WO2008055196A9 - Genemap of the human genes associated with male pattern baldness - Google Patents

Genemap of the human genes associated with male pattern baldness

Info

Publication number
WO2008055196A9
WO2008055196A9 PCT/US2007/083147 US2007083147W WO2008055196A9 WO 2008055196 A9 WO2008055196 A9 WO 2008055196A9 US 2007083147 W US2007083147 W US 2007083147W WO 2008055196 A9 WO2008055196 A9 WO 2008055196A9
Authority
WO
WIPO (PCT)
Prior art keywords
baldness
gene
tables
sample
expression
Prior art date
Application number
PCT/US2007/083147
Other languages
French (fr)
Other versions
WO2008055196A3 (en
WO2008055196A2 (en
Inventor
John Verner Raelson
Walter Edward Bradley
Randall David Little
Bruno Paquin
Quynh Nguyen-Huu
Tim Keith
Pascal Croteau
Abdelmajid Belouchi
Rene Allard
Sophie Debrus
Eerdewegh Paul Van
Jonathan Segal
Original Assignee
Genizon Biosciences Inc
John Verner Raelson
Walter Edward Bradley
Randall David Little
Bruno Paquin
Quynh Nguyen-Huu
Tim Keith
Pascal Croteau
Abdelmajid Belouchi
Rene Allard
Sophie Debrus
Eerdewegh Paul Van
Jonathan Segal
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 Genizon Biosciences Inc, John Verner Raelson, Walter Edward Bradley, Randall David Little, Bruno Paquin, Quynh Nguyen-Huu, Tim Keith, Pascal Croteau, Abdelmajid Belouchi, Rene Allard, Sophie Debrus, Eerdewegh Paul Van, Jonathan Segal filed Critical Genizon Biosciences Inc
Publication of WO2008055196A2 publication Critical patent/WO2008055196A2/en
Publication of WO2008055196A9 publication Critical patent/WO2008055196A9/en
Publication of WO2008055196A3 publication Critical patent/WO2008055196A3/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
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • G16B20/20Allele or variant detection, e.g. single nucleotide polymorphism [SNP] detection
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • G16B20/40Population genetics; Linkage disequilibrium
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B25/00ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression
    • G16B25/10Gene or protein expression profiling; Expression-ratio estimation or normalisation
    • 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
    • C12Q2600/136Screening for pharmacological compounds
    • 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
    • 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
    • C12Q2600/172Haplotypes
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B25/00ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression

Definitions

  • the invention relates to the field of genomics and genetics, including genome analysis and the study of DNA variations.
  • the invention relates to the fields of pharmacogenomics, diagnostics, patient therapy and the use of genetic haplotype information to predict an individual's susceptibility to baldness and/or their response to a particular drug or drugs, so that drugs tailored to genetic differences of population groups may be developed and/or administered to the appropriate population.
  • the invention also relates to a GeneMap for baldness, which links variations in DNA (including both genie and non-genic regions) to an individual's susceptibility to baldness and/or response to a particular drug or drugs.
  • the invention further relates to the genes disclosed in the GeneMap (see Tables 2-4), which is related to methods and reagents for detection of an individual's increased or decreased risk for baldness by identifying at least one polymorphism in one or a combination of the genes from the GeneMap. Also related are the candidate regions identified in Table 1 , which are associated with baldness.
  • the invention further relates to nucleotide sequences of those genes including genomic DNA sequences, cDNA sequences, single nucleotide polymorphisms (SNPs), alleles and haplotypes (see Sequence Listing and Tables 5, 6.1 and 6.2).
  • the invention further relates to isolated nucleic acids comprising these nucleotide sequences and isolated polypeptides or peptides encoded thereby. Also related are expression vectors and host cells comprising the disclosed nucleic acids or fragments thereof, as well as antibodies that bind to the encoded polypeptides or peptides.
  • the present invention further relates to ligands that modulate the activity of the disclosed genes or gene products.
  • the invention relates to diagnostics and therapeutics for baldness, utilizing the disclosed nucleic acids, SNPs, chromosomal regions, gene maps, polypeptides or peptides, antibodies and/or ligands and small molecules that activate or repress relevant signaling events.
  • Baldness affects a large proportion of the population; about 35% of men begin to bald by the time they are 35 years old, and about two-thirds are either bald or have a balding pattern by age 60. Although inherited baldness affects more men than women, the incidence of baldness in women is significant, since it amounts to a third or half of that in men before menopause, and increases greatly after that. Baldness is neither physically disabling nor a life-threatening disease, and is only of cosmetic importance, but it may profoundly affect self-esteem and/or cause psychological stress and anxiety.
  • Changes that contribute to the development of baldness include alterations in the growth cycle of hair. Hair typically progresses through cycles comprising three phases: anagen (active hair growth), catagen (transition phase), and telogen (resting phase during which the hair shaft is shed prior to new growth). As baldness progresses, there is a shift in the percentages of hair follicles in each phase, with the majority shifting from anagen to telogen. The size of hair follicles is also known to decrease while the total number remains relatively constant. Baldness results when the oid shaft is no longer replaced. In most cases, the hair follicle remains alive and the potential for hair re-growth is preserved.
  • the DNA sequences between two human genomes are 99.9% identical.
  • the variations in DNA sequence between individuals can be as an example, deletions of small or large stretches of DNA, insertions of stretches of DNA, variations in the number of repetitive DNA elements, and changes in single base positions in the genome called "single nucleotide polymorphisms" (SNPs).
  • SNPs single nucleotide polymorphisms
  • the candidate gene approach relies on the analysis of a gene in patients who have a disorder in which the gene is thought to play a role. This approach is limited in utility because it only provides for the investigation of genes with known functions. Although variant sequences of candidate genes may be identified using this approach, it is inherently limited by the fact that variant sequences in other genes that contribute to the phenotype will be necessarily missed when the technique is employed. Recently, the first genome-wide scan (GWS) has been shown to be efficient in identifying baldness susceptibility markers, such as the androgen receptor on chromosome X (Hillmer et al., 2005).
  • GWS first genome-wide scan
  • a GWS searches throughout the genome without any a priori hypothesis and consequently can identify genes that are not obvious candidates for the disorder as well as genes that are relevant candidates for the disorder, as well as chromosomal regions that are structurally important where an "associated allele", a "particular allele of a polymorphic locus", or the likes can influence the expression of specific genes.
  • LD linkage disequilibrium
  • identifying susceptibility genes associated with baldness and their respective biochemical pathways will facilitate the development of effective treatments for baldness and hair loss, and will permit the identification of diagnostic markers which will predict the propensity for balding and permit therapeutic intervention before baldness can occur.
  • the identification of those genetic markers would provide the basis for novel genetic tests and eliminate or reduce the therapeutic methods currently used.
  • the identification of those genetic markers will also provide the development of effective therapeutic intervention for a much greater proportion of the individuals affected by baldness than the current methods, which are effective only in a small number of baldness cases.
  • the present invention satisfies this need and provides related advantages as well.
  • Allele One of a pair, or series, of forms of a gene or non-genic region that occur at a given locus in a chromosome. Alleles are symbolized with the same basic symbol ⁇ e.g., B for dominant and b for recessive; B1 , B2, Bn for n additive alleles at a locus). In a normal diploid cell there are two alleles of any one gene (one from each parent), which occupy the same relative position (locus) on homologous chromosomes. Within a population there may be more than two alleles of a gene. See multiple alleles. SNPs also have alleles, i.e., the two (or more) nucleotides that characterize the SNP.
  • Amplification of nucleic acids refers to methods such as polymerase chain reaction (PCR), ligation amplification (or ligase chain reaction, LCR) and amplification methods based on the use of Q-beta replicase. These methods are well known in the art and are described, for example, in U.S. Patent Nos. 4,683,195 and 4,683,202. Reagents and hardware for conducting PCR are commercially available. Primers useful for amplifying sequences from the disorder region are preferably complementary to, and preferably hybridize specifically to, sequences in the disorder region or in regions that flank a target region therein. Genes from Tables 2-4 generated by amplification may be sequenced directly. Alternatively, the amplified sequence(s) may be cloned prior to sequence analysis.
  • PCR polymerase chain reaction
  • LCR ligase chain reaction
  • Antigenic component is a moiety tnat binds to its specific aruiQody witn sufficiently high affinity to form a detectable antigen-antibody complex.
  • Antibodies refer to polyclonal and/or monoclonal antibodies and fragments thereof, and immunologic binding equivalents thereof, that can bind to proteins and fragments thereof or to nucleic acid sequences from the disorder region, particularly from the disorder gene products or a portion thereof.
  • the term antibody is used both to refer to a homogeneous molecular entity, or a mixture such as a serum product made up of a plurality of different molecular entities.
  • Proteins may be prepared synthetically in a protein synthesizer and coupled to a carrier molecule and injected over several months into rabbits. Rabbit sera are tested for immunoreactivity to the protein or fragment.
  • Monoclonal antibodies may be made by injecting mice with the proteins, or fragments thereof Monoclonal antibodies will be screened by ELISA and tested for specific immunoreactivity with protein or fragments thereof (Harlow et al 1988, Antibodies A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY) These antibodies will be useful in assays as well as therapeutics
  • Associated allele refers to an allele at a polymorphic locus that is associated with a particular phenotype of interest, e g , a predisposition to a disorder or a particular drug response
  • Baldness encompasses the complete or partial loss of hair and a variety of different types of alopecia (e g , alopecia areata, alopecia totalis, alopecia universalis, alopecia diffusa, alopeciapartialis and androgenic alopecia) due to intrinsic factors (e g , aging, hormone production and/or hormonal imbalances, pregnancy, etc ) or genetic factors, as well as disease- or extrinsic factors-related hair loss (e g , thyroid function imbalance, autoimmune disorders, stress, vitamin deficiency and/or other dietary imbalances, chemotherapy, radiotherapy or other treatments, etc ) Baldness of the scalp is characterized by, e g , loss of activity and/or miniaturization of hair follicles hair loss, slowing of hair growth, thinning of the hair, appearance of shorter and weaker hairs, as well as any of a number of characteristic structural and/or molecular features
  • baldness refers to
  • cDNA refers to complementary or copy DNA produced from an RNA template by the action of RNA-dependent DNA polymerase (reverse transcriptase)
  • a cDNA clone means a duplex DNA sequence complementary to an RNA molecule of interest, included in a cloning vector or PCR amplified
  • cDNA library refers to a collection of recombinant DNA molecules containing cDNA inserts that together comprise essentially all of the expressed genes of an organism or tissue.
  • a cDNA library can be prepared by methods known to one skilled in the art (see, e.g., Cowell and Austin, 1997, "DNA Library Protocols," Methods in Molecular Biology). Generally, RNA is first isolated from the cells of the desired organism, and the RNA is used to prepare cDNA molecules.
  • Cloning refers to the use of recombinant DNA techniques to insert a particular gene or other DNA sequence into a vector molecule. In order to successfully clone a desired gene, it is necessary to use methods for generating DNA fragments, for joining the fragments to vector molecules, for introducing the composite DNA molecule into a host cell in which it can replicate, and for selecting the clone having the target gene from amongst the recipient host cells.
  • Cloning vector refers to a plasmid or phage DNA or other DNA molecule that is able to replicate in a host cell.
  • the cloning vector is typically characterized by one or more endonuclease recognition sites at which such DNA sequences may be cleaved in a determinable fashion without loss of an essential biological function of the DNA, and which may contain a selectable marker suitable for use in the identification of cells containing the vector.
  • Coding sequence or a protein-coding sequence is a polynucleotide sequence capable of being transcribed into mRNA and/or capable of being translated into a polypeptide or peptide.
  • the boundaries of the coding sequence are typically determined by a translation start codon at the 5'-terminus and a translation stop codon at the 3'-terminus.
  • Complement of a nucleic acid sequence refers to the antisense sequence that participates in Watson-Crick base-pairing with the original sequence.
  • Disorder region refers to the portions of the human chromosomes displayed in Table 1 bounded by the markers from Tables 2-5, 6.1 and 6.2.
  • Disorder-associated nucleic acid or polypeptide sequence refers to a nucleic acid sequence that maps to region of Table 1 or the polypeptides encoded therein (Tables 2-5, 6.1 and 6.2 SNPs, nucleic acids, and polypeptides).
  • nucleic acids this encompasses sequences that are identical or complementary to the gene sequences from Tables 2-4, as well as sequence-conservative, function-conservative, and non-conservative variants thereof.
  • polypeptides this encompasses sequences that are identical to the polypeptide, as well as function-conservative and non-conservative variants thereof.
  • alleles of naturally-occurring polymorphisms causative of baldness such as, but not limited to, alleles that cause altered expression of genes of Tables 2-4 and alleles that cause altered protein levels or stability (e.g., decreased levels, increased levels, expression in an inappropriate tissue type, increased stability, and decreased stability.
  • Expression vector refers to a vehicle or plasmid that is capable of expressing a gene that has been cloned into it, after transformation or integration in a host cell.
  • the cloned gene is usually placed under the control of (i.e., operably linked to) a regulatory sequence.
  • Function-conservative variants are those in which a change in one or more nucleotides in a given codon position results in a polypeptide sequence in which a given amino acid residue in the polypeptide has been replaced by a conservative amino acid substitution. Function-conservative variants also include analogs of a given polypeptide and any polypeptides that have the ability to elicit antibodies specific to a designated polypeptide.
  • Founder population Also called a population isolate, this is a large number of people who have mostly descended, in genetic isolation from other populations, from a much smaller number of people who lived many generations ago.
  • Gene refers to a DNA sequence that encodes through its template or messenger RNA a sequence of amino acids characteristic of a specific peptide, polypeptide, or protein.
  • the term "gene” also refers to DNA sequence that encodes an RNA product.
  • the term gene as used herein with reference to genomic DNA includes intervening, non-coding regions, as well as regulatory regions, and can include 5' and 3' ends.
  • a gene sequence is wild-type if such sequence is usually found in individuals unaffected by the disorder or condition of interest. However, environmental factors and other genes can also play an important role in the ultimate determination of the disorder. In the context of complex disorders involving multiple genes (oligogenic disorder), the wild type, or normal sequence can also be associated with a measurable risk or susceptibility, receiving its reference status based on its frequency in the general population.
  • GeneMaps are defined as groups of gene(s) that are directly or indirectly involved in at least one phenotype of a disorder. As such, GeneMaps enable the development of synergistic diagnostic products, creating "theranostics”. Genotype: Set of alleles at a specified locus or loci.
