seth et al . , Proc.
Natl. Acad. Sci. USA 101:16683 (2004); Champion et al . , Circulation 108:2790 (2003); Li et al . , Gene Ther. 10:1807
(2003); Vassalli et al . , Int. J. Cardiol. 90:229 (2003); del
Monte et al . , Circulation 105:904 (2002); Hoshijima et al.,
Nat. Med. 8:864 (2002); Eizema et al . , Circulation 101:2193
(2000); Miyamoto et al . , Proc. Natl. Acad. Sci. USA 97:793 (2000); He et al . , Circulation 100:974 (1999). Recent reports have demonstrated the use of AAV vectors for sustained gene expression in mouse and hamster myocardium and arteries for over one year (Li et al., Gene Ther. 10:1807 (2003); Vassalli et al . , Int. J. Cardiol. 90:229 (2003)) . In particular, expression vectors based on AAV serotype 6 have been shown to efficiently transduce both skeletal and cardiac muscle (e.g., Blankinship et al . , MoI . Ther. 10:671 (2004)). The present invention also provides for the use of coxsackie viral vectors for delivery of desired siRNA sequences.
Following introduction of the desired siRNA molecules into cells, changes in desired gene product levels can be measured if desired. Desired gene products include, for example, desired mRNA and desired polypeptide, and both can be measured using methods well-known to those skilled in the art. For example, desired mRNA can be directly detected and quantified using, e.g., Northern hybridization, in situ hybridization, dot and slot blots, or oligonucleotide arrays, or can be amplified before detection and quantitation using, e.g., polymerase chain reaction (PCR), reverse-transcription-PCR (RT-PCR) , PCR-enzyme-linked immunosorbent assay (PCR-ELISA) , or ligase chain reaction (LCR) .
Desired polypeptide (or fragments thereof) can be detected and quantified using various well-known immunological assays, such as, e.g., enzyme-linked immunosorbent assay (ELISA) , radioimmunoassay (RIA) , immunoprecipitation, immunofluorescence, and Western blotting. Anti-deaired antibodies (preferably anti-human desired) for use in immunological assays are commercially available from, e.g., EMD Biosciences (San Diego, CA),
Upstate (Charlottesville, VA), Abeam (Cambridge, MA),
Affinity Bioreagents (Golden, CO) and Novus Biologicals
(Littleton, CO) , or may be produced by methods well-known to those skilled in the art . Specific embodiments according to the methods of the present invention will now be described in the following examples. The examples are illustrative only, and are not intended to limit the remainder of the disclosure in any way.
EXAMPLES Example 1
RNA-isolation and Rβversθ transcription reaction. Applicants analyzed . the CAG-repeat sequences in the Huntington's disease gene using the following allele- specific reverse transcription reaction. Table 2 defines the sequences for various allele-specific reverse transcription primers for use in determining which allele of a heterozygous patient's Huntington's disease gene contains the disease-causing allele, in accordance with the subject invention. The subject SNP sites in the Huntington's disease gene (IT15) are designated using the identification number provided by the National Center for Biotechnology- Information (NCBI) database, accessible at: http : //www .ncbi . nlm. nih. gov/entrez/query. fcgi?db=snp .
Table 2
Figure imgf000031_0001
Figure imgf000032_0001
Cell culture and genotyp±ng.
Fibroblasts purchased from Coriell Cell Repositories
(Camden, NJ) were cultured at 37°C with 5% CO2 and maximum humidity. The growth medium was minimum essential medium containing 20% FBS, 1% PSN antibiotics, 1% fungizone, 2% non-essential amino acids, 2% amino acids and 2% vitamins
(all reagents from Invitrogen) . Genomic DNA was isolated using the DNeasy kit (Qiagen) and nucleotide identity for HD alleles at various SNP positions was determined by pyrosequencing conducted by Isogen Lifescience (The
Netherlands) .
Identification of a heterozygous HD individual. Fibroblasts from 21 individuals from different kindreds, diagnosed with HD or known to be at risk for HD, were purchased from Coriell Cell Repositories (Camden, NJ) . DNA was harvested from each fibroblast culture and pyrosequenced to determine the nucleotide identity at eleven known single nucleotide polymorphism (SNP) sites within the protein coding region of the HD sequence. Heterozygosity was found at four of the eleven SNP sites tested. Ten of the 21 genotyped fibroblast cultures (see Table 3) were heterozygous for at least 1 SNP site. The results showed that the donor of GM04022 fibroblasts is heterozygous at SNP position rs363125 (NCBI) , harboring both an A-allele (adenine) and a C-allele (cytosine) at nucleotide position
5310 in the HD reference sequence NM_002111.6. The fibroblasts from this donor (here called donor-1) were selected for further study. These fibroblasts are from a female Caucasian donor who was 28 years old at the time of cell collection. Her mother, who was 58 years old at the time of the cell collection, was reportedly diagnosed with HD at age 49.
Figure imgf000034_0001
Development of allele-speciiric reverse transcription .
As described above, donor-1 was determined to be heterozygous (adenine versus cytosine) at SNP site rs363125. In order to design an allele-specific RNA interference-based therapy for donor-1, it needs to be determined which of these SNP sequences is associated with the expanded CAG repeat mutation that is located approximately 5000 nucleotides upstream from the SNP position. Appropriate siRNA or shRNA targeting SNP site rs363125 should only reduce protein expression from the allele that contains the expanded CAG repeat and should therefore only be specific for the associated SNP. Correspondence between the SNP identity and the expanded allele cannot be readily determined by cDNA sequencing or by comparing the lengths of PCR products spanning the SNP position and the CAG repeat region. To solve this problem, the inventors developed a strategy that uses SNP-specific reverse transcription (RT) primers to selectively generate cDNA from only one allelic species of HD mRNA. The primers each contained either guanine or thymine at the 3' terminal position, corresponding to SNP site rs363125, as shown in FIG. 1. Primers of various lengths were tested. The 3 '-guanine and 3' -thymine primer were each designed to specifically facilitate reverse transcription (also known as first strand synthesis) of only the mRNA containing cytosine or adenine at the SNP position,- respectively. Following reverse transcription with either type of RT primer, PCR was conducted on the resulting cDNA using PCR primers that amplify a product spanning the CAG-repeat sequence in the first exon of the HD gene. RNA isolated from the fibroblasts was reverse transcribed using the Superscript III RT kit (Invitrogen) in the presence of 100 nM of one of the following DNA primers: the 20-mer 5'-GTGTTCTTCTAGCGTTGAAT-S', SEQ ID NO: 52 (or a shorter, corresponding 15-mer or 10-mer ending in T-3', corresponding to SEQ ID NO: 47 and SEQ ID NO: 42, respectively) or the 20-mer primer δ'-GTGTTCTTCTAGCGTTGAAG- 3' SEQ ID NO: 64 (or a shorter, corresponding 15-mer or 10- mer primer ending in G-3', corresponding to SEQ ID NO: 59 and SEQ ID NO: 54, respectively) at 100 nM . The CAG repeat sequence on either RT product was then amplified by PCR (Bio-Rad iCycler) using Accuprime GC-Rxch DNA-polymerase (invitrogen) , and forward primer 5'-GCCTTCGAGTCCCTCAAGT -3' and reverse primer 5'-GACAATGATTCACACGGTCT-S' at 0.2 μM each (SEQ ID NO: 85 and SEQ ID NO: 86, respectively) . The resulting PCR products contain the complete CAG repeat sequence of one of the two alleles of the GM04022 cells. CAG-repeat size for each allelic RT products was determined using standard 1.5% agarose gel electrophoresis with ethidium bromide staining, and also by sequencing of the products of the PCR amplification of the CAG repeat region.
Gel electrophoresis of the respective PCR products (FIG. 2) showed that the RT-PCR assay using the 3' -thymine primer in the RT step yielded a product of approximately 300 base pairs. In contrast, the RT-PCR assay using the 3'- guanine primer in the RT step yielded a PCR product of approximately 360 base pairs. RT primers that were 15 bases in length yielded cDNA from which a PCR product corresponding to only one allele from heterozygous donor-1 was preferentially amplified. Allele specific RT primers of different lengths were also designed and tested for SNP1936032 and SNP362331, both of which were also determined to be heterozygous in donor-1 (Table 3). These experiments showed that primers of 15 bases in length were also optimal for allele-specifiσ transcription from the respective SNP sites. CorrespσndencB between a SNP identity and an individual' s expanded allele.
Based on the relative sizes of the PCR products in
Figure 2, it may be determined that the CAG-repeat region on the allele containing cytosine at SNP rs363125 contains approximately 20 more repeats than the allele containing adenine at that SNP, with a total CAG-repeat length of about
40 repeats. Sequencing of the PCR products confirmed that the CAG repeat region of the donor-1 allele containing cytosine at SNP position rs363125 contains 44 versus 19 CAG codons, respectively. Therefore, the allele containing cytosine at SNP position rs363125 is the disease-causing allele, and an appropriate allele-specific therapy for donor-1 would deliver an siRNA or shRNA providing the sequence specified here as siRNA 363125_C~16.
Example 2
Development of SNP-specific real time PCR assays. In order to determine whether allele-specific suppression of HD mRNA is occurring in cells, it is necessary to be able to quantify the amount of HD mRNA corresponding to each allele individually. Molecular beacons are synthetic oligonucleotide probes that have a fluorophore and a quencher covalently linked to the respective ends of the σligo. In solution, the beacon adopts a hairpin conformation, causing the fluorophore to be quenched. However, upon hybridization with complementary DNA in a PCR reaction, the hairpin conformation is lost and fluorescence from the fluorophore can be detected. Beacons can be constructed such that as little as a single nucleotide mismatch between the beacon and the complementary