  • Haplotype The allelic pattern of a group of (usually contiguous) DNA markers or other polymorphic loci along an individual chromosome or double helical DNA segment. Haplotypes identify individual chromosomes or chromosome segments. The presence of shared haplotype patterns among a group of individuals implies that the locus defined by the haplotype has been inherited, identical by descent (IBD), from a common ancestor. Detection of identical by descent haplotypes is the basis of linkage disequilibrium (LD) mapping. Haplotypes are broken down through the generations by recombination and mutation. In some instances, a specific allele or haplotype may be associated with susceptibility to a disorder or condition of interest, e.g., baldness. In other instances, an allele or haplotype may be associated with a decrease in susceptibility to a disorder or condition of interest, i.e., a protective sequence.
  • IBD identical by descent
  • Detection of identical by descent haplotypes
  • Host includes prokaryotes and eukaryotes.
  • the term includes an organism or cell that is the recipient of an expression vector (e.g., autonomously replicating or integrating vector).
  • Hybridizable nucleic acids are hybridizable to each other when at least one strand of the nucleic acid can anneal to another nucleic acid strand under defined stringency conditions.
  • hybridization requires that the two nucleic acids contain at least 10 substantially complementary nucleotides; depending on the stringency of hybridization, however, mismatches may be tolerated.
  • the appropriate stringency for hybridizing nucleic acids depends on the length of the nucleic acids and the degree of complementarity, and can be determined in accordance with the methods described herein.
  • IBD Identity by descent
  • Identity is a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, as determined by comparing the sequences. In the art, identity also means the degree of sequence relatedness between polypeptide or polynucleotide sequences, as the case may be, as determined by the match between strings of such sequences. Identity and similarity can be readily calculated by known methods, including but not limited to those described in A.M. Lesk (ed), 1988, Computational Molecular Biology, Oxford University Press, NY; D.W. Smith (ed), 1993, Biocomputing. Informatics and Genome Projects, Academic Press, NY; A.M. Griffin and H. G. Griffin, H.
  • Immunogenic component is a moiety that is capable of eliciting a humoral and/or cellular immune response in a host animal.
  • Isolated nucleic acids are nucleic acids separated away from other components (e.g., DNA, RNA, and protein) with which they are associated (e.g., as obtained from cells, chemical synthesis systems, or phage or nucleic acid libraries). Isolated nucleic acids are at least 60% free, preferably 75% free, and most preferably 90% free from other associated components. In accordance with the present invention, isolated nucleic acids can be obtained by methods described herein, or other established methods, including isolation from natural sources (e.g., cells, tissues, or organs), chemical synthesis, recombinant methods, combinations of recombinant and chemical methods, and library screening methods.
  • natural sources e.g., cells, tissues, or organs
  • chemical synthesis e.g., recombinant methods, combinations of recombinant and chemical methods, and library screening methods.
  • Isolated polypeptides or peptides are those that are separated from other components (e.g., DNA, RNA, and other polypeptides or peptides) with which they are associated (e.g., as obtained from cells, translation systems, or chemical synthesis systems).
  • isolated polypeptides or peptides are at least 10% pure; more preferably, 80% or 90% pure.
  • Isolated polypeptides and peptides include those obtained by methods described herein, or other established methods, including isolation from natural sources (e.g., cells, tissues, or organs), chemical synthesis, recombinant methods, or combinations of recombinant and chemical methods.
  • Proteins or polypeptides referred to herein as recombinant are proteins or polypeptides produced by the expression of recombinant nucleic acids.
  • a portion as used herein with regard to a protein or polypeptide refers to fragments of that protein or polypeptide. The fragments can range in size from 5 amino acid residues to all but one residue of the entire protein sequence. Thus, a portion or fragment can be at least 5, 5-50, 50-100, I00-200, 200-400, 400-800, or more consecutive amino acid residues of a protein or polypeptide.
  • Linkage disequilibrium (LD) the situation in which the alleles for two or more loci do not occur together in individuals sampled from a population at frequencies predicted by the product of their individual allele frequencies.
  • markers that are in LD do not follow Mendel's second law of independent random segregation.
  • LD can be caused by any of several demographic or population artifacts as well as by the presence of genetic linkage between markers. However, when these artifacts are controlled and eliminated as sources of LD, then LD results directly from the fact that the loci involved are located close to each other on the same chromosome so that specific combinations of alleles for different markers (haplotypes) are inherited together.
  • Markers that are in high LD can be assumed to be located near each other and a marker or haplotype that is in high LD with a genetic trait can be assumed to be located near the gene that affects that trait. The physical proximity of markers can be measured in family studies where it is called linkage or in population studies where it is called linkage disequilibrium.
  • LD mapping population based gene mapping, which locates disorder genes by identifying regions of the genome where haplotypes or marker variation patterns are shared statistically more frequently among disorder patients compared to healthy controls. This method is based upon the assumption that many of the patients will have inherited an allele associated with the disorder from a common ancestor (IBD), and that this allele will be in LD with the disorder gene.
  • IBD common ancestor
  • Locus a specific position along a chromosome or DNA sequence.
  • a locus could be a gene, a marker, a chromosomal band or a specific sequence of one or more nucleotides.
  • MAF Minor allele frequency
  • Markers an identifiable DNA sequence that is variable (polymorphic) for different individuals within a population. These sequences facilitate the study of inheritance of a trait or a gene. Such markers are used in mapping the order of genes along chromosomes and in following the inheritance of particular genes; genes closely linked to the marker or in LD with the marker will generally be inherited with it. Two types of markers are commonly used in genetic analysis, microsatellites and SNPs.
  • Microsatellite DNA of eukaryotic cells comprising a repetitive, short sequence of DNA that is present as tandem repeats and in highly variable copy number, flanked by sequences unique to that locus.
  • Mutant sequence if it differs from one or more wild-type sequences.
  • a nucleic acid from a gene listed in Tables 2-4 containing a particular allele of a single nucleotide polymorphism may be a mutant sequence.
  • the individual carrying this allele has increased susceptibility toward the disorder/trait or condition of interest.
  • the mutant sequence might also refer to an allele that decreases the susceptibility toward a disorder/trait or condition of interest and thus acts in a protective manner.
  • the term mutation may also be used to describe a specific allele of a polymorphic locus.
  • Non-conservative variants are those in which a change in one or more nucleotides in a given codon position results in a polypeptide sequence in which a given amino acid residue in a polypeptide has been replaced by a non-conservative amino acid substitution.
  • Non-conservative variants also include polypeptides comprising non- conservative amino acid substitutions.
  • Nucleic acid or polynucleotide purine- and pyrimidine-containing polymers of any length, either polyribonucleotides or polydeoxyribonucleotide or mixed polyribo polydeoxyribonucleotides. This includes single-and double-stranded molecules, i.e., DNA-DNA, DNA-RNA and RNA-RNA hybrids, as well as protein nucleic acids (PNA) formed by conjugating bases to an amino acid backbone. This also includes nucleic acids containing modified bases.
  • PNA protein nucleic acids
  • Nucleotide a nucleotide, the unit of a DNA molecule, is composed of a base, a 2 - deoxyribose and phosphate ester(s) attached at the 5' carbon of the deoxyribose. For its incorporation in DNA, the nucleotide needs to possess three phosphate esters but it is converted into a monoester in the process.
  • Operably linked means that the promoter controls the initiation of expression of the gene.
  • a promoter is operably linked to a sequence of proximal DNA if upon introduction into a host cell the promoter determines the transcription of the proximal DNA sequence(s) into one or more species of RNA.
  • a promoter is operably linked to a DNA sequence if the promoter is capable of initiating transcription of that DNA sequence.
  • Ortholog denotes a gene or polypeptide obtained from one species that has homology to an analogous gene or polypeptide from a different species.
  • Paralog denotes a gene or polypeptide obtained from a given species that has homology to a distinct gene or polypeptide from that same species.
  • Phenotype any visible, detectable or otherwise measurable property of an organism such as symptoms of, or susceptibility to, a disorder.
  • Polymorphism occurrence of two or more alternative genomic sequences or alleles between or among different genomes or individuals at a single locus.
  • a polymorphic site thus refers specifically to the locus at which the variation occurs.
  • an individual carrying a particular allele of a polymorphism has an increased or decreased susceptibility toward a disorder or condition of interest.
  • a portion as used with regard to a nucleic acid or polynucleotide refers to fragments of that nucleic acid or polynucleotide.
  • the fragments can range in size from 8 nucleotides to all but one nucleotide of the entire gene sequence.
  • the fragments are at least about 8 to about 10 nucleotides in length; at least about 12 nucleotides in length; at least about 15 to about 20 nucleotides in length; at least about 25 nucleotides in length; or at least about 35 to about 55 nucleotides in length.
  • Probe or primer refers to a nucleic acid or oligonucleotide that forms a hybrid structure with a sequence in a target region of a nucleic acid due to complementarity of the probe or primer sequence to at least one portion of the target region sequence.
  • Protein and polypeptide are synonymous. Peptides are defined as fragments or portions of polypeptides, preferably fragments or portions having at least one functional activity (e.g., proteolysis, adhesion, fusion, antigenic, or intracellular activity) as the complete polypeptide sequence.
  • functional activity e.g., proteolysis, adhesion, fusion, antigenic, or intracellular activity
  • Recombinant nucleic acids nuclei acids which have been produced by recombinant DNA methodology, including those nucleic acids that are generated by procedures which rely upon a method of artificial replication, such as the polymerase chain reaction (PCR) and/or cloning into a vector using restriction enzymes. Portions of recombinant nucleic acids which code for polypeptides can be identified and isolated by, for example, the method of M. Jasin et al., U.S. Patent No. 4,952,501. Regulatory sequence: refers to a nucleic acid sequence that controls or regulates expression of structural genes when operably linked to those genes.
  • Regulatory sequences will vary depending on whether the vector is designed to express the operably linked gene in a prokaryotic or eukaryotic host, and may contain transcriptional elements such as enhancer elements, termination sequences, tissue-specificity elements and/or translational initiation and termination sites.
  • Sample refers to a biological sample, such as, for example, tissue or fluid isolated from an individual or animal (including, without limitation, plasma, serum, cerebrospinal fluid, lymph, tears, nails, hair, saliva, milk, pus, and tissue exudates and secretions) or from in vitro cell culture-constituents, as well as samples obtained from, for example, a laboratory procedure.
  • tissue or fluid isolated from an individual or animal (including, without limitation, plasma, serum, cerebrospinal fluid, lymph, tears, nails, hair, saliva, milk, pus, and tissue exudates and secretions) or from in vitro cell culture-constituents, as well as samples obtained from, for example, a laboratory procedure.
  • Single nucleotide polymorphism variation of a single nucleotide. This includes the replacement of one nucleotide by another and deletion or insertion of a single nucleotide.
  • SNPs are biallelic markers although tri- and tetra-allelic markers also exist.
  • SNP A ⁇ C may comprise allele C or allele A (Tables 5, 6.1 and 6.2).
  • a nucleic acid molecule comprising SNP A ⁇ C may include a C or A at the polymorphic position.
  • haplotype is used, e.g. the genotype of the SNPs in a single DNA strand that are linked to one another.
  • haplotype is used to describe a combination of SNP alleles, e.g., the alleles of the SNPs found together on a single DNA molecule.
  • the SNPs in a haplotype are in linkage disequilibrium with one another.
  • variants are those in which a change of one or more nucleotides in a given codon position results in no alteration in the amino acid encoded at that position (Ae., silent mutation).
  • nucleic acid or fragment thereof is substantially homologous to another if, when optimally aligned (with appropriate nucleotide insertions and/or deletions) with the other nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least 60% of the nucleotide bases, usually at least 70%, more usually at least 80%, preferably at least 90%, and more preferably at least 95-98% of the nucleotide bases.
  • substantial homology exists when a nucleic acid or fragment thereof will hybridize, under selective hybridization conditions, to another nucleic acid (or a complementary strand thereof). Selectivity of hybridization exists when hybridization which is substantially more selective than total lack of specificity occurs.
  • selective hybridization will occur when there is at least about 55% sequence identity over a stretch of at least about nine or more nucleotides, preferably at least about 65%, more preferably at least about 75%, and most preferably at least about 90% (M. Kanehisa, 1984, NucL Acids Res. 11 :203-213).
  • the length of homology comparison, as described, may be over longer stretches, and in certain embodiments will often be over a stretch of at least 14 nucleotides, usually at least 20 nucleotides, more usually at least 24 nucleotides, typically at least 28 nucleotides, more typically at least 32 nucleotides, and preferably at least 36 or more nucleotides.
  • Wild-type gene from Tables 2-4 refers to the reference sequence.
  • the wild-type gene sequences from Tables 2-4 used to identify the variants (single nucleotide polymorphisms, alleles, and haplotypes) described in detail herein.
  • MPB male pattern baldness
  • Androgenic alopecia which is inherited as an autosomal dominant trait with partial penetrance, is known to be androgen-dependent. Androgens are steroid hormones found circulating at varying levels in both men and women. They are essential in sex differentiation, development, and reproductive function. This is evidenced in the fact tnat castrated males do not develop baldness.
  • Hair follicles initially appear in utero. No new follicles are created after birth, and it is believed that none are lost in adult life. However, in androgenic alopecia, hair follicles do become progressively smaller (miniaturized). Hair follicles exhibit cyclic activity. Each period of active growth of hair (anagen) alternates with a resting period (telogen), separated by a relatively short transition phase (catagen). Hair growth on the human scalp is a mosaic of follicular activity with each follicle at a stage independent of its neighbors. In the human scalp, anagen may occupy three years or more; however, the percentage of follicles in telogen increases with age, resulting in a gradual thinning.
  • telogen the ratio of telogen to anagen is increased still further.
  • in androgenic alopecia the hairs in affected areas become steadily shorter and finer, and ultimately may be reduced to the short, fine, unpigmented hair known as vellus hair.
  • Testosterone is the major circulating androgen. Because circulating testosterone is largely bound to sex hormone binding globulin (SHBG), the availability of testosterone depends not only on its total concentration, but also on the level of SHBG. Past studies have indicated that plasma testosterone levels in androgenic alopecia appear to be normal, but SHBG levels tend to be low.