DNA is sufficient for the probe to be more stable in its self-annealed state than in the probe -cDNA hybrid. The inventors designed two such beacons corresponding to the two allelic variants of SNP rs363125 (Fig. 3) . Absence of genomic DNA carry over in the RNA isolates was confirmed by PCR using forward primer S'-CTCGCCTCCAGCATGAAAGT-S' and reverse primer S'-GTGACTGGGGCATTGATT-B', (SEQ ID NOS: 87 and 88, respectively) which amplify monocyte chemo-attractant protein-1 (MCP-I) genomic DNA. Genomic DNA from HeLa cells served as a positive control for the PCR reaction, and only total RNA samples from fibroblasts verified to be without detectable genomic DNA were used in further analyses . Concentration and integrity of the isolated RNA were checked by Experion automated electrophoresis (Bio-Rad, StdSens analysis kit) of the 28S/18S ribosomal RNA. RNA was reverse transcribed into cDNA using the iScript cDNA synthesis kit (BioRad) , after which allele-specific real time PCR was conducted using the molecular beacon method. The sequences of the molecular beacons are described in Figure 3 and correspond to SEQ ID NOs: 89 (3 '-BHQI- tcacgcaGATCGCAACTTAATGACAGGGtgcgtga-FAM-51 ) and 90 (3 '-BHQI- gcgctagAGATCGCAACTTCATGACAGGGGTAGctagcgc- FAM-5'), for the "A" and the "C" alleles, respectively. In these sequences, the "loop sequence" which binds to a portion of the targeted portion of the PCR product is in capital letters, while the "stem" portions, which form the double strand of the beacon if the beacon is not bound to the targeted portion of the PCR product. The sequences of the primers for the "A" allele correspond to SEQ ID NOs: 91 <5' CCTTCTCTCCGTATTTAATCTCCTGTA-3 ' ) and 92 (51 TCATTTCCACCTTCAGCTGTTTTGTAA-3 •) for the forward and the reverse primers, respectively. The sequences of the primers for the "C" allele correspond to SEQ ID NOs 93 (51 AGATATTGTTCTTTCTCGTATTCAGG-S ') and 94 (51 TGCTCACTCATTTCCACCTTC-3 ' ) for the forward and the reverse primers, respectively. Detection of the PCR product was conducted using molecular beacons (Proligo, France) each of which detects only a PCR product containing a complementary nucleotide at the SNP position. The two alleles were quantified in separate PCR reactions, each containing 0.3 μM of the appropriate molecular beacon. During elongation, increase of the FAM fluorescence signal was recorded. The data were quantified against a standard curve generated using a serial dilution of a plasmid containing the PCR product (Baseclear genesynthesis , The Netherlands). Results were normalized against GAPDH, amplified using IQ SYBR Green Supermix (Bio-Rad) with forward primer 5'- ACTCCTCCACCTTTGACGC-3' (SEQ ID NO: 95) and reverse primer 5' -GTTGCTGTAGCCAAATTCGTT-3 ' (SEQ ID NO : 96).
As shown in Figure 4, these two beacons are specific for their respective SNP sequences in real time PCR reactions. In these PCR reactions plasmids were used as template DNA that were engineered and verified to contain one or the other allelic variant of the target sequence. Both beacon MB363125_A and MB363125_C were found to yield a PCR amplification signal only from their corresponding DNA template .
Allele-specific suppression of huntingtin mRNA. Fibroblasts were cultured in 25cm2 culture flasks (Nunc) as described, but without the addition of PSN antibiotics and Fungizone. Lipofectamine 2000 (Invitrogen) was used to conduct siRNA transfection at three different conditions: 1) mock transfection (n=8) ; 2) transfection with scrambled siRNA (Ambion) (n=4) ; and 3) transfection with siRNA sequence 5'-GAAGUACUGUCCCCAUCUCdTdT-S', SEQ ID NO:. 97, (Ambion) (n=7) at a concentration of 100 nM. The latter siRNA has a guanine located at position 16 relative to the 3' end of the complementary region of the target huntingtin mRNA, providing specificity for the allele containing cytosine at the SNP position. A parallel cell culture was transfected with a fluorescently labeled Block-It siRNA (Invitrogen) to verify efficiency of Lipofectamine 2000 transfection . The cells were incubated overnight at 37°c, 5% CO2 and maximum humidity. The cultures were then washed with PBS and fluorescence microscopy (Leica DM-IRB) was used to confirm transfection in the Block-It transfected cultures, which were considered representative for all transfection conditions. The cells were cultured for another day before RNA isolation as described, approximately 48 hours post- transfection. Fibroblasts from donor-1 were transfected with siRNA designed to specifically target the mRNA containing cytosine at SNP site rs363125. This siRNA molecule (siRNA 363125_C- 16) was designed such that the cytosine nucleotide of the SNP is located at position 16 relative to the 5' end of the sense strand of the siRNA molecule. The amount of mRNA from both endogenous alleles was separately quantified using the molecular beacons developed to be specific for the allelic variants at this SNP site. The results showed that about 48 hours following treatment of the fibroblasts with siRNA 363125_C-16, mRNA transcripts containing cytosine at position rs363125 were detected at levels approximately 80% lower (p < 0.01, two-tailed) from that detected in controls that were mock transfected, or transfected with a scrambled siRNA (Fig. 5; ANOVA of treatment by allele interaction effect, p = 0.0002) . Transcripts containing adenine at position rs363125 were present at levels statistically not different from (p = 0.5145) and numerically similar to the controls .
All publications cited in the specification, both patent publications and non-patent publications, are indicative of the level of skill of those skilled in the art to which this invention pertains. All these publications are herein fully incorporated by reference to the same extent as if each individual publication were specifically and individually indicated as being incorporated by reference.