  • SHBG sex hormone binding globulin
  • Androgens such as DHT
  • DHT androgen receptors
  • antiandrogens spironolactone, cyproterone acetate, flutamide and cimetidine
  • DHT ligands
  • Androgenic alopecia is a strongly heritable condition and is inherited as a complex trait. Familial aggregation, twin studies and consistent ethnic differences in trait frequency have strongly supported the important role of genetic factors in the cause of baldness. Niermann et al., (1964) and Hayakawa et al., (1992) reported the concordance of androgenic alopecia in all the monozygotic pairs and half of the dizygotics pairs they studied, suggesting a very high heritability and an autosomal dominant inheritance pattern.
  • Minoxidil is a potent vasodilator wnich has been used as a treatment for hypertension
  • a noted side effect of this treatment was the growth of hair on parts of the body Research indicates that minoxidil did not help the majority of balding men, although it did help a specific population of minimally balding young men, who had begun to bald within five years
  • the reason for the effectiveness of minoxidil is not known, but is suggested to be due to an increase in blood flow caused by the vasodilating effect of the drug
  • the longterm effects of minoxidil treatment are also not known
  • Another treatment is the topical use of progesterone, a molecule similar to testosterone, which competes with testosterone for binding 5-alpha-reductase, the enzyme that converts testosterone to DHT, and results in lowering the production of DHT from testosterone
  • progesterone a molecule similar to testosterone
  • the present invention is based on the discovery of genes associated with baldness.
  • disorder-associated loci candidate regions listed in Table 1 and genes listed in Tables 2-4) are therefore identified by the statistically significant differences in allele frequencies between the cases and the controls.
  • 65 candidate regions showing a difference with a -Iog10 P value of 3.0 or higher are identified along with previously known region that has been previously reported to be associated with baldness (example: the androgen receptor on Chromosome X).
  • the invention provides a method for the discovery of genes associated with baldness and the construction of a GeneMap for baldness in a human population, comprising the following steps (see Example section herein):
  • Step 1 Recruit patients (cases) and controls
  • 500 patients diagnosed for baldness along with 500 control individuals are recruited from the Quebec Founder Population (QFP).
  • less than 500 male patients diagnosed for baldness along with less than 500 male control individuals are recruited from the Quebec Founder Population (QFP).
  • more than 500 male patients diagnosed for baldness along with more than 500 male control individuals are recruited from the Quebec Founder Population (QFP).
  • the present invention is performed as a whole or partially with DNA samples from individuals of another founder population than the Quebec population or from the general population.
  • Step 2 DNA extraction and dosage
  • sample comprising cells or nucleic acids from patients or controls may be used.
  • Preferred samples are those easily obtained from the patient or control.
  • Such samples include, but are not limited to blood, peripheral lymphocytes, buccal swabs, epithelial cell swabs, nails, hair, bronchoalveolar lavage fluid, sputum, or other body fluid or tissue obtained from an individual.
  • DNA is extracted from such samples in the quantity and quality necessary to perform the invention using conventional DNA extraction and dosage techniques.
  • the present invention is not linked to any DNA extraction or dosage platform in particular.
  • the extracted DNA from case and control samples from recruited individuals is pooled together in various pools. Pools are designed to segregate probands (cases) from controls.
  • proband pools consisting of DNA extracted from recruited individuals, are separated according to level of baldness as determined on the Norwood/Hamilton scale (Norwood 1975), and further separated by age group.
  • Control pools consisting of DNA extracted from control individuals, but is not limited to pools separated as determined by Norwood/Hamilton scales.
  • the preferred pools distribution is as follows- six proband pools consisted of Norwood/Hamilton type Vl and 2 proband pools consisted of Norwood/Hamilton type VII.
  • the control pools consisted of 5 pools of 57 samples with Norwood/Hamilton type I, and 3 pools of 60, 60, or 65 samples with Norwood/Hamilton type II.
  • proband samples of 468 male subjects are used to construct the case pools and 470 male controls are used to construct the control pools.
  • probant samples of less or more than 500 male subjects are used to construct the case pools and less or more than 500 male controls are used to construct the control pools.
  • Step 3 Genotype the proband and control pools
  • assay specific and/or locus-specific and/or allele-specific oligonucleotides for every SNP marker of the present invention are organized onto one or more arrays.
  • the genotype at each SNP locus is revealed by hybridizing short PCR fragments comprising each SNP locus onto these arrays.
  • the arrays permit a high-throughput genome wide association study using DNA samples from individuals of the Quebec founder population.
  • Such assay-specific and/or locus- specific and/or allele-specific oligonucleotides necessary for scoring each SNP of the present invention are preferably organized onto a solid support.
  • Such supports can be arrayed on wafers, glass slides, beads or any other type of solid support.
  • the assay-specific and/or locus-specific and/or allele-specific oligonucleotides are not organized onto a solid support but are still used as a whole, in panels or one by one.
  • the present invention is therefore not linked to any genotyping platform in particular.
  • one or more portions of the SNPs maps are used to screen the whole genome, a subset of chromosomes, a chromosome, a subset of genomic regions or a single genomic region.
  • Step 4 Exclude the markers that did not pass the quality control of the assay.
  • the quality controls consist of, but are not limited to, the following criteria: eliminate SNPs that are non-polymorphic in the Quebec founder population or have ⁇ 10% minor allele frequency (MAF).
  • Step 5 Perform the genetic analysis on the results obtained
  • genetic analysis is performed on all the genotypes from step 3.
  • genetic analysis is performed on a total of 248,535 SNPs.
  • the data analysis compares the relative fluorescence intensities of features corresponding to the reference allele of a given SNP with those corresponding to the alternate allele, to calculate a p-hat value.
  • the latter is proportional to the fluorescence signal from perfect match features for the reference allele divided by the sum of fluorescence signals from perfect match features for the reference plus the alternate alleles.
  • P-hat assumes values close to 1 (typically 0.9) for pure reference samples and close to 0 (typically 0.1 ) for pure alternate samples, and can be used as a measured estimate of the reference allele frequency of a SNP in a DNA pool.
  • delta p-hat is calculated using the weighted average of case and control p-hats. Delta p-hat is a reliable estimate of the allele frequency difference between the cases and controls.
  • the data is analyzed according to the p-hat value obtained from the previous embodiment on each pool.
  • Single marker P values are calculated for all markers within the genome wide scan map as described in Example 3 herein, using the p-hat value.
  • the combined P values across multi-marker sliding windows are calculated after the method of Fisher (described in Example 3 herein).
  • step 5 the candidate regions that were identified by step 5 are further mapped and confirmed for the purpose of refinement and validation.
  • the cases and controls are individually genotyped to confirm the candidate regions.
  • the confirmed candidate regions are processed by fine mapping to refine the candidate regions.
  • this fine mapping is performed with a density of genetic markers higher than in the genome wide scan (step 3) using any genotyping platform available in the art.
  • Such fine mapping can be, but is not limited to, typing the allele via an allele-specific elongation assay that is then ligated to a locus-specific oligonucleotide.
  • Such assays can be performed directly on the genomic DNA at a highly multiplex level and the products can be amplified using universal oligonucleotides.
  • the density of genetic markers can be, but is not limited to, a set of SNP markers with an average inter-marker distance of 1 -4 Kb distributed over about 400 Kb to 1 Mb, roughly centered at the highest point of the GWS association.
  • the preferred samples are those obtained from baldness samples including the ones used for the GWS.
  • samples used for the GWS can be supplemented with other samples to increase total cases and controls.
  • 650 cases and 650 controls samples can be used for the fine mapping genotyping analysis.
  • the genetic analysis of the results obtained using haplotype information (available after confirmatory mapping individual samples, see Example section herein) as well as single-marker association (as performed as in step 5, described herein) are performed as described herein (step 5 and Example section).
  • the candidate regions that are validated and confirmed after this analysis proceed to a gene mining step described in Example 5, herein, to characterize their marker and genetic content.
  • Step 7 SNP and DNA polymorphism discovery
  • all the candidate genes and regions identified in step 6 are sequenced for polymorphism identification.
  • the entire region, including all introns, is sequenced to identify all polymorphisms.
  • the candidate genes are prioritized for sequencing, and only functional gene elements (promoters, exons and splice sites) are sequenced.
  • previously identified polymorphisms in the candidate regions can also be used.
  • SNPs from dbSNP, Perlegen Sciences, Inc., or others can also be used rather than resequencing the candidate regions to identify polymorphisms.
  • the discovery of SNPs and DNA polymorphisms generally compi ises a s ⁇ e ⁇ consisting of determining the major haplotypes in the region to be sequenced.
  • the preferred samples are selected according to which haplotypes contribute to the association signal observed in the region to be sequenced.
  • the purpose is to select a set of samples that cover all the major haplotypes in the given region.
  • Each major haplotype is preferably analyzed in at least a few individuals.
  • Any analytical procedure may be used to detect the presence or absence of variant nucleotides at one or more polymorphic positions of the invention.
  • allelic variation requires a mutation discrimination technique, optionally an amplification reaction and optionally a signal generation system. Any means of mutation detection or discrimination may be used. For instance, DNA sequencing, scanning methods, hybridization, extension based methods, incorporation based methods, restriction enzyme-based methods and ligation-based methods may be used in the methods of the invention.
  • Sequencing methods include, but are not limited to, direct sequencing, and sequencing by hybridization.
  • Scanning methods include, but are not limited to, protein truncation test (PTT), single-strand conformation polymorphism analysis (SSCP), denaturing gradient gel electrophoresis (DGGE), temperature gradient gel electrophoresis (TGGE), cleavage, heteroduplex analysis, chemical mismatch cleavage (CMC), and enzymatic mismatch cleavage.
  • Hybridization-based methods of detection include, but are not limited to, solid phase hybridization such as dot blots, multiple allele specific diagnostic assay (MASDA), reverse dot blots, and oligonucleotide arrays (DNA Chips).
  • Solution phase hybridization amplification methods may also be used, such as Taqman.
  • Extension based methods include, but are not limited to, amplification refraction mutation systems (ARMS), amplification refractory mutation systems (ALEX), and competitive oligonucleotide priming systems (COPS).
  • Incorporation based methods include, but are not limited to, mini-sequencing and arrayed primer extension (APEX).
  • Restriction enzyme-based detection systems include, but are not limited to, restriction site generating PCR.
  • ligation based detection methods include, but are not limited to, oligonucleotide ligation assays (OLA).
  • Signal generation or detection systems that may be used in the methods of the invention include, but are not limited to, fluorescence methods such as fluorescence resonance energy transfer (FRET), fluorescence quenching, fluorescence polarization as well as other chemiluminescence, electrochemiluminescence, Raman, radioactivity, colometric methods, hybridization protection assays and mass spectrometry methods.
  • Further amplification methods include, but are not limited to self sustained replication (SSR), nucleic acid sequence based amplification (NASBA), ligase chain reaction (LCR), strand displacement amplification (SDA) and branched DNA (B-DNA).
  • SSR self sustained replication
  • NASBA nucleic acid sequence based amplification
  • LCR ligase chain reaction
  • SDA strand displacement amplification
  • B-DNA branched DNA
  • This step further maps the candidate regions and genes confirmed in the previous step to identify and validate the responsible polymorphisms associated with baldness in the human population.
  • the discovered SNPs and polymorphisms of step 7 are ultrafine mapped at a higher density of markers than the fine mapping described herein using the same technology described in step 6
  • GeneMap for baldness
  • the gene content of this GeneMap is described in more detail below
  • Such GeneMap can be used for other methods of the invention comprising the diagnostic methods described herein, the susceptibility to baldness, the response to a particular drug, the efficacy of a particular drug, the screening methods described herein and the treatment methods described herein
  • the GeneMap consists of genes and targets, in a variety of combinations, identified from the candidate regions listed in Table 1 In the preferred embodiment, all genes from Tables 2-4 are present in the GeneMap
  • IPA Ingenuity database
  • ACTA1 AKR1 B7, AR 1 CCNE1 , CD79B, CDC25B, CITED2, CYP17A1.
  • DHFR EBF1.
  • HSPA1 B KLKBI 1 LHB, proliferation, Tissue SLC2A4, TCF3,
  • Network 1 contains 44 nodes (35 original and 9 manual additions) and includes the 4 input genes (Figure 1 ). A short description of these 4 genes follows. By virtue of their role in hair follicle morphogenesis and/or androgen signaling, several genes from this network are very good candidates for involvement in the pathophysiology of MPB.
  • EDA2R gene is also known as XEDAR, EDAA2R, EDA-A2R and TNFRSF27.
  • This protein is a type III transmembrane protein of the TNFR (tumor necrosis factor receptor) superfamily, and contains 3 cysteine-rich repeats and a single transmembrane domain but lacks an N-terminal signal peptide (Yan et al 2000). Multiple alternatively spliced transcript variants have been found for this gene, but some variants lack sufficient support.
  • This receptor specifically binds EDA-A2, an isoform of the anhidrotic ectodermal dysplasia (EDA) gene (see below).
  • EDA-A2 binding to EDA2R leads to interaction of its intracellular domain with TRAF3 and TRAF6 and activation of both NF- KB and JNK pathways (Botchkarev and Fessing 2005).
  • E16 and E17 XEDAR expression was expressed in large amounts in maturing follicles.
  • the pattern of expression was confined to hair follicles (Yan et al 2000).
  • AR The androgen receptor gene (AR) is more than 90 kb long and codes for a protein that has 3 major functional domains: the N-terminal domain, DNA-binding domain, and androgen-binding domain.
  • the protein functions as a steroid-hormone activated transcription factor. Upon binding the hormone ligand, the receptor dissociates from accessory proteins, translocates into the nucleus, dimerizes, and then stimulates transcription of androgen responsive genes.
  • This gene contains 2 polymorphic trinucleotide repeat segments that encode polyglutamine and polyglycine tracts in the N- terminal transactivation domain of its protein. The involvement of androgen signaling in the development of MPB is well known.
  • EBF1 the gene early B cell factor 1 encodes a transcription which plays an essential role in B-cell develoment.
  • a role of the immune system on hair follicle cycling has been hypothesized (Stenn and Paus 2001 ).
  • TCF family members LEF1 and TCF1 were first characterized by their role in T- and B-cell development (Staal and Clevers 2000), and were shown subsequently to be involved in hair morphogenesis (Fuchs et al 2001 , Schmidt-Ullrich and Paus 2005)
  • expression of LEF1 is decreased in Ebf+/- E2a+/- mice pro-B cells (O'Riordan and Grosschedl 1999).