Claims

1. A method for determining which single nucleotide polymorphism variant of an allele from a gene isolated from a heterozygous patient is on the same mRNA transcript as a disease-causing mutation at a remote region of the gene's mRNA comprising: a) an allele-specific reverse transcription reaction using an allele-specific primer which recognizes one single nucleotide polymorphism variant, and b) analysis of an allele-specific cDNA product from the allele-specific reverse transcription reaction at the remote region of the gene to determine the presence or absence of the mutation on the allele-specific cDNA product, wherein the allele-specific primer is shorter than about 20 nucleotides .
2. The method of claim 1, wherein the 3' end of the allele-specific primer is positioned at the single nucleotide polymorphism nucleotide position.
3. A method of claim 1 or 2 further comprising: c) at least a second allele-specific reverse transcription reaction using at least a second allele-specific primer which recognizes at least a second single nucleotide polymorphism variant, and d) analysis of at least a second allele-specific cDNA product from at least the second allele-specific reverse transcription reaction at the remote region of the gene to determine the presence or absence of the mutation on at least the second allele-specific cDNA product.
4. The method of claim 3 wherein the 3' end of at least the second allele-specific primer is positioned at the single nucleotide polymorphism nucleotide position.
5. A method of treating a patient comprising: a) determining which single nucleotide polymorphism variant is on the same mRNA transcript as a disease causing mutation according to the method of claim 1, .and b) applying an allele-specific therapy to the single nucleotide polymorphism variant.
6. The method of claim 5 wherein the allele-specific therapy comprises an RNA molecule comprising a double-stranded portion, and the single nucleotide polymorphism site is located within seven nucleotides from an end of the double stranded portion.
7. The method of claim 5 or 6, wherein the allele- specific therapy comprises allele-specific RNA interference using siRNA or shRNA.
8. The method of any one of claims 5-7, wherein the patient is at risk of developing a neurodegenerative disorder selected from the group consisting of Dystophia myotonica, Spinocerebellar ataxia type 1, Spinocerebellar ataxia type 2, Spinocerebellar ataxia type 3, Spinocerebellar ataxia type 6, Spinocerebellar ataxia type 7, Spinocerebellar ataxia type 8, Spinocerebellar ataxia type 17, Huntington disease-like 2, Spinal and bulbar muscular atrophy, Huntington' s disease, Dentatorubral-pallidoluysian atrophy, Oculopharyngeal dystrophy, Congenital central hypoventilation syndrome, Infantile spasms, Synpolydactyly, Cleidocranial dysplasia, Holoprosencephaly, Hand-foot-genital syndrome, Type II blephorophimosis, ptosis, and epicanthus inversus syndrome. allele-specific therapy operates at the same single nucleotide polymorphism site used to determine which mRNA transcript contains the disease-causing allele in said patient.
10. The method of any one of claims 5-9, wherein the allele-specific therapy operates at a different single nucleotide polymorphism site than the site used to determine which mRNA transcript contains the disease-causing allele in said patient .
11. The method of any one of claims 5-10, wherein the double stranded portion is between 15 and 23 nucleotides long.
12. The method of any one of claims 5-11, wherein the double stranded portion does not contain a mismatch in a position adjacent to the single nucleotide polymorphism site.
13. The method of any one of claims 5-12, wherein one strand of the double-stranded portion is 100% identical to the portion of the targeted mRNA transcript.
14. The method of any one of claims 8-13, wherein the disease is Huntington' s disease.
15. The method of claim 14, wherein the single nucleotide polymorphism site is rs262125.
16. A kit for determining which single nucleotide polymorphism variant of an allele of a heterozygous patient is on the same mRNA transcript as a disease-causing mutation located at a remote region of the gene's mRNA comprising a) an allele-specific primer which recognizes one single nucleotide polymorphism variant, and b) a set of instructions.
17. The method of claim 16, wherein the allele-specific primer is shorter than about 20 nucleotides.
18. The kit of claim 16 or 17, further comprising: at least a second allele-specific primer which recognizes at least a second single nucleotide polymorphism variant.
19. The kit of any one of claims 16-18, wherein the 3' end of the allele-specific primer is positioned at the single nucleotide polymorphism nucleotide position.
20. The kit of any one of claims 16-19, wherein the 3' end of the allele-specific primer is not positioned at the single nucleotide polymorphism nucleotide position.
21. The kit of any one of claims 18-20, wherein the 3' end of at least the second allele-specific primer is positioned at the single nucleotide polymorphism nucleotide position.
22. The kit of any one of claims 18-20, wherein the 3' end of at least the second allele-specific primer is not positioned at the single nucleotide polymorphism nucleotide position.
23. The kit of any one of claims 16-22, wherein the allele-specific primer is selected from the group consisting Of SEQ. ID. 1 - SEQ. ID. 84.
24. The kit of any one of claims 16-23, wherein at least the second allele-specific primer is selected from the group consisting of SEQ. ID. 11 - SEQ. ID. 20 if the allele-specific primer is selected from the group consisting of SBQ. ID. 1 ~ selected from the group consisting of SEQ. ID. 31 - SEQ, ID. 40 if the allele- specific primer is selected from the group consisting of SEQ. ID. 21 - SEQ. ID. 30; or at least the second allele-specific primer is selected from the group consisting of SEQ. ID. 53 - SEQ. ID. 64 if the allele-specific primer is selected from the group consisting of SEQ. ID. 41 - SEQ. ID. 52; or at least the second allele-specific primer is selected from the group consisting of SEQ. ID. 75 - SEQ. ID. 84 if the allele-specific primer is selected from the group consisting of SEQ. ID. 65 - SEQ. ID. 74.
25. The kit of claim 24, wherein the allele-specific primer is selected from the group consisting of SEQ. ID..42, SEQ. ID. 47, SEQ. ID. 52, SEQ. ID. 54, SEQ. ID. 59, and SEQ. ID. 64.
PCT/US2007/012259 2005-06-28 2007-05-23 Methods and kits for linking polymorphic sequences to expanded repeat mutations WO2007139811A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP07777234A EP2029773A4 (en) 2006-05-24 2007-05-23 Methods and kits for linking polymorphic sequences to expanded repeat mutations
US12/560,178 US9133517B2 (en) 2005-06-28 2009-09-15 Methods and sequences to preferentially suppress expression of mutated huntingtin

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11/439,858 US20080280843A1 (en) 2006-05-24 2006-05-24 Methods and kits for linking polymorphic sequences to expanded repeat mutations
US11/439,858 2006-05-24

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/439,858 Continuation-In-Part US20080280843A1 (en) 2005-06-28 2006-05-24 Methods and kits for linking polymorphic sequences to expanded repeat mutations

Related Child Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/064532 Continuation-In-Part WO2008147887A1 (en) 2005-06-28 2008-05-22 Methods and kits for linking polymorphic sequences to expanded repeat mutations

Publications (1)

Publication Number Publication Date
WO2007139811A1 true WO2007139811A1 (en) 2007-12-06

Family

ID=38778977

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/012259 WO2007139811A1 (en) 2005-06-28 2007-05-23 Methods and kits for linking polymorphic sequences to expanded repeat mutations

Country Status (3)

Country Link
US (1) US20080280843A1 (en)
EP (2) EP2298937A1 (en)
WO (1) WO2007139811A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008147887A1 (en) * 2007-05-23 2008-12-04 Medtronic, Inc. Methods and kits for linking polymorphic sequences to expanded repeat mutations
US7732591B2 (en) 2003-11-25 2010-06-08 Medtronic, Inc. Compositions, devices and methods for treatment of huntington's disease through intracranial delivery of sirna
US7819842B2 (en) 2006-11-21 2010-10-26 Medtronic, Inc. Chronically implantable guide tube for repeated intermittent delivery of materials or fluids to targeted tissue sites
US7829694B2 (en) 2002-11-26 2010-11-09 Medtronic, Inc. Treatment of neurodegenerative disease through intracranial delivery of siRNA
US7902352B2 (en) 2005-05-06 2011-03-08 Medtronic, Inc. Isolated nucleic acid duplex for reducing huntington gene expression
US7988668B2 (en) 2006-11-21 2011-08-02 Medtronic, Inc. Microsyringe for pre-packaged delivery of pharmaceuticals
US8058251B2 (en) 2002-11-26 2011-11-15 Kaemmerer William F Devices, systems and methods for improving memory and/or cognitive function through brain delivery of siRNA
US8119611B2 (en) 2002-11-26 2012-02-21 Medtronic, Inc. Treatment of neurodegenerative disease through intracranial delivery of SIRNA
US8258112B2 (en) 2005-05-06 2012-09-04 Medtronic, Inc Methods and sequences to suppress primate huntington gene Expression
US8324367B2 (en) 2006-11-03 2012-12-04 Medtronic, Inc. Compositions and methods for making therapies delivered by viral vectors reversible for safety and allele-specificity
WO2013160563A1 (en) * 2012-04-25 2013-10-31 Ho Tho Huu Method for competitive allele-specific cdna synthesis and differential amplification of the cdna products
US8957198B2 (en) 2003-02-03 2015-02-17 Medtronic, Inc. Compositions, devices and methods for treatment of Huntington's disease through intracranial delivery of sirna
EP2780475A4 (en) * 2011-11-17 2015-06-17 Clarient Diagnostic Services Inc Method of allele-specific amplification
US9133517B2 (en) 2005-06-28 2015-09-15 Medtronics, Inc. Methods and sequences to preferentially suppress expression of mutated huntingtin
US9375440B2 (en) 2006-11-03 2016-06-28 Medtronic, Inc. Compositions and methods for making therapies delivered by viral vectors reversible for safety and allele-specificity

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117487923A (en) * 2023-12-27 2024-02-02 湖南家辉生物技术有限公司 Application of HABP2 gene mutant as detection target, detection reagent and/or detection kit with HABP2 gene mutant

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030152947A1 (en) * 2001-06-15 2003-08-14 Crossman David C. Methods for detecting and treating the early onset of aging-related conditions
US20050282198A1 (en) * 1997-05-29 2005-12-22 Interleukin Genetics, Inc. Diagnostics and therapeutics for diseases associated with an IL-1 inflammatory haplotype
US20060014165A1 (en) * 2003-07-14 2006-01-19 Decode Genetics Ehf. Methods of diagnosis and treatment for asthma and other respiratory diseases based on haplotype association