  • EBF1 interacts with the transcription factor RUNX1 (Maier et al 2004) which has a role in hair morphogenesis (Raveh et al 2006). Those evidence make the EBF1 gene a very relevant candidate to play a role in hair follicle morphogenesis, and hence MPB.
  • EDA The anhidrotic ectodermal dysplasia (EDA) gene encodes various isoforms of ectodysplasin.
  • EDA-A1 and EDA-A2 are two of the 8 isoforms of EDA.
  • EDA-A1 is the longest form and encodes a 391 -residue protein with a domain similar to TNF at the COOH terminus.
  • EDA-A1 binds specifically EDA1 R receptor. Except for a two-residue deletion (Glu308 and Val309 in this domain), EDA-A2 is identical to EDA-A1.
  • EDA-A2 is generated through the use of an alternative internal splice donor site.
  • EDA-A1 and EDA-A2 are 2 trimeric type Il membrane proteins both containing a short intracellular domain, transmembrane region, and extracellular portion with collagenous domain and a TNF ligand motif in the C terminal region. Cleavage of EDA-A1/2 by the furin-like enzyme leads to formation of soluble extracellular molecule which is able to interact with corresponding receptors and mediate the signal. Mutations in EDA give rise to a clinical syndrome "hypohydrotic ectodermal dysplasia" characterized by loss of hair, sweat glands, and teeth (OMIM 300451 ). Mutations in EDA are responsible for most of the clinical cases studied to date. In a minority of patients, this phenotype is due to mutations in a distinct gene termed downless in mice and EDAR in humans (or EDA1 R) (OMIM 604095).
  • the analysis is performed by looking for direct and indirect interactions. From this analysis 35 genes are mapped to the Ingenuity database and assigned to 1 genetic network as defined by IPA ( Figure 2). Table C contains information about the gene content of the network, as well as the top functions assigned to those biochemical pathways.
  • Network 1 b contains 35 original nodes and includes the 4 input genes (Figure 2).
  • Figure 2 For descriptions of the EDA2R, AR, EBF1 and EDA gene, please refer to text above about networks from direct analysis only.
  • Figure 3 below lists the symbols used in both figures 1 and 2.
  • nucleic acid sequences of the present invention may be derived from a variety of sources including DNA, miRNA, cDNA, synthetic DNA, synthetic RNA, derivatives, mimetics or combinations thereof. Such sequences may comprise genomic DNA, which may or may not include naturally occurring introns, genie regions, nongenic regions, and regulatory regions. Moreover, such genomic DNA may be obtained in association with promoter regions or poly (A) sequences. The sequences, genomic DNA, or cDNA may be obtained in any of several ways. Genomic DNA can be extracted and purified from suitable cells by means well known in the art. Alternatively, mRNA can be isolated from a cell and used to produce cDNA by reverse transcription or other means.
  • nucleic acids described herein are used in certain embodiments of the methods of the present invention for production of RNA, proteins or polypeptides, through incorporation into cells, tissues, or organisms.
  • DNA containing all or part of the coding sequence for the genes described in Tables 2-4, or the SNP markers described in Tables 5, 6.1 and 6.2, is incorporated into a vector for expression of the encoded polypeptide in suitable host cells.
  • the invention also comprises the use of the nucleotide
  • nucleic acids of the present invention find use as primers and templates for the recombinant production of baldness-associated peptides or polypeptides, for chromosome and gene mapping, to provide antisense sequences, for tissue distribution studies, to locate and obtain full length genes, to identify and obtain homologous sequences (wild-type and mutants), and in diagnostic applications.
  • an antisense nucleic acid or oligonucleotide is wholly or partially complementary to, and can hybridize with, a target nucleic acid (either DNA or RNA) having the sequence of SEQ ID NO:1 , NO:3 or any SEQ ID from Tables 2-5, 6.1 and 6.2.
  • a target nucleic acid either DNA or RNA
  • an antisense nucleic acid or oligonucleotide comprising 16 nucleotides can be sufficient to inhibit expression of at least one gene from Tables 2-4.
  • an antisense nucleic acid or oligonucleotide can be complementary to 5 1 or 3' untranslated regions, or can overlap the translation initiation codon (5 1 untranslated and translated regions) of at least one gene from Tables 2-4, or its functional equivalent.
  • the antisense nucleic acid is wholly or partially complementary to, and can hybridize with, a target nucleic acid that encodes a polypeptide from a gene described in Tables 2-4.
  • oligonucleotides can be constructed which will bind to duplex nucleic acid (i.e., DNA:DNA or DNA:RNA), to form a stable triple helix containing or triplex nucleic acid.
  • duplex nucleic acid i.e., DNA:DNA or DNA:RNA
  • triplex oligonucleotides can inhibit transcription and/or expression of a gene from Tables 2-4, or its functional equivalent (M. D. Frank-Kamenetskii et a/., 1995).
  • Triplex oligonucleotides are constructed using the basepairing rules of triple helix formation and the nucleotide sequence of the genes described in Tables 2-4.
  • oligonucleotide refers to naturally-occurring species or synthetic species formed from naturally-occurring subunits or their close homologs.
  • the term may also refer to moieties that function similarly to oligonucleotides, but have non-naturally-occurring portions.
  • oligonucleotides may have altered sugar moieties or inter-sugar linkages. Exemplary among these are phosphorothioate and other sulfur containing species which are known in the art.
  • At least one of the phosphodiester bonds of the oligonucleotide has been substituted with a structure that functions to enhance the ability of the compositions to penetrate into the region of cells where the RNA whose activity is to be modulated is located. It is preferred that such substitutions comprise phosphorothioate bonds, methyl phosphonate bonds, or short chain alkyl or cycloalkyl structures.
  • the phosphodiester bonds are substituted with structures which are, at once, substantially non-ionic and ⁇ on chira!, or with structures which are chira' and enantiomerically specific. Persons of ordinary skill in the art will be able to select other linkages for use in the practice of the invention.
  • Oligonucleotides may also include species that include at least some modified base forms. Thus, purines and pyrimidines other than those normally found in nature may be so employed. Similarly, modifications on the furanosyl portions of the nucleotide subunits may also be effected, as long as the essential tenets of this invention are adhered to. Examples of such modifications are 2'-O-alkyl- and T- halogen-substituted nucleotides. Some non-limiting examples of modifications at the 2' position of sugar moieties which are useful in the present invention include OH, SH, SCH3, F, OCH3, OCN, O(CH2), NH2 and O(CH2)n CH3, where n is from 1 to about 10.
  • oligonucleotides are functionally interchangeable with natural oligonucleotides or synthesized oligonucleotides, which have one or more differences from the natural structure All such analogs are comprehended by this invention so long as they function effectively to hybridize with at least one gene from Tables 2-4 DNA or RNA to inhibit the function thereof
  • the oligonucleotides in accordance with this invention preferably comprise from about 3 to about 50 subunits It is more preferred that such oligonucleotides and analogs comprise from about 8 to about 25 subunits and still more preferred to have from about 12 to about 20 subunits
  • a "subunit" is a base and sugar combination suitably bound to adjacent subunits through phosphodiester or other bonds
  • Antisense nucleic acids or oligonucleotides can be produced by standard techniques (see, e g , Shewmaker et al , U S Patent No 6,107,065)
  • the oligonucleotides used in accordance with this invention may be conveniently and routinely made through the well-known technique of solid phase synthesis Any other means for such synthesis may also be employed, however, the actual synthesis of the oligonucleotides is well within the abilities of the practitioner It is also well known to prepare other oligonucleotide such as phosphorothioates and alkylated derivative
  • oligonucleotides of this invention are designed to be hyb ⁇ dizable with RNA (e g , mRNA) or DNA from genes described in Tables 2-4
  • RNA e g , mRNA
  • an oligonucleotide e g , DNA oligonucleotide
  • an oligonucleotide that can hybridize to the translation initiation site of the mRNA of a gene described in Tables 2-4 can be used to prevent translation of the mRNA
  • oligonucleotides that bind to the double-stranded DNA of a gene from Tables 2-4 can be adm ⁇ n ⁇ s f ered
  • Such oligonucleotides can form a t ⁇ o'ev COiSt-Uc + and nh'bit the transcription of the DNA encoding polypeptides of the genes described
  • antisense oligonucleotides may be targeted to hybridize to the following regions mRNA cap region, translation initiation site, translational termination site, transcription initiation site, transcription termination site, polyadenylation signal, 3 1 untranslated region, 5' untranslated region, 5'cod ⁇ ng region, mid coding region, and 3' coding region
  • the complementary oligonucleotide is designed to hybridize to the most unique 5' sequence of a gene described in Tables 2-4, including any of about 15-35 nucleotides spanning the 5' coding sequence.
  • the antisense oligonucleotide can be synthesized, formulated as a pharmaceutical composition, and administered to a subject.
  • expression vectors derived from retroviruses, adenovirus, herpes or vaccinia viruses, or from various bacterial plasmids may be used for delivery of nucleotide sequences to the targeted organ, tissue or cell population.
  • Methods which are well known to those skilled in the art can be used to construct recombinant vectors which will express nucleic acid sequence that is complementary to the nucleic acid sequence encoding a polypeptide from the genes described in Tables 2-4. These techniques are described both in Sambrook et al., 1989 and in Ausubel et al., 1992.
  • expression of at least one gene from Tables 2-4 can be inhibited by transforming a cell or tissue with an expression vector that expresses high levels of untranslatable sense or antisense sequences. Even in the absence of integration into the DNA, such vectors may continue to transcribe RNA molecules until they are disabled by endogenous nucleases. Transient expression may last for a month or more with a nonreplicating vector, and even longer if appropriate replication elements are included in the vector system.
  • Various assays may be used to test the ability of gene-specific antisense oligonucleotides to inhibit the expression of at least one gene from Tables 2-4.
  • mRNA levels of the genes described in Tables 2-4 can be assessed by Northern blot analysis (Sambrook et al., 1989; Ausubel et al., 1992; J. C. Alwine et al. 1977; I. M. Bird, 1998), quantitative or semi-quantitative RT-PCR analysis (see, e.g., W.M. Freeman et al., 1999; Ren et al., 1998; J. M. CaIe er al., 1998), or in situ hybridization (reviewed by A.K. Raap, 1998).
  • antisense oligonucleotides may be assessed by measuring levels of the polypeptide from the genes described in Tables 2-4, e.g., by western blot analysis, indirect immunofluorescence and immunoprecipitation techniques (see, e.g., J. M. Walker, 1998, Protein Protocols on CD-ROM, Humana Press, Totowa, NJ). Any other means for such detection may also be employed, and is well within the abilities of the practitioner. Mapping Technologies
  • mapping technologies may be based on amplification methods, restriction enzyme cleavage methods, hybridization methods, sequencing methods, and cleavage methods using agents.
  • Amplification methods include: self sustained sequence replication (Guatelli et al., 1990), transcriptional amplification system (Kwoh et al., 1989), Q-Beta Replicase (Lizardi et al., 1988), isothermal amplification (e.g. Dean et al., 2002; and Hafner et al., 2001 ), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of ordinary skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low number.
  • Restriction enzyme cleavage methods include: isolating sample and control DNA, amplification (optional), digestion with one or more restriction endonucleases, determination of fragment length sizes by gel electrophoresis and comparing samples and controls. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA.
  • sequence specific ribozymes see, e.g., U.S. Pat. No. 5,498,531
  • DNAzyme e.g. U.S. Pat. No. 5,807,718 can be used to score for the presence of specific mutations by development or loss of a ribozyme or DNAzyme cleavage site.
  • SNPs and SNP maps of the invention can be identified or generate ⁇ Dy hybridizing sample nucleic acids, e.g., DNA or RNA, to high density arrays or bead arrays containing oligonucleotide probes corresponding to the SNPS of Tables 5, 6.1 and 6.2 (see the Affymetrix arrays and lllumina bead sets at www.affymetrix.com and www.illumina.com and see Cronin et al., 1996; or Kozal et al., 1996).
  • sample nucleic acids e.g., DNA or RNA
  • sequencing reactions can be used to directly sequence nucleic acids for the presence or the absence of one or more SNPs of Tables 5, 6.1 and 6.2.
  • Examples of sequencing reactions include those based on techniques developed by Maxam and Gilbert (1977) or Sanger (1977).
  • any of a variety of automated sequencing procedures can be utilized, including sequencing by mass spectrometry (see, e.g. PCT International Publication No. WO 94/16101 ; Cohen et al., 1996; and Griffin et a/., 1993), real-time pyrophosphate sequencing method (Ronaghi et a/., 1998; and Permutt et al., 2001 ) and sequencing by hybridization (see e.g. Drmanac et al., 2002).
  • RNA/RNA, DNA/DNA or RNA/DNA heteroduplexes Other methods of detecting SNPs include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA, DNA/DNA or RNA/DNA heteroduplexes (Myers et al., 1985).
  • mismatch cleavage starts by providing heteroduplexes formed by hybridizing (labeled) RNA or DNA containing a wild-type sequence with potentially mutant RNA or DNA obtained from a sample.
  • the double-stranded duplexes are treated with an agent who cleaves single- stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands.
  • RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with S1 nuclease to enzymatically digest the mismatched regions.
  • either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of a mutation or SNP (see, for example, Cotton et al., 1988; and Saleeba et al., 1992).
  • the control DNA or RNA can be labeled for detection.
  • the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping SNPs.
  • DNA mismatch repair enzymes
  • the mutY enzyme of E. coli c'eaves A at G/A mismatches (Hsu et a/ , 1994)
  • Other examples include, but are not limited to, the MutHLS enzyme complex of E. coli (Smith and Modrich Proc. 1996) and CeI 1 from the celery (Kulinski et al., 2000) both cleave the DNA at various mismatches.
  • a probe based on a polymorphic site corresponding to a SNP of Tables 5, 6.1 and 6.2 is hybridized to a cDNA or other DNA product from a test cell or cells.
  • the duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like (see, for example, U.S. Pat. No. 5,459,039).
  • the screen can be performed in vivo following the insertion of the heteroduplexes in an appropriate vector. The whole procedure is known to those ordinary skilled in the art and is referred to as mismatch repair detection (see e.g. Fakhrai-Rad et al., 2004).
  • alterations in electrophoretic mobility can be used to identify SNPs in a sample.
  • single strand conformation polymorphism SSCP
  • SSCP single strand conformation polymorphism
  • Single-stranded DNA fragments of case and control nucleic acids will be denatured and allowed to renature.