Family Cites Families (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683202A (en) * 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4965188A (en) * 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4800159A (en) * 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences
US6013431A (en) * 1990-02-16 2000-01-11 Molecular Tool, Inc. Method for determining specific nucleotide variations by primer extension in the presence of mixture of labeled nucleotides and terminators
WO1992020400A1 (en) * 1991-05-24 1992-11-26 Sumitomo Pharmaceuticals Company, Limited Instrument for applying pharmaceutical to inside of brain
US5236908A (en) * 1991-06-07 1993-08-17 Gensia Pharmaceuticals, Inc. Methods of treating injury to the central nervous system
EP0655090B1 (en) * 1992-04-27 2000-12-27 The Trustees Of Dartmouth College Detection of gene sequences in biological fluids
US5354326A (en) * 1993-01-27 1994-10-11 Medtronic, Inc. Screening cable connector for interface to implanted lead
AU7568094A (en) * 1993-08-12 1995-03-14 Cytotherapeutics, Inc. Improved compositions and methods for the delivery of biologically active molecules using genetically altered cells contained in biocompatible immunoisolatory capsules
AU7676894A (en) * 1993-08-27 1995-03-21 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Convection-enhanced drug delivery
US5624803A (en) * 1993-10-14 1997-04-29 The Regents Of The University Of California In vivo oligonucleotide generator, and methods of testing the binding affinity of triplex forming oligonucleotides derived therefrom
AU1552995A (en) * 1993-12-17 1995-07-03 Spinal Cord Society Method for inducing dna synthesis in neurons
EP0755454B1 (en) * 1994-04-13 2008-02-13 The Rockefeller University Aav-mediated delivery of dna to cells of the nervous system
US6294202B1 (en) * 1994-10-06 2001-09-25 Genzyme Corporation Compositions containing polyanionic polysaccharides and hydrophobic bioabsorbable polymers
US5534350A (en) * 1994-12-28 1996-07-09 Liou; Derlin Powerfree glove and its making method
WO1996033761A1 (en) * 1995-04-28 1996-10-31 Medtronic, Inc. Intraparenchymal infusion catheter system
US7069634B1 (en) * 1995-04-28 2006-07-04 Medtronic, Inc. Method for manufacturing a catheter
US5840059A (en) * 1995-06-07 1998-11-24 Cardiogenesis Corporation Therapeutic and diagnostic agent delivery
US5942455A (en) * 1995-11-14 1999-08-24 Drexel University Synthesis of 312 phases and composites thereof
JPH09268067A (en) * 1996-03-29 1997-10-14 Asahi Glass Co Ltd Production of silicon carbide member
US7189222B2 (en) * 1996-04-30 2007-03-13 Medtronic, Inc. Therapeutic method of treatment of alzheimer's disease
US5735814A (en) * 1996-04-30 1998-04-07 Medtronic, Inc. Techniques of treating neurodegenerative disorders by brain infusion
US5976109A (en) * 1996-04-30 1999-11-02 Medtronic, Inc. Apparatus for drug infusion implanted within a living body
WO2000062828A1 (en) * 1996-04-30 2000-10-26 Medtronic, Inc. Autologous fibrin sealant and method for making the same
CA2257197A1 (en) * 1996-06-03 1997-12-11 Jeffrey Edberg Fc receptor polymorphism
EP0869186A4 (en) * 1996-07-18 2002-04-03 Srl Inc Method for the diagnosis of spinocerebellar ataxia type 2 and primers therefor
AU4266597A (en) * 1996-09-11 1998-04-14 General Hospital Corporation, The Use of a non-mammalian dna virus to express an exogenous gene in a mammalian cell
EP0954612A2 (en) 1996-11-06 1999-11-10 Sequenom, Inc. Dna diagnostics based on mass spectrometry
US5882561A (en) * 1996-11-22 1999-03-16 Drexel University Process for making a dense ceramic workpiece
GB9701684D0 (en) * 1997-01-28 1997-03-19 Smithkline Beecham Plc Novel compounds
US5968059A (en) * 1997-03-06 1999-10-19 Scimed Life Systems, Inc. Transmyocardial revascularization catheter and method
GB9706463D0 (en) * 1997-03-27 1997-05-14 Medical Res Council A model of inflamation in the central nervous system for use in the study of disease
NZ333334A (en) * 1997-04-17 2001-06-29 Frank L Sorgi Delivery system for gene therapy to the brain
US5782892A (en) * 1997-04-25 1998-07-21 Medtronic, Inc. Medical lead adaptor for external medical device
US5931861A (en) * 1997-04-25 1999-08-03 Medtronic, Inc. Medical lead adaptor having rotatable locking clip mechanism
US6042579A (en) * 1997-04-30 2000-03-28 Medtronic, Inc. Techniques for treating neurodegenerative disorders by infusion of nerve growth factors into the brain
US6110459A (en) * 1997-05-28 2000-08-29 Mickle; Donald A. G. Transplants for myocardial scars and methods and cellular preparations
US6231969B1 (en) * 1997-08-11 2001-05-15 Drexel University Corrosion, oxidation and/or wear-resistant coatings
US6187906B1 (en) * 1997-08-11 2001-02-13 Aukland Uniservices Limited Methods to improve neural outcome
US6207425B1 (en) * 1997-09-11 2001-03-27 City Of Hope Bidirectional PCR amplification of specific alleles
US6376471B1 (en) * 1997-10-10 2002-04-23 Johns Hopkins University Gene delivery compositions and methods
US6151525A (en) * 1997-11-07 2000-11-21 Medtronic, Inc. Method and system for myocardial identifier repair
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US6436392B1 (en) * 1998-05-20 2002-08-20 University Of Iowa Research Foundation Adeno-associated virus vectors
EP1080202B1 (en) * 1998-05-27 2006-01-25 Avigen, Inc. Convection-enhanced delivery of aav vectors encoding aadc
US6245884B1 (en) * 1998-10-16 2001-06-12 Vivian Y. H. Hook Secretases related to alzheimer's dementia
US6835194B2 (en) * 1999-03-18 2004-12-28 Durect Corporation Implantable devices and methods for treatment of pain by delivery of fentanyl and fentanyl congeners
US6291243B1 (en) * 1999-04-28 2001-09-18 The Board Of Trustees Of The Leland Stanford Jr. University P element derived vector and methods for its use
GB9928248D0 (en) * 1999-12-01 2000-01-26 Gill Steven S An implantable guide tube for neurosurgery
US6310048B1 (en) * 1999-12-09 2001-10-30 St. Louis University Antisense modulation of amyloid beta protein expression
US6461989B1 (en) * 1999-12-22 2002-10-08 Drexel University Process for forming 312 phase materials and process for sintering the same