  • the secondary structure of single-stranded nucleic acids varies according to sequence. The resulting alteration in electrophoretic mobility enables the detection of even a single base change.
  • the DNA fragments may be labeled or detected with labeled probes.
  • RNA rather than DNA
  • the method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Kee et al., 1991 ).
  • the movement of mutant or wild-type fragments in a polyacrylamide gel containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al., 1985).
  • DGGE denaturing gradient gel electrophoresis
  • DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR.
  • a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum et al., 1987).
  • the mutant fragment is detected using denaturing HPLC (see e.g. Hoogendoorn et a/ . 2000)
  • oligonucleotide primers may be prepared in which the SNP is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al., 1986; Saiki et al., 1989). Such oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
  • the amplification, the allele-specific hybridization and the detection can be done in a single assay following the principle of the 5' nuclease assay (e.g. see Livak et al., 1995).
  • the associated allele, a particular allele of a polymorphic locus, or the like is amplified by PCR in the presence of both allele-specific oligonucleotides, each specific for one or the other allele.
  • Each probe has a different fluorescent dye at the 5' end and a quencher at the 3' end.
  • the Taq polymerase via its 5' exonuclease activity will release the corresponding dyes. The latter will thus reveal the genotype of the amplified product.
  • Hybridization assays may also be carried out with a temperature gradient following the principle of dynamic allele-specific hybridization or like e.g. Jobs et al., (2003); and Bourgeois and Labuda, (2004).
  • the hybridization is done using one of the two allele-specific oligonucleotides labeled with a fluorescent dye, an intercalating quencher under a gradually increasing temperature.
  • the probe is hybridized to both the mismatched and full-matched template.
  • the probe melts at a lower temperature when hybridized to the template with a mismatch.
  • the release of the probe is captured by an emission of the fluorescent dye, away from the quencher.
  • the probe melts at a higher temperature when hybridized to the template with no mismatch.
  • the temperature-dependent fluorescence signals therefore indicate the absence or presence of an associated allele, a particular allele of a polymorphic locus, or the like (e.g. Jobs et al., 2003).
  • the hybridization is done under a gradually decreasing temperature. In this case, both allele-specific oligonucleotides are hybridized to the template competitively. At high temperature none of the two probes are hybridized. Once the optimal temperature of the full-matched probe is reached, it hybridizes and leaves no target for the mismatched probe (e.g. Bourgeois and Labuda, 2004). In the latter case, if the allele-specific probes are differently labeled, then they are hybridized to a single PCR-amphfied target. If the probes are labeled with the same dye, then the probe cocktail is hybridized to twice to identical templates with only one labeled probes, different in the two cocktails, in the presence of the unlabeled competitive probe.
  • Oligonucleotides used as primers for specific amplification may carry the associated allele, a particular allele of a polymorphic locus, or the like, also referred to as "mutation" of interest in the center of the molecule, so that amplification depends on differential hybridization (Gibbs et al., 1989) or at the extreme 3' end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner, 1993).
  • amplification may also be performed using Taq ligase for amplification (Barany, 1991 ).
  • ligation will occur only if there is a perfect match at the 3' end of the 5' sequence making it possible to detect the presence of a known associated allele, a particular allele of a polymorphic locus, or the like at a specific site by looking for the presence or absence of amplification.
  • the products of such an oligonucleotide ligation assay can also be detected by means of gel electrophoresis.
  • the oligonucleotides may contain universal tags used in PCR amplification and zip code tags that are different for each allele. The zip code tags are used to isolate a specific, labeled oligonucleotide that may contain a mobility modifier (e.g. Grossman et al., 1994).
  • allele-specific elongation followed by ligation will form a template for PCR amplification.
  • elongation will occur only if there is a perfect match at the 3' end of the allele-specific oligonucleotide using a DNA polymerase.
  • This reaction is performed directly on the genomic DNA and the extension/ligation products are amplified by PCR.
  • the oligonucleotides contain universal tags allowing amplification at a high multiplex level and a zip code for SNP identification.
  • the PCR tags are designed in such a way that the two alleles of a SNP are amplified by different forward primers, each having a different dye.
  • the zip code tags are the same for both alleles of a given SNPs and they are used for hybridization of the PCR-amplified products to oligonucleotides bound to a solid support, chip, bead array or like.
  • Fan et al. Cold Spring Harbor Symposia on Quantitative Biology, Vol. LXVIII, pp. 69-78 2003.
  • Another alternative includes the single-bass extension'ligat ; oi assay ⁇ ,sirg s molecular inversion probe, consisting of a single, long oligonucleotide (see e.g. Hardenbol et al., 2003).
  • the oligonucleotide hybridizes on both side of the SNP locus directly on the genomic DNA, leaving a one-base gap at the SNP locus.
  • the gap- filling, one-base extension/ligation is performed in four tubes, each having a different dNTP.
  • the oligonucleotide is circularized whereas unreactive, linear oligonucleotides are degraded using an exonuclease such as exonuclease I of E. coli.
  • the circular oligonucleotides are then linearized and the products are amplified and labeled using universal tags on the oligonucleotides.
  • the original oligonucleotide also contains a SNP-specific zip code allowing hybridization to oligonucleotides bound to a solid support, chip, and bead array or like. This reaction can be performed at a high multiplexed level.
  • the associated allele, a particular allele of a polymorphic locus, or the like is scored by single-base extension (see e.g. U.S. Pat. No. 5,888,819).
  • the template is first amplified by PCR.
  • the extension oligonucleotide is then hybridized next to the SNP locus and the extension reaction is performed using a thermostable polymerase such as ThermoSequenase (GE Healthcare) in the presence of labeled ddNTPs. This reaction can therefore be cycled several times. The identity of the labeled ddNTP incorporated will reveal the genotype at the SNP locus.
  • the labeled products can be detected by means of gel electrophoresis, fluorescence polarization (e.g. Chen et al., 1999) or by hybridization to oligonucleotides bound to a solid support, chip, and bead array or like. In the latter case, the extension oligonucleotide will contain a SNP-specific zip code tag.
  • a SNP is scored by selective termination of extension.
  • the template is first amplified by PCR and the extension oligonucleotide hybridizes in vicinity to the SNP locus, close to but not necessarily adjacent to it.
  • the extension reaction is carried out using a thermostable polymerase such as Thermo Sequenase (GE Healthcare) in the presence of a mix of dNTPs and at least one ddNTP.
  • Thermo Sequenase GE Healthcare
  • Thermo Sequenase GE Healthcare
  • Thermo Sequenase GE Healthcare
  • the extension product can then be detected by means of gel electrophoresis, in which case the extension products need to be labeled, or by mass spectrometry (see e.g. Storm ef al., 2003).
  • SNPs are detected using an invasive cleavage assay (see U.S. Pat. No. 6,090,543).
  • oligonucleotides per SNP to interrogate but these are used in a two step-reaction. During the primary reaction, three of the designed oligonucleotides are first hybridized directly to the genomic DNA. One of them is locus- specific and hybridizes up to the SNP locus (the pairing of the 3' base at the SNP locus is not necessary).
  • the present invention provides methods for identifying agents that modulate the expression of a nucleic acid encoding a gene from Tables 2-4. Such methods may utilize any available means of monitoring for changes in the expression level of the nucleic acids of the invention.
  • an agent is said to modulate the expression of a nucleic acid of the invention if it is capable of up- or down- regulating expression of the nucleic acid in a cell.
  • Such cells can be obtained from any parts of the body such as the scalp, blood, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, genitalia, vessels and endothelium.
  • Some non-limiting examples of cells that can be used are: hair (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
  • hair follicle, bulb, root, shaft and papilla
  • scalp muscle cells
  • nervous cells blood and vessels
  • dermis dermis
  • epidermis and other skin cells cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
  • the expression of a nucleic acid encoding a gene of the invention in a cell or tissue sample is monitored directly by hybridization to the nucleic acids of the invention.
  • Cell lines or tissues are exposed to the agent to be tested under appropriate conditions and time and total RNA or mRNA is isolated by standart procedures such as those disclosed in Sambrook et al., (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press).
  • Probes to detect differences in RNA expression levels between cells exposed to the agent and control cells may be prepared as described above. Hybridization conditions are modified using known methods, such as those described by Sambrook et al., and Ausubel ef al., as required for each probe. Hybridization of total cellular RNA or RNA enriched for polyA RNA can be accomplished in any available format. For instance, total cellular RNA or RNA enriched for polyA RNA can be affixed to a solid support and the solid support exposed to at least one probe comprising at least one, or part of one of the sequences of the invention under conditions in which the probe will specifically hybridize.
  • nucleic acid fragments comprising at least one, or part of one of the sequences of the invention can be affixed to a solid support, such as a silicon chip or a porous glass wafer.
  • the chip or wafer can then be exposed to total cellular RNA or polyA RNA from a sample under conditions in which the affixed sequences will specifically hybridize to the RNA.
  • agents which up or down regulate expression are identified.
  • the present invention provides methods for identifying agents that modulate at least one activity of the proteins described in Tables 2-4. Such methods may utilize any means of monitoring or detecting the desired activity.
  • an agent is said to modulate the expression of a protein of the invention if it is capable of up- or down- regulating expression of the protein in a cell.
  • Such cells can be obtained from any parts of the body such as the scalp, blood, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, genitalia, vessels and endothelium.
  • Some non-limiting examples of cells that can be used are: hair (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
  • hair follicle, bulb, root, shaft and papilla
  • scalp muscle cells
  • nervous cells blood and vessels
  • dermis dermis
  • epidermis and other skin cells cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
  • the specific activity of a protein of the invention may be assayed in a cell population that has been exposed to the agent to be tested and compared to an unexposed control cell population may be assayed.
  • Cell lines or populations are exposed to the agent to be tested under appropriate conditions and time.
  • Cellular lysates may be prepared from the exposed cell line or population and a control, unexposed cell line or population. The cellular lysates are then analyzed with the probe.
  • Antibody probes can be prepared by immunizing suitable mammalian hosts utilizing appropriate immunization protocols using the proteins of the invention or antigen- containing fragments thereof. To enhance immunogenicity, these proteins or fragments can be conjugated to suitable carriers. Methods for preparing immunogenic conjugates with carriers such as BSA, KLH or other carrier proteins are well known in the art. In some circumstances, direct conjugation using, for example, carbodiimide reagents may be effective; in other instances linking reagents such as those supplied by Pierce Chemical Co. (Rockford, IL) may be desirable to provide accessibility to the hapten.
  • the hapten peptides can be extended at either the amino or carboxy terminus with a cysteine residue or interspersed with cysteine residues, for example, to facilitate linking to a carrier.
  • Administration of the immunogens is conducted generally by injection over a suitable time period and with use of suitable adjuvants, as is generally understood in the art.
  • suitable adjuvants as is generally understood in the art.
  • titers of antibodies are taken to determine adequacy of antibody formation. While the polyclonal antisera produced in this way may be satisfactory for some applications, for pharmaceutical compositions, use of monoclonal preparations is preferred.
  • Immortalized cell lines which secrete the desired monoclonal antibodies may be prepared using standard methods, see e.g., Kohler & Milstein (1992) or modifications which affect immortalization of lymphocytes or spleen cells, as is generally known.
  • the immortalized cell lines secreting the desired antibodies can be screened by immunoassay in which the antigen is the peptide hapten, polypeptide or protein.
  • the cells can be cultured either in vitro or by production in ascites fluid.
  • the desired monoclonal antibodies may be recovered from the culture supernatant or from the ascites supernatant.
  • Fragments of the monoclonal antibodies or the polyclonal antisera which contain the immunologically significant portion(s) can be used as antagonists, as well as the intact antibodies.
  • Use of immunologically reactive fragments, such as Fab or Fab' fragments, is often preferable, especially in a therapeutic context, as these fragments are generally less immunogenic than the whole immunoglobulin.
  • the antibodies or fragments may also be produced, using current technology, by recombinant means.
  • Antibody regions that bind specifically to the desired regions of the protein can also be produced in the context of chimeras derived from multiple species.
  • Antibody regions that bind specifically to the desired regions of the protein can also be produced in the context of chimeras from multiple species, for instance, humanized antibodies.
  • the antibody can therefore be a humanized antibody or a human antibody, as described in U.S. Patent 5,585,089 or Riechmann et al. (1988).
  • Agents that are assayed in the above method can be randomly selected or rationally selected or designed.
  • an agent is said to be randomly selected when the agent is chosen randomly without considering the specific sequences involved in the association of the a protein of the invention alone or with its associated substrates, binding partners, etc.
  • An example of randomly selected agents is the use of a chemical library or a peptide combinatorial library, or a growth broth of an organism.
  • an agent is said to be rationally selected or designed when the agent is chosen on a non-random basis which takes into account the sequence of the target site or its conformation in connection with the agent's action
  • Agents can be rationally selected or rationally designed by utilizing the peptide sequences that make up these sites
  • a rationally selected peptide agent can be a peptide whose ammo acid sequence is identical to or a derivative of any functional consensus site
  • the agents of the present invention can be, as examples, oligonucleotides, antisense polynucleotides, interfering RNA, peptides, peptide mimetics, antibodies, antibody fragments, small molecules, vitamin derivatives, as well as carbohydrates
  • Peptide agents of the invention can be prepared using standard solid phase (or solution phase) peptide synthesis methods, as is known in the art
  • the DNA encoding these peptides may be synthesized using commercially available oligonucleotide synthesis instrumentation and produced recombinantly
  • Another class of agents of the present invention includes antibodies or fragments thereof that bind to a protein encoded by a gene in Tables 2-4
  • Antibody agents can be obtained by immunization of suitable mammalian subjects with peptides, containing as antigenic regions, those portions of the protein intended to be targeted by the antibodies (see section above of antibodies as probes for standard antibody preparation methodologies)
  • the present invention includes peptide mimetics that mimic the three-dimensional structure of the protein encoded by a gene from Tables 2-4
  • Such peptide mimetics may have significant advantages over naturally occurring peptides 'nclud ⁇ n g for grea+er chemical stab'li+y enhanced pharmacological properties (half-life, absorption, potency, efficacy, etc ), altered specificity (e g , a broad-spectrum of biological activities), reduced antigenicity and others
  • mimetics are peptide-containing molecules that mimic elements of protein secondary structure
  • the underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions, such as those of antibody and antigen
  • a peptide mimetic is expected to permit molecular interactions similar to the natural molecule
  • peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those
  • Peptide mimetics that are structurally similar to therapeutically useful peptides may be used to produce an equivalent therapeutic or prophylactic effect.