US20030092003A1 (en) * 1999-12-29 2003-05-15 Ribozyme Pharmaceuticals, Inc. Method and reagent for the treatment of Alzheimer's disease
US20050032733A1 (en) * 2001-05-18 2005-02-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SiNA)
US6632671B2 (en) * 2000-02-28 2003-10-14 Genesegues, Inc. Nanoparticle encapsulation system and method
US6468524B1 (en) * 2000-03-22 2002-10-22 The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services AAV4 vector and uses thereof
US6551290B1 (en) * 2000-03-31 2003-04-22 Medtronic, Inc. Catheter for target specific drug delivery
US6945969B1 (en) * 2000-03-31 2005-09-20 Medtronic, Inc. Catheter for target specific drug delivery
US6372250B1 (en) * 2000-04-25 2002-04-16 The Regents Of The University Of California Non-invasive gene targeting to the brain
US20030039964A1 (en) * 2000-05-09 2003-02-27 Giffard Philip Morrison Method
US20020042388A1 (en) * 2001-05-01 2002-04-11 Cooper Mark J. Lyophilizable and enhanced compacted nucleic acids
US20020090622A1 (en) * 2000-08-23 2002-07-11 Monisola Adeokun Chemical compounds
US20050209179A1 (en) * 2000-08-30 2005-09-22 Sirna Therapeutics, Inc. RNA interference mediated treatment of Alzheimer's disease using short interfering nucleic acid (siNA)
US20030190635A1 (en) * 2002-02-20 2003-10-09 Mcswiggen James A. RNA interference mediated treatment of Alzheimer's disease using short interfering RNA
US6866859B2 (en) * 2000-08-30 2005-03-15 Biocoat Incorporated Bi-laminar, hyaluronan coatings with silver-based anti-microbial properties
US6623927B1 (en) * 2000-11-08 2003-09-23 Council Of Scientific And Industrial Research Method of detection of allelic variants of SCA2 gene
CA2327208A1 (en) * 2000-11-30 2002-05-30 The Government Of The United States Of America Methods of increasing distribution of therapeutic agents
EP1386004A4 (en) * 2001-04-05 2005-02-16 Ribozyme Pharm Inc Modulation of gene expression associated with inflammation proliferation and neurite outgrowth, using nucleic acid based technologies
AU2002256418A1 (en) * 2001-04-27 2002-11-11 Vertex Pharmaceuticals Incorporated Inhibitors of bace
US20050277133A1 (en) * 2001-05-18 2005-12-15 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
US20030109476A1 (en) * 2001-08-07 2003-06-12 Kmiec Eric B. Compositions and methods for the prevention and treatment of Huntington's disease
US7560254B2 (en) * 2001-08-29 2009-07-14 Ge Healthcare Bio-Sciences Corp. Allele specific primer extension
US6944497B2 (en) * 2001-10-31 2005-09-13 Medtronic, Inc. System and method of treating stuttering by neuromodulation
US20030120282A1 (en) * 2001-12-24 2003-06-26 Scouten Charles W. Stereotaxic manipulator with retrofitted linear scales and digital display device
US7294504B1 (en) * 2001-12-27 2007-11-13 Allele Biotechnology & Pharmaceuticals, Inc. Methods and compositions for DNA mediated gene silencing
US20050096284A1 (en) * 2002-02-20 2005-05-05 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
US7270653B2 (en) * 2002-02-20 2007-09-18 Abbott Research Group Methods of treating abnormal biological conditions using metal oxides
US20040023855A1 (en) * 2002-04-08 2004-02-05 John Constance M. Biologic modulations with nanoparticles
US20040018520A1 (en) * 2002-04-22 2004-01-29 James Thompson Trans-splicing enzymatic nucleic acid mediated biopharmaceutical and protein
AU2003238796A1 (en) 2002-05-28 2003-12-12 U.S. Genomics, Inc. Methods and apparati using single polymer analysis
US7097976B2 (en) * 2002-06-17 2006-08-29 Affymetrix, Inc. Methods of analysis of allelic imbalance
US7008403B1 (en) * 2002-07-19 2006-03-07 Cognitive Ventures Corporation Infusion pump and method for use
US20040023390A1 (en) * 2002-08-05 2004-02-05 Davidson Beverly L. SiRNA-mediated gene silencing with viral vectors
US20050042646A1 (en) * 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
US20040241854A1 (en) 2002-08-05 2004-12-02 Davidson Beverly L. siRNA-mediated gene silencing
EP2213738B1 (en) * 2002-11-14 2012-10-10 Dharmacon, Inc. siRNA molecules targeting Bcl-2
US20050048641A1 (en) * 2002-11-26 2005-03-03 Medtronic, Inc. System and method for delivering polynucleotides to the central nervous system
US8946151B2 (en) * 2003-02-24 2015-02-03 Northern Bristol N.H.S. Trust Frenchay Hospital Method of treating Parkinson's disease in humans by convection-enhanced infusion of glial cell-line derived neurotrophic factor to the putamen
US8512290B2 (en) * 2003-03-20 2013-08-20 Boston Scientific Scimed, Inc. Devices and methods for delivering therapeutic or diagnostic agents
US7632640B2 (en) * 2003-12-08 2009-12-15 The Clinic For Special Children Association of TSPYL polymorphisms with SIDDT syndrome
WO2005065242A2 (en) * 2003-12-29 2005-07-21 Am Biosolutions Method of treating cancer using platelet releasate
WO2005070104A2 (en) * 2004-01-09 2005-08-04 The University Of Tennessee Research Foundation REAL-TIME POLYMERASE CHAIN REACTION-BASED GENOTYPING ASSAY FOR β2-ADRENERGIC RECEPTOR SINGLE NUCLEODITE POLYMORPHISM
US20050177866A1 (en) * 2004-02-09 2005-08-11 Kirsch Steven T. Method and system for acceleration of secure socket layer transactions in a network
US20050202075A1 (en) * 2004-03-12 2005-09-15 Pardridge William M. Delivery of genes encoding short hairpin RNA using receptor-specific nanocontainers
AU2005248147A1 (en) * 2004-05-11 2005-12-08 Alphagen Co., Ltd. Polynucleotides for causing RNA interference and method for inhibiting gene expression using the same
US7563572B2 (en) * 2004-08-02 2009-07-21 University Of Utah Research Foundation Method for long range allele-specific PCR
EP2353651A3 (en) * 2004-10-22 2011-09-07 Neurologix, Inc. Use of apoptosis inhibiting compounds in degenerative neurological disorders
NL1028134C2 (en) * 2005-01-27 2006-07-31 Sara Lee De Nv Method for preparing a drink suitable for consumption from at least two ingredients to be dissolved and / or extracted and an amount of liquid.
AU2006224971B2 (en) * 2005-03-18 2009-07-02 Boston University A method for the detection of chromosomal aneuploidies
TW200635542A (en) * 2005-04-01 2006-10-16 Dharma Drum Mountain Method of establishing and using commemorative material
EP1941059A4 (en) * 2005-10-28 2010-11-03 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of huntingtin gene

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050282198A1 (en) * 1997-05-29 2005-12-22 Interleukin Genetics, Inc. Diagnostics and therapeutics for diseases associated with an IL-1 inflammatory haplotype
US20030152947A1 (en) * 2001-06-15 2003-08-14 Crossman David C. Methods for detecting and treating the early onset of aging-related conditions
US20060014165A1 (en) * 2003-07-14 2006-01-19 Decode Genetics Ehf. Methods of diagnosis and treatment for asthma and other respiratory diseases based on haplotype association

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7829694B2 (en) 2002-11-26 2010-11-09 Medtronic, Inc. Treatment of neurodegenerative disease through intracranial delivery of siRNA
US8618069B2 (en) 2002-11-26 2013-12-31 Medtronic, Inc. Devices, systems and methods for improving memory and/or cognitive function through brain delivery of siRNA
US8415319B2 (en) 2002-11-26 2013-04-09 Medtronic, Inc. Devices, systems and methods for improving memory and/or cognitive function through brain delivery of siRNA
US8119611B2 (en) 2002-11-26 2012-02-21 Medtronic, Inc. Treatment of neurodegenerative disease through intracranial delivery of SIRNA
US8058251B2 (en) 2002-11-26 2011-11-15 Kaemmerer William F Devices, systems and methods for improving memory and/or cognitive function through brain delivery of siRNA
US8957198B2 (en) 2003-02-03 2015-02-17 Medtronic, Inc. Compositions, devices and methods for treatment of Huntington's disease through intracranial delivery of sirna
US7732591B2 (en) 2003-11-25 2010-06-08 Medtronic, Inc. Compositions, devices and methods for treatment of huntington's disease through intracranial delivery of sirna
US7902352B2 (en) 2005-05-06 2011-03-08 Medtronic, Inc. Isolated nucleic acid duplex for reducing huntington gene expression
US8258112B2 (en) 2005-05-06 2012-09-04 Medtronic, Inc Methods and sequences to suppress primate huntington gene Expression
US9133517B2 (en) 2005-06-28 2015-09-15 Medtronics, Inc. Methods and sequences to preferentially suppress expression of mutated huntingtin
US9273356B2 (en) 2006-05-24 2016-03-01 Medtronic, Inc. Methods and kits for linking polymorphic sequences to expanded repeat mutations
US8324367B2 (en) 2006-11-03 2012-12-04 Medtronic, Inc. Compositions and methods for making therapies delivered by viral vectors reversible for safety and allele-specificity
US9375440B2 (en) 2006-11-03 2016-06-28 Medtronic, Inc. Compositions and methods for making therapies delivered by viral vectors reversible for safety and allele-specificity
US7819842B2 (en) 2006-11-21 2010-10-26 Medtronic, Inc. Chronically implantable guide tube for repeated intermittent delivery of materials or fluids to targeted tissue sites
US7988668B2 (en) 2006-11-21 2011-08-02 Medtronic, Inc. Microsyringe for pre-packaged delivery of pharmaceuticals
WO2008147930A3 (en) * 2007-05-23 2009-03-26 Medtronic Inc Methods and kits for linking polymorphic sequences to expanded repeat mutations
WO2008147930A2 (en) * 2007-05-23 2008-12-04 Medtronic, Inc. Methods and kits for linking polymorphic sequences to expanded repeat mutations
WO2008147887A1 (en) * 2007-05-23 2008-12-04 Medtronic, Inc. Methods and kits for linking polymorphic sequences to expanded repeat mutations
EP2780475A4 (en) * 2011-11-17 2015-06-17 Clarient Diagnostic Services Inc Method of allele-specific amplification
EP2780478A4 (en) * 2011-11-17 2015-06-17 Clarient Diagnostic Services Inc Detection of abl mutant by allele-specific amplificaton
WO2013160563A1 (en) * 2012-04-25 2013-10-31 Ho Tho Huu Method for competitive allele-specific cdna synthesis and differential amplification of the cdna products
US9879316B2 (en) 2012-04-25 2018-01-30 Tho Huu Ho Method for competitive allele-specific cDNA synthesis and differential amplification of the cDNA products

Also Published As

Publication number Publication date
EP2298937A1 (en) 2011-03-23
US20080280843A1 (en) 2008-11-13
EP2029773A4 (en) 2009-11-11
EP2029773A1 (en) 2009-03-04

Similar Documents

Publication Publication Date Title
US9273356B2 (en) Methods and kits for linking polymorphic sequences to expanded repeat mutations
EP2298937A1 (en) Methods and kits for linking polymorphic sequences to expanded repeat mutations
JP5695383B2 (en) Hair shape susceptibility gene
JP5695385B2 (en) Hair shape susceptibility gene
WO2010048497A1 (en) Genetic profile of the markers associated with alzheimer&#39;s disease
US20020037508A1 (en) Human single nucleotide polymorphisms
JP5695384B2 (en) Hair shape susceptibility gene
US20080050358A1 (en) Identification of Snps Associated with Hyperlipidemia, Dyslipidemia and Defective Carbohydrate Metabolism
US20050255498A1 (en) APOC1 genetic markers associated with age of onset of Alzheimer&#39;s Disease
EP1819834B1 (en) Tools and methods for the quantification of dna repeats
US20030032099A1 (en) Methods for predicting susceptibility to obesity and obesity-associated health problems
JP7199106B2 (en) Method and kit for determining hyperexcitability in subject
JP5904501B2 (en) Method for detecting type 2 diabetes
WO2021050608A1 (en) Novel genetic markers for postural orthostatic tachycardia syndrome (pots) and methods of use thereof for diagnosis and treatment of the same
WO2010084668A1 (en) Diagnostic method for nephrotic syndrome, prophylactic or therapeutic agent for nephrotic syndrome, and method for screening the prophylactic or therapeutic agent
US20030008301A1 (en) Association between schizophrenia and a two-marker haplotype near PILB gene
JP2009022269A (en) Method for examining obesity, and method for screening prophylactic or curative agent for obesity
EP2586787A1 (en) DEP2 and its uses in major depressive disorder and other related disorders
JP4317376B2 (en) Method for detecting diabetic nephropathy gene
US20110097711A1 (en) Method of judging inflammatory disease by using single nucleotdie polymorphism
US20030087798A1 (en) Atopy-associated sequence variants on chromosome 12
Liu Identification of genetic and epigenetic risk factors for psoriasis and psoratic arthritis
WO2009036513A1 (en) Diagnostic and therapeutic protocols
JPWO2006068111A1 (en) Method for determining phenotype associated with polymorphism of PPARγ gene
EP1379688A2 (en) Polymorphisms in the human cmoat gene and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07777234

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007777234

Country of ref document: EP