  • peptide mimetics are structurally similar to a paradigm polypeptide (i.e., a polypeptide that has a biochemical property or pharmacological activity), but have one or more peptide linkages optionally replaced by a linkage using methods known in the art.
  • Labeling of peptide mimetics usually involves covalent attachment of one or more labels, directly or through a spacer (e.g., an amide group), to non-interfering position(s) on the peptide mimetic that are predicted by quantitative structure-activity data and molecular modeling.
  • Such non- interfering positions generally are positions that do not form direct contacts with the macromolecule(s) to which the peptide mimetic binds to produce the therapeutic effect.
  • Derivitization (e.g., labeling) of peptide mimetics should not substantially interfere with the desired biological or pharmacological activity of the peptide mimetic.
  • the use of peptide mimetics can be enhanced through the use of combinatorial chemistry to create drug libraries.
  • the design of peptide mimetics can be aided by identifying amino acid mutations that increase or decrease binding of the protein to its binding partners. Approaches that can be used include the yeast two hybrid method (see Chien et al., 1991 ) and the phage display method.
  • the two hybrid method detects protein-protein interactions in yeast (Fields et al., 1989).
  • the phage display method detects the interaction between an immobilized protein and a protein that is expressed on the surface of phages such as lambda and M13 (Amberg ef al., 1993; Hogrefe et al., 1993). These methods allow positive and negative selection for protein-protein interactions and the identification of the sequences that determine these interactions.
  • the present invention also relates to methods for diagnosing baldness or a related disorder, preferably androgenic alopecia, a disposition to such disorder, predisposition to such a disorder and/or disorder progression.
  • the steps comprise contacting a target sample with (a) nucleic molecule(s) or fragments thereof and comparing the concentration of individual mRNA(s) with the concentration of the corresponding mRNA(s) from at least one healthy donor.
  • samples are from any parts of the body such as the scalp, blood, dermis, epidermis and other skin cells, cutaneous surfaces, intert ⁇ gious areas, genitalia, vessels and endothelium
  • Some non-limiting examples of cells that can be used are hair (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and
  • RNA is obtained from cells according to standard procedures and, preferably, reverse-transcribed
  • a DNAse treatment in order to get rid of contaminating genomic DNA
  • Some non-limiting examples of cells that can be used are hair (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes
  • the nucleic acid molecule or fragment is typically a nucleic acid probe for hybridization or a primer for PCR
  • the person skilled in the art is in a position to design suitable nucleic acids probes based on the information provided in the Tables of the present invention
  • the target cellular component, / e mRNA, e g in skin, may be detected directly in situ, e g by in situ hybridization or it may be isolated from other cell components by common methods known to those skilled in the art before contacting with a probe Detection methods include Northern blot analysis, RNase protection, in situ methods e g in situ hybridization in vtm amplification r ⁇ e+hods (PCP LCR QRMA replicase or RNA-transc ⁇ ption/amplification (TAS, 3SR), reverse dot blot disclosed in EP-B10237362) and other detection assays that are known to those skilled in the art Products obtained by in vitro amplification can be detected according to established methods, e g by
  • the probes (or primers) (or, alternatively, the reverse-transcribed sample mRNAs) can be detectably labeled, for example, with a radioisotope, a bioluminescent compound, a chemiluminescent compound, a fluorescent compound, a metal chelate, or an enzyme
  • the present invention also relates to the use of the nucleic acid molecules or fragments described above for the preparation of a diagnostic composition for the diagnosis of baldness or a disposition to such a disorder.
  • the present invention also relates to the use of the nucleic acid molecules of the present invention for the isolation or development of a compound which is useful for therapy of baldness.
  • the nucleic acid molecules of the invention and the data obtained using said nucleic acid molecules for diagnosis of baldness might allow for the identification of further genes which are specifically dysregulated, and thus may be considered as potential targets for therapeutic interventions.
  • the invention further provides prognostic assays that can be used to identify subjects having or at risk of developing baldness.
  • a test sample is obtained from a subject and the amount and/or concentration of the nucleic acid described in Tables 2- 4 is determined; wherein the presence of an associated allele, a particular allele of a polymorphic locus, or the likes in the nucleic acids sequences of this invention (see SEQ ID from Tables 5, 6.1 and 6.2) can be diagnostic for a subject having or at risk of developing baldness.
  • a test sample refers to a biological sample obtained from a subject of interest.
  • a test sample can be a biological fluid, a cell sample, or tissue.
  • a biological fluid can be, but is not limited to saliva, serum, mucus, urine, stools, spermatozoids, vaginal secretions, lymph, amiotic liquid, pleural liquid and tears.
  • Cells can be, but are not limited to hair cells (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, bnge ⁇ ans cells, CD4 f and CD3+ T cells and lymphocytes
  • the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, polypeptide, nucleic acid such as antisense DNA or interfering RNA (RNAi), small molecule or other drug candidate) to treat baldness.
  • an agent e.g., an agonist, antagonist, peptidomimetic, polypeptide, nucleic acid such as antisense DNA or interfering RNA (RNAi), small molecule or other drug candidate
  • these assays can be used to predict whether an individual will have an efficacious response or will experience adverse events in response to such an agent.
  • such methods can be used to determine whether a subject can be effectively treated with an agent that modulates the expression and/or activity of a gene from Tables 2-4, or the nucleic acids described herein.
  • an association study may be performed to identify polymorphisms from Tables 5, 6.1 and 6.2 that are associated with a given response to the agent, e.g., an efficacious response or the likelihood of one or more adverse events.
  • one embodiment of the present invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with aberrant expression or activity of a gene from Tables 2-4, in which a test sample is obtained and nucleic acids or polypeptides from Tables 2-5, 6.1 and 6.2 are detected (e.g., wherein the presence of a particular level of expression of a gene from Tables 2-4 or a particular allelic variant of such gene, such as polymorphism from Tables 2-5, 6.1 and 6.2, is diagnostic for a subject that can be administered an agent to treat a disorder such as baldness).
  • the method includes obtaining a sample from a subject suspected of having baldness or an affected individual and exposing such sample to an agent.
  • the expression and/or activity of the nucleic acids and or genes of the invention are monitored before and after treatment with such agent to assess the effect of such agent. After analysis of the expression values, one skilled in the art can determine whether such agent can effectively treat such subject.
  • the method includes obtaining a sample from a subject having or susceptible to developing baldness and determining the allelic constitution of polymorphisms from Table 1 that are associated with a particular response to an agent. After analysis of the allelic constitution of the individual at the associated polymorphisms, one skilled in the art can determine whether such agent can effectively treat such subject.
  • the methods of the invention can also be used to detect genetic alterations in a gene from Tables 2-4, thereby determining if a subject with the lesioned gene is at risk for a disorder associated with baldness.
  • the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic alteration characterized by at least one alteration linked to or affecting the integrity of a gene from Tables 2-4 encoding a polypeptide or the misexpression of such gene.
  • such genetic alterations can be detected by ascertaining the existence of at least one of: (1 ) a deletion of one or more nucleotides from a gene from Tables 2-4; (2) an addition of one or more nucleotides to a gene from Tables 2-4; (3) a substitution of one or more nucleotides of a gene from Tables 2-4; (4) a chromosomal rearrangement of a gene from Tables 2-4; (5) an alteration in the level of a messenger RNA transcript of a gene from Tables 2-4; (6) aberrant modification of a gene from Tables 2-4, such as of the methylation pattern of the genomic DNA, (7) the presence of a non-wild type splicing pattern of a messenger RNA transcript of a gene from Tables 2-4; (8) inappropriate post- translational modification of a polypeptide encoded by a gene from Tables 2-4; and (9) alternative promoter use.
  • a preferred biological sample is a peripheral blood sample obtained by conventional means from a subject.
  • Another preferred biological sample is a buccal swab.
  • Other biological samples can be, but is not limited to, urine, stools, spermatozoids, vaginal secretions, lymph, amiotic liquid, pleural liquid and tears.
  • detection of the alteration involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Pat. Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et a/., 1988; and Nakazawa et al., 1994), the latter of which can be particularly useful for detecting point mutations in a gene from Tables 2-4 (see Abavaya et al., 1995).
  • PCR polymerase chain reaction
  • LCR ligation chain reaction
  • This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic DNA, mRNA, or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers which specifically hybridize to a gene from Tables 2-4 under conditions such that hybridization and amplification of the nucleic acid from Tables 2-4 (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample.
  • PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with some of the techniques used for detecting a mutation, an associated allele, a particular allele of a polymorphic locus, or the like described herein.
  • Alternative amplification methods include: self sustained sequence replication (Guatelli et al., 1990), transcriptional amplification system (Kwoh et al., 1989), Q-Beta Replicase (Lizardi et al., 1988), isothermal amplification (e.g. Dean ef a/., 2002); and Hafner et al.,
  • alterations in a gene from Tables 2-4, from a sample cell can be identified by identifying changes in a restriction enzyme cleavage pattern.
  • sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates a mutation(s), an associated allele, a particular allele of a polymorphic locus, or the like in the sample DNA.
  • sequence specific ribozymes see, e.g., U.S. Pat. No. 5,498,531 or DNAzyme e.g. U.S. Pat. No. 5,807,718) can be used to score for the presence of specific associated allele, a particular allele of a polymorphic locus, or the likes by development or loss of a ribozyme or DNAzyme cleavage site.
  • the present invention also relates to further methods for diagnosing baldness or a related disorder, preferably androgenic alopecia, a disposition to such disorder, predisposition to such a disorder and/or disorder progression.
  • the steps comprise contacting a target sample with (a) nucleic molecule(s) or fragments thereof and determining the presence or absence of a particular allele of a polymorphism that confers a disorder-related phenotype (e.g., predisposition to such a disorder and/or disorder progression).
  • the presence of at least one allele from Tables 5, 6.1 and 6.2 that is associated with baldness disorder ("associated allele"), at least 5 or 10 associated alleles from Tables 5, 6.1 and 6.2, at least 50 associated alleles from Tables 5, 6.1 and 6.2, at least 100 associated alleles from Tables 5, 6.1 and 6.2, or at least 200 associated alleles from Table 5-7 determined in the sample is an indication of baldness or a related disorder, a disposition or predisposition to such kinds of disorders, or a prognosis for such disorder progression.
  • Samples may be obtained from any parts of the body such as the scalp, blood, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, genitalia, vessels and endothelium.
  • Some non-limiting examples of cells that can be used are hair (follicle, bulb, root, shaft and papilla) cells, scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
  • alterations in a gene from Tables 2-4 can be identified by hybridizing sample and control nucleic acids, e.g., DNA or RNA, to high density arrays or bead arrays containing tens to thousands of oligonucleotide probes (Cronin et al., 1996; Kozal et al., 1996).
  • alterations in a gene from Tables 2-4 can be identified in two dimensional arrays containing light-generated DNA probes as described in Cronin et al., (1996). Briefly, a first hybridization array of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear arrays of sequential overlapping probes.
  • This step allows the identification of point mutations, associated alleles, particular alleles of a polymorphic locus, or the like.
  • This step is followed by a second hybridization array that allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants, mutations, alleles detected
  • Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene
  • any of a variety of sequencing reactions known in the art can be used to directly sequence a gene from Tables 2-4 and detect an associated allele, a particular allele of a polymorphic locus, or the like by comparing the sequence of the sample gene from Tables 2-4 with the corresponding wild-type (control) sequence
  • Examples of sequencing reactions include those based on techniques developed by Maxam and Gilbert (1977) or Sanger (1977)
  • any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays (Bio/Techniques 19 448, 1995) including sequencing by mass spectrometry (see, e g PCT International Publication No WO 94/16101 , Cohen et al , 1996, and Griffin ef al 1993), real-time pyrophosphate sequencing method (Ronaghi et al , 1998, and Permutt et al , 2001 ) and sequencing by hybridization (see e g Drmanac ef al , 2002)
  • RNA/RNA duplexes can be treated with RNase and DNA/DNA hybrids treated with S1 nuclease to enzymatically digest the mismatched regions
  • the imutY enzyme of E. coli cleaves A at G/A mismatches (Hsu ef a/., 1994).
  • Other examples include, but are not limited to, the MutHLS enzyme complex of E. coli (Smith and Modrich., 1996) and CeI 1 from the celery (Kulinski ef a/., 2000) both cleave the DNA at various mismatches.
  • a probe based on a gene sequence from Tables 2-4 is hybridized to a cDNA or other DNA product from a test cell or cells. The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected using electrophoresis protocols or the like.
  • the screen can be performed in vivo following the insertion of the heteroduplexes in an appropriate vector.
  • the whole procedure is known to those ordinary skilled in the art and is referred to as mismatch repair detection (see e.g. Fakhrai-Rad ef a/., 2004).
  • alterations in electrophoretic mobility can be used to identify an associated allele, a particular allele of a polymorphic locus, or the likes in genes from Tables 2-4.
  • SSCP single strand conformation polymorphism
  • Single- stranded DNA fragments of sample and control nucleic acids from Tables 2-4 will be denatured and allowed to renature.
  • the secondary structure of single-stranded nucleic acids varies according to sequence; the resulting alteration in electrophoretic mobility enables the detection of even a single base change.
  • the DNA fragments may be labeled or detected with labeled probes.
  • the sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence.
  • the method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Kee et al., 1991 ).
  • the movement of mutant or wild-type fragments in a polyacrylamide gel containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers ef a/., 1985).
  • DGGE denaturing gradient gel electrophoresis
  • DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR.
  • a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum ef al., 1987).
  • the mutant fragment is detected using denaturing HPLC (see e.g. Hoogendoorn et al., 2000).
  • oligonucleotide primers may be prepared in which the known associated allele, particular allele of a polymorphic locus, or the like is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al., 1986; Saiki ef a/., 1989).
  • Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different associated allele, a particular allele of a polymorphic locus, or the likes where the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
  • the amplification, the allele-specific hybridization and the detection can be done in a single assay following the principle of the 5' nuclease assay (e.g. see Livak ef al., 1995).
  • the associated allele, a particular allele of a polymorphic locus, or the like locus is amplified by PCR in the presence of both allele- specific oligonucleotides, each specific for one or the other allele.
  • Each probe has a different fluorescent dye at the 5' end and a quencher at the 3' end.
  • the Taq polymerase via its 5' exonuclease activity will release the corresponding dyes. The latter will thus reveal the genotype of the amplified product.
  • the hybridization may also be carried out with a temperature gradient following the principle of dynamic allele-specific hybridization or like (e.g. Jobs ef al., 2003; and Bourgeois and Labuda, 2004).
  • the hybridization is done using one of the two allele-specific oligonucleotides labeled with a fluorescent dye, an intercalating quencher under a gradually increasing temperature.
  • the probe is hybridized to both the mismatched and full-matched template.
  • the probe melts at a lower temperature when hybridized to the template with a mismatch.
  • the release of the probe is captured by an emission of the fluorescent dye, away from the quencher.
  • the probe melts at a higher temperature when hybridized to the template with no mismatch.
  • the temperature-dependent fluorescence signals therefore indicate the absence or presence of the associated allele, particular allele of a polymorphic locus, or the like (e.g. Jobs et al. supra).
  • the hybridization is done under a gradually decreasing temperature. In this case, both allele-specific oligonucleotides are hybridized to the template competitively. At high temperature none of the two probes is hybridized. Once the optimal temperature of the full-matched probe is reached, it hybridizes and leaves no target for the mismatched probe. In the latter case, if the allele-specific probes are differently labeled, then they are hybridized to a single PCR-amplified target. If the probes are labeled with the same dye, then the probe cocktail is hybridizes twice to identical templates with only one labeled probes, different in the two cocktails, in the presence of the unlabeled competitive probe.
  • allele specific amplification technology that depends on selective PCR amplification may be used in conjunction with the present invention.
  • Oligonucleotides used as primers for specific amplification may carry the associated allele, particular allele of a polymorphic locus, or the like of interest in the center of the molecule, so that amplification depends on differential hybridization (Gibbs et al., 1989) or at the extreme 3 1 end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner, 1993).
  • amplification may also be performed using Taq ligase for amplification (Barany, 1991 ). In such cases, ligation will occur only if there is a perfect match at the 3' end of the 5' sequence making it possible to detect the presence of a known associated allele, a particular allele of a polymorphic locus, or the like at a specific site by looking for the presence or absence of amplification.
  • oligonucleotide ligation assay can also be detected by means of gel electrophoresis.
  • the oligonucleotides may contain universal tags used in PCR amplification and zip code tags that are different for each allele.
  • the zip code tags are used to isolate a specific, labeled oligonucleotide that may contain a mobility modifier (e.g. Grossman ef a/., 1994).
  • allele-specific elongation followed by ligation will form a template for PCR amplification. In such cases, elongation will occur only if there is a perfect match at the 3' end of the allele-specific oligonucleotide using a DNA polymerase.
  • the oligonucleotides contain universal tags allowing amplification at a high multiplex level and a zip code for SNP identification
  • the PCR tags are designed in such a way that the two alleles of a SNP are amplified by different forward primers, each having a different dye
  • the zip code tags are the same for both alleles of a given SNP and they are used for hybridization of the PCR-amplified products to oligonucleotides bound to a solid support, chip, bead array or like
  • Fan et al Cold Spring Harbor Symposia on Quantitative Biology, VoI LXVIII, pp 69-78, 2003
  • Another alternative includes the single-base extension/ligation assay using a molecular inversion probe, consisting of a single, long oligonucleotide (see e g Hardenbol et al , 2003)
  • the oligonucleotide hybridizes on both side of the SNP locus directly on the genomic DNA, leaving a one-base gap at the SNP locus
  • the gap- filhng, one-base extension/ligation is performed in four tubes, each having a different dNTP
  • the oligonucleotide is circularized whereas unreactive, linear oligonucleotides are degraded using an exonulease such as exonuclease I of E coli
  • the circular oligonucleotides are then linearized and the products are amplified and labeled using universal tags on the oligonucleotides
  • the original oligonucleotide also contains a SNP-specific zip code allowing hybridization to oligonucleo
  • the associated allele, particular allele of a polymorphic locus, or the like is scored by single-base extension (see e g U S Pat No 5,888,819)
  • the template is first amplified by PCR
  • the extension oligonucleotide is then hybridized next to + he SNP locus and the extension reaction is performed LG'ng a thermostable polymerase such as ThermoSequenase (GE Healthcare) in the presence of labeled ddNTPs
  • This reaction can therefore be cycled several times
  • the identity of the labeled ddNTP incorporated will reveal the genotype at the SNP locus
  • the labeled products can be detected by means of gel electrophoresis, fluorescence polarization (e g Chen et al., 1999) or by hybridization to oligonucleotides bound to a solid support, chip, bead array or like In the latter case, the extension oligonucleotide will contain a SNP-specific
  • the variant is scored by selective termination of extension
  • the template is first amplified by PCR and the extension oligonucleotide hybridizes in vicinity to the SNP locus, close to but not necessarily adjacent to it
  • the extension reaction is carried out using a thermostable polymerase such as Thermo Sequenase (GE Healthcare) in the presence of a mix of dNTPs and at least one ddNTP.
  • Thermo Sequenase GE Healthcare
  • Thermo Sequenase GE Healthcare
  • Thermo Sequenase GE Healthcare
  • the extension product can then be detected by means of gel electrophoresis, in which case the extension products need to be labeled, or by mass spectrometry (see e.g. Storm et al., 2003).
  • the associated allele, particular allele of a polymorphic locus, or the like is detected using an invasive cleavage assay (see U.S. Pat. No. 6,090,543).
  • an invasive cleavage assay see U.S. Pat. No. 6,090,543
  • allele-specific oligonucleotides that hybridize in tandem to the locus-specific probe but also contain a 5' flap that is specific for each allele of the SNP.
  • this creates a structure that is recognized by a cleavase enzyme (U.S. Pat. No. 6,090,606) and the allele-specific flap is released.
  • the flap fragments hybridize to a specific cassette to recreate the same structure as above except that the cleavage will release a small DNA fragment labeled with a fluorescent dye that can be detected using regular fluorescence detector. In the cassette, the emission of the dye is inhibited by a quencher.
  • microsatellites can also be useful to detect the genetic predisposition of an individual to a given disorder.
  • Microsatellites consist of short sequence motifs of one or a few nucleotides repeated in tandem. The most common motifs are polynucleotide runs, dinucleotide repeats (particularly the CA repeats) and trinucleotide repeats. However, other types of repeats can also be used.
  • the microsatellites are very useful for genetic mapping because they are highly polymorphic in their length.
  • Microsatellite markers can be typed by various means, including but not limited to DNA PCR fragment sizing, oligonucleotide ligation assay and mass spectrometry.
  • the locus of the microsatellite is amplified by PCR and the size of the PCR fragment will be directly correlated to the length of the microsatellite repeat.
  • the size of the PCR fragment can be detected by regular means of gel electrophoresis.
  • the fragment can be labeled internally during PCR or by using end-labeled oligonucleotides in the PCR reaction (e.g. Mansfield et al., 1996).
  • the size of the PCR fragment is determined by mass spectrometry. In such a case, however, the flanking sequences need to be eliminated. This can be achieved by ribozyme cleavage of an RNA transcript of the microsatellite repeat (Krebs et al., 2001 ).
  • the microsatellite locus is amplified using oligonucleotides that include a T7 promoter on one end and a ribozyme motif on the other end. Transcription of the amplified fragments will yield an RNA substrate for the ribozyme, releasing small RNA fragments that contain the repeated region. The size of the latter is determined by mass spectrometry.
  • the flanking sequences are specifically degraded. This is achieved by replacing the dTTP in the PCR reaction by dUTP.
  • dUTP nucleosides are then removed by uracyl DNA glycosylases and the resulting basic sites are cleaved by either abasic endonucleases such as human AP endonuclease or chemical agents such as piperidine.
  • Bases can also be modified post- PCR by chemical agents such as dimethyl sulfate and then cleaved by other chemical agents such as piperidine (see e.g. Maxam and Gilbert, 1977; U.S. Pat. No. 5,869,242; and U.S. Patent pending serial No. 60/335,068).
  • an oligonucleotide ligation assay can be performed.
  • the microsatellite locus is first amplified by PCR.
  • different oligonucleotides can be submitted to ligation at the center of the repeat with a set of oligonucleotides covering all the possible lengths of the marker at a given locus (Zirvi ef al., 1999).
  • the methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid selected from the SEQ ID of Tables 2-5, 6.1 and 6.2, or antibody reagent described herein, which may be conveniently used, for example, in a clinical setting to diagnose patient exhibiting symptoms or a family history of a disorder or disorder involving abnormal activity of genes from Tables 2-4.
  • Method to treat an animal suspected of having baldness comprising at least one probe nucleic acid selected from the SEQ ID of Tables 2-5, 6.1 and 6.2, or antibody reagent described herein, which may be conveniently used, for example, in a clinical setting to diagnose patient exhibiting symptoms or a family history of a disorder or disorder involving abnormal activity of genes from Tables 2-4.
  • the present invention provides methods of treating a disorder associated with baldness by expressing in vivo the nucleic acids of at least one gene from Tables 2-4.
  • These nucleic acids can be inserted into any of a number of well-known vectors for the transfection of target cells and organisms as described below.
  • the nucleic acids are transfected into cells, ex vivo or in vivo, through the interaction of the vector and the target cell.
  • Non-viral vector delivery systems include DNA plasmids, naked nucleic acid, and nucleic acid complexed with a delivery vehicle such as a liposome.
  • Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell.
  • RNA or DNA viral based systems for the delivery of nucleic acids take advantage of highly evolved processes for targeting a virus to specific cells in the body and trafficking the viral payload to the nucleus.
  • Viral vectors can be administered directly to patients (in vivo) or they can be used to treat cells in vitro and the modified cells are administered to patients (ex vivo).
  • Conventional viral based systems for the delivery of nucleic acids could include retroviral, lentivirus, adenoviral, adeno-associated and herpes simplex virus vectors for gene transfer.
  • Viral vectors are currently the most efficient and versatile method of gene transfer in target cells and tissues. Integration in the host genome is possible with the retrovirus, lentivirus, and adeno-associated virus gene transfer methods, often resulting in long term expression of the inserted transgene. Additionally, high transduction efficiencies have been observed in many different cell types and target tissues.
  • Lentiviral vectors are retroviral vector that are able to transduce or infect non-dividing cells and typically produce high viral titers. Selection of a retroviral gene transfer system would therefore depend on the target tissue. Retroviral vectors are comprised of cis-acting long terminal repeats with packaging capacity for up to 6-10 kb of foreign sequence. The minimum cis-acting LTRs are sufficient for replication and packaging of the vectors, which are then used to integrate the therapeutic gene into the target cell to provide permanent transgene expression.
  • Widely used retroviral vectors include those based upon murine leukemia virus (MuLV), gibbon ape leukemia virus (GaLV), Simian lmmuno deficiency virus (SIV), human immuno deficiency virus (HIV), and combinations thereof (see, e.g., Buchscher ef al., 1992; Johann ef al., 1992; Sommerfelt ef al., 1990; Wilson et al., 1989; Miller et a/.,1999;and PCT/US94/05700).
  • MiLV murine leukemia virus
  • GaLV gibbon ape leukemia virus
  • SIV Simian lmmuno deficiency virus
  • HV human immuno deficiency virus
  • Adenoviral based systems are typically used.
  • Adenoviral based vectors are capable of very high transduction efficiency in many cell types and do not require cell division. With such vectors, high titer and levels of expression have been obtained. This vector can be produced in large quantities in a relatively simple system.
  • Adeno-associated virus (“AAV”) vectors are also used to transduce cells with target nucleic acids, e.g., in the in vitro production of nucleic acid ⁇ ard pept'des, and for in vivo and ⁇ x vivo gene therapy procedures (see, e.g., West ef al., 1987; U.S. Pat. No.
  • pLASN and MFG- S are examples are retroviral vectors that have been used in clinical trials (Dunbar ef al., 1995; Kohn ef al., 1995; Malech ef al., 1997).
  • PA317/pLASN was the first therapeutic vector used in a gene therapy trial (Blaese ef al., 1995). Transduction efficiencies of 50% or greater have been observed for MFG-S packaged vectors (Ellem ef al., 1997; and Dranoff et al., 1997).
  • Recombinant adeno-associated virus vectors are a promising alternative gene delivery systems based on the defective and nonpathogenic parvovirus adeno- associated type 2 virus. All vectors are derived from a plasmid that retains only the AAV 145 bp inverted terminal repeats flanking the transgene expression cassette. Efficient gene transfer and stable transgene delivery due to integration into the genomes of the transduced cell are key features for this vector system. (Wagner ef a/., 1998, Kearns ef a/1996).
  • Ad vectors Replication-deficient recombinant adenoviral vectors (Ad) are predominantly used in transient expression gene therapy; because they can be produced at high titer and they readily infect a number of different cell types. Most adenovirus vectors are engineered such that a transgene replaces the Ad E1 a, E1 b, and E3 genes; subsequently the replication defector vector is propagated in human 293 cells that supply deleted gene function in trans. Ad vectors can transduce multiple types of tissues in vivo, including nondividing, differentiated cells such as those found in the liver, kidney and muscle system tissues. Conventional Ad vectors have a large carrying capacity.
  • Ad vector An example of the use of an Ad vector in a clinical trial involved polynucleotide therapy for antitumor immunization with intramuscular injection (Sterman et al., 1998). Additional examples of the use of adenovirus vectors for gene transfer in clinical trials include Rosenecker ef al., 1996; Sterman ef a/., 1998; Welsh ef a/., 1995; Alvarez ef a/., 1997; Topf ef a/., 1998.
  • Packaging cells are used to form virus particles that are capable of infecting a host cell. Such cells include 293 cells, which package adenovirus, and ⁇ 2 cells or PA317 cells, which package retrovirus.
  • Viral vectors used in gene therapy are usually generated by producer cell line that packages a nucleic acid vector into a viral particle. The vectors typically contain the minimal viral sequences required for packaging and subsequent integration into a host, other viral sequences being replaced by an expression cassette for the protein to be expressed. The missing viral functions are supplied in trans by the packaging cell line. For example, AAV vectors used in gene therapy typically only possess ITR sequences from the AAV genome which are required for packaging and integration into the host genome.
  • Viral DNA is packaged in a cell line, which contains a helper plasmid encoding the other AAV genes, namely rep and cap, but lacking ITR sequences.
  • the cell line is also infected with adenovirus as a helper.
  • the helper virus promotes replication of the AAV vector and expression of AAV genes from the helper plasmid.
  • the helper plasmid is not packaged in significant amounts due to a lack of ITR sequences. Contamination with adenovirus can be reduced by, e.g., heat treatment to which adenovirus is more sensitive than AAV.
  • a viral vector is typically modified to have specificity for a given cell type by expressing a ligand as a fusion protein with a viral coat protein on the viruses outer surface.
  • the ligand is chosen to have affinity for a receptor known to be present on the cell type of interest.
  • Moloney murine leukemia virus can be modified to express human heregulin fused to gp70, and the recombinant virus infects certain human breast cancer cells expressing human epidermal growth factor receptor. This principle can be extended to other pairs of virus expressing a ligand fusion protein and target cell expressing a receptor.
  • filamentous phage can be engineered to display antibody fragments (e.g., Fab or Fv) having specific binding affinity for virtually any chosen cellular receptor.
  • antibody fragments e.g., Fab or Fv
  • Such vectors can be engineered to contain specific uptake sequences thought to favor uptake by specific target cells.
  • Gene therapy vectors can be delivered in vivo by administration to an individual patient, typically by systemic administration (e.g., intravenous, intraperitoneal, intramuscular, subdermai, or intracranial infusion) or topical application.
  • vectors can be delivered to cells ex vivo, such as cells explanted from an individual patient (e.g., lymphocytes, bone marrow aspirates, and tissue biopsy) or universal donor hematopoietic stem cells, followed by reimplantation of the cells into a patient, usually after selection for cells which have incorporated the vector.
  • Ex vivo cell transfection for diagnostics, research, or for gene therapy is well known to those of skill in the art.
  • cells are isolated from the subject organism, transfected with a nucleic acid (gene or cDNA), and re-infused back into the subject organism (e.g., patient).
  • a nucleic acid gene or cDNA
  • Various cell types suitable for ex vivo transfection are well known to those of skill in the art (see, e.g., Freshney et al., 1994; and the references cited therein for a discussion of how to isolate and culture cells from patients).
  • stem cells are used in ex vivo procedures for cell transfection and gene therapy
  • the advantage to using stem cells is that they can be differentiated into other cell types in vitro, or can be introduced into a mammal (such as the donor of the cells) where they will engraft in the bone marrow Methods for differentiating CD34+ cells in vitro into clinically important immune cell types using cytokines such a GM-CSF, IFN- ⁇ and TNF- ⁇ are known (see lnaba ef a/ , 1992)
  • Stem cells are isolated for transduction and differentiation using known methods
  • stem cells are isolated from bone marrow cells by panning the bone marrow cells with antibodies which bind unwanted cells, such as CD4+ and CD8+ (T cells), CD45+ (panB cells), GR-1 (granulocytes), and lad (differentiated antigen presenting cells)
  • Vectors e g , retroviruses, adenoviruses, liposomes, etc ) containing therapeutic nucleic acids can be also administered directly to the organism for transduction of cells in vivo Alternatively, naked DNA can be administered
  • nucleic acids from Tables 2-4 are administered in any suitable manner, preferably with the pharmaceutically acceptable carriers described above Suitable methods of administering such nucleic acids are available and well known to those of skill in the art, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route (see Samulski et al , 1989)
  • the present invention is not limited to any method of administering such nucleic acids but preferentially uses the methods described herein
  • the present invention further provides other methods of treating baldness such as administering to an individual having baldness an effective amount of an agent that regulates the expression, activity or physical state of at least one gene from Tables 2-4
  • An "effective amount" of an agent is an amount that modulates a level of expression or activity of a gene from Tables 2-4, in a cell in the individual at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80% or more, compared to a level of the respective gene from Tables 2-4 in a cell in the individual in the absence of the compound
  • the preventive or therapeutic agents of the present invention may be administered, either orally or parenterally, systemically or locally
  • intravenous injection such as drip infusion, intramuscular injection, intraperitoneal injection, subcutaneous injection, suppositories, intestinal lavage, oral enteric coated tablets, and the like can be selected, and the method of administration may be chosen, as appropriate, depending on the
  • the effective dosage is chosen from the range of 0.01 mg to 100 mg per kg of body weight per administration. Alternatively, the dosage in the range of 1 to 1000 mg, preferably 5 to 50 mg per patient may be chosen.
  • the therapeutic efficacy of the treatment may be monitored by observing various parts of the body, such as the scalp, by any monitoring method known in the art. Others ways of monitoring efficacy can be, but are not limited to monitoring hair growth, size of hair follicles, length and thickness of hair, activity of hair follicles, hair loss and status of hair at the crown of the head.
  • the present invention further provides a method of treating an individual clinically diagnosed with baldness.
  • the methods generally comprises analyzing a biological sample that includes a cell, in some cases, a skin cell, from an individual clinically diagnosed with baldness for the presence of modified levels of expression of at least 1 gene, at least 10 genes, at least 50 genes from Tables 2-4.
  • a treatment plan that is most effective for individuals clinically diagnosed as having a condition associated with baldness is then selected on the basis of the detected expression of such genes in a cell.
  • Treatment may include administering a composition that includes an agent that modulates the expression or activity of a protein from Tables 2-4 in the cell. Information obtained as described in the methods above can also be used to predict the response of the individual to a particular agent.
  • the invention further provides a method for predicting a patient's likelihood to respond to a drug treatment for a condition associated with baldness, comprising determining whether modified levels of a gene from Tables 2- 4 is present in a cell, wherein tne presence of protein is predictive of the patient's likelihood to respond to a drug treatment for the condition.
  • a method for predicting a patient's likelihood to respond to a drug treatment for a condition associated with baldness comprising determining whether modified levels of a gene from Tables 2- 4 is present in a cell, wherein tne presence of protein is predictive of the patient's likelihood to respond to a drug treatment for the condition.
  • Examples of the prevention or improvement of symptoms accompanied by baldness that can monitor for effectiveness include prevention or improvement of hair growth on the scalp, the size of hair follicles, length and thickness of hair, activity of hair follicles, hair loss and status of hair at the crown of the head.
  • the invention also provides a method of predicting a response to therapy in a subject having baldness by determining the presence or absence in the subject of one or more markers associated with baldness described in Tables 2-5, 6.1 and 6.2, diagnosing the subject in which the one or more markers are present as having baldness, and predicting a response to a therapy based on the diagnosis e.g., response to therapy may include an efficacious response and/or one or more adverse events.
  • the invention also provides a method of optimizing therapy in a subject having baldness by determining the presence or absence in the subject of one or more markers associated with a clinical subtype of baldness, diagnosing the subject in which the one or more markers are present as having a particular clinical subtype of baldness, and treating the subject having a particular clinical subtype of baldness based on the diagnosis.
  • baldness can be treated with surgical procedures, such as hair transplants and scalp reduction.
  • Another treatment is the topical use of progesterone, a molecule similar to testosterone. The side effects of this treatment are not known and again this treatment has not proven to be efficient on a vast majority of balding individuals.
  • Example 1 Identification of cases and controls
  • the Quebec founder population has two distinct advantages over general populations for LD mapping. Because it is relatively young, about 12 to 15 generations from mid 17th century to present, and because it has a limited but sufficient number of founders, approximately 2600 effective founders (Charbonneau et al. 1987), the Quebec population is characterized both by extended LD and by decreased genetic heterogeneity. The increased extent of LD allows the detection of disorder genes using a reasonable marker density, while still allowing the increased meiotic resolution of population-based mapping.
  • the number of founders is small enough to result in increased LD and reduced allelic heterogeneity, yet large enough to insure that all of the major disorder genes involved in general populations are present in Quebec.
  • Reduced allelic heterogeneity will act to increase relative risk imparted by the remaining alleles and so increase the power of case/control studies to detect gene an associated allele, a particular allele of a polymorphic locus, or the likes involved in complex disorders within the Quebec population.
  • the specific combination of age in generations, optimal number of founders and large present population size makes the QFP optimal for LD-based gene mapping. The existence of close family relationships among the selected individuals is verified. When subjects are linked by a common relative, according to the information available, only one is kept f or the study.
  • Patient inclusion criteria for the study included diagnosis for baldness with the onset of the condition occurring before age 25.
  • the extraction method yielded high molecular weight DNA, and the quality of every DNA sample was verified by agarose gel electrophoresis. Genomic DNA appeared on the gel as a large band of very high molecular weight. The remaining two buffy coats were stored at -80 0 C as backups.
  • the samples were collected as 468 male patients (cases) and 470 male controls.
  • the DNA extracted from cases and control samples is pooled together in various pools. Pools are designed to segregate probands (cases) from controls.
  • Proband pools consisting of DNA extracted from recruited cases, were separated according to level of baldness as determined on the Norwood/Hamilton scale (Norwood 1975), and further separated by age group.
  • Six proband pools consisted of Norwood/Hamilton type Vl, and 2 proband pools consisted of Norwood/Hamilton type VII.
  • Genotyping was performed using Perlegen's ultra-high-throughput platform. Loci of interest were amplified and hybridized to wafers containing arrays of oligonucleotides. Allele discrimination was performed through allele-specific hybridization. In total, 248,535 SNPs, spread over 3 microarrays, were genotyped.
  • This set of markers contained the QLDM (Quebec LD Map), a map created specifically for the Quebec founder population, which possesses a base density of one marker per 40 kb and up to one marker per 10 kb in low-LD regions, the lower the LD is in a given area, the higher the marker density will be.
  • QLDM Quebec LD Map
  • the QLDM markers and other markers were selected from various databases including the -1.6 million SNP database of Perlegen Life Sciences (Patil, 2001 ), the hapmap consortium database and dbSNP at NCBI.
  • the SNPs were chosen to maximize uniformity of genetic coverage and as much as possible, with a minor allele frequency of 10% or higher.
  • the genotyping information was entered into a Unified Genotype Database (a proprietary database under development) from which it was accessed using custom-built programs for export to the genetic analysis pipeline. Analyses of these genotypes were performed with the statistical tools described in Example 3.
  • the GWS permitted the identification of 65 candidate regions that are further analyzed by the Fine Mapping approache described below. Four candidate regions out of the 65 were presently confirmed use the Fine Mapping approach described below. The rest of the candidate regions Fine Mapping is ongoing.
  • Example 2 The raw data generated by the GWS approach (Example 2 herein) was analyzed by various means to identify candidate regions (see also Fine Mapping described in Example 5).
  • the data analysis process compares the relative fluorescence intensities of features corresponding to the reference allele of a given SNP with those corresponding to the alternate allele, to calculate a p-hat value.
  • the latter is proportional to the fluorescence signal from perfect match features for the reference allele divided by the sum of fluorescence signals from perfect match features for the reference plus the alternate alleles.
  • P-hat assumes values close to 1 (typically 0.9) for pure reference samples and close to 0 (typically 0.1 ) for pure alternate samples, and can be used as a measured estimate of the reference allele frequency of a SNP in a DNA pool.
  • delta p-hat The difference between case and controi poois, delta p-hat, is calcuiaied using the weig ⁇ ted average of case and control p-hats. Delta p-hat is a reliable estimate of the allele frequency difference between the cases and controls.
  • the data consisted of 8 pools of cases and 8 pools of controls, with a p-hat value for each pool (provided by Perlegen's analysis methods described above).
  • the weighted mean p-hat value for all case pools was calculated as 2- ⁇ ⁇ ⁇ Z- ⁇ ⁇ where Ni is the number of alleles (2 X the number of people) in each case pool and the weighted
  • p-hat value for all control pools were calculated as Z-. y J 2-, i where Li is the number of alleles in each control pool.
  • the estimate of delta-phat, the difference in allele frequency between cases and controls was X - Y and the significance of delta-phat was X - Y
  • T X ( / A 1 ⁇ 1 1 ) ⁇ N , ( Phat , ⁇ *J and
  • Y 1 Yin/ 3 Workers, 14th edition Hafner Press N.Y. 1970 pp 99-100) where ⁇ *-> ' with 2k degrees of freedom where Pi is the P value for association of each of k markers within the sliding window.
  • the combined P values identify regions of multiple single marker associations.
  • Fisner ' s method assumes independence of association for each marker which id known not to be the case because of strong LD between adjacent markers. Therefore the magnitude of the combined P value was affected both by LD and association and these cannot be taken at face value.
  • the combined P values were best used as a means to identify regions of multi-marker association which can then be assessed on the basis of the magnitude of single marker association.
  • Fine mapping genotyping all cases and controls samples individually
  • Fine mapping is carried out using the lllumina BeadStation 500GX SNP genotyping platform.
  • Alleles are genotyped using an allele-specific elongation assay that involves ligation to a locus-specific oligonucleotide. The assay is performed directly on genomic DNA at a highly multiplex level and the products are amplified using universal oligonucleotides.
  • a set of SNP markers is selected with an average inter-marker distance varying with the mean extent of LD throughout the region as determined by delta-M (I M), where M is the number of markers present in the 300 kb window centered at each reference marker is defined as the square root of the average r2 or ⁇ 2ij measures of LD between all ( ⁇ M(M-1 ) ⁇ /2) pairwise comparisons of all (M) markers within the 300 kb window (Dawson et a/., 2002).
  • Regions with a signal harbouring a high -Log10 P value and with mean delta-M of 0.3 or below are mapped with a target density of one marker per 10 kb.
  • Regions showing a signal with a high -Log10 P value and with mean delta-M between 0.3 and 0.35 as well as selected regions with a signal with a lower -Log10 P value and a delta-M value below 0.35 are mapped with a target density of one marker per 10-20 kb.
  • the principle is that low-LD regions will be mapped at a higher SNP density.
  • Selected regions with a delta-M value above 0.35 are mapped with a density of one marker per 20-30 kb, including the markers used in the GWS.
  • the selected regions are delimited by the location where the LDSTATS -Log10 P values reach the background level.
  • the cohort consisted of 652 cases and 652 controls (these included the cases and controls used in the GWS and other cases and controls recruited following the GWS genotyping using the same criteria described in Example 1 ).
  • a unique consensus sequence was constructed for each splice variant and a trained reviewer assessed each alignment. This assessment included examination of all putative splice junctions for consensus splice donor/acceptor sequences, putative start codons, consensus Kozak sequences and upstream in-frame stops, and the location of polyadenylation signals. In addition, conserved noncoding sequences (CNSs) that could potentially be involved in regulatory functions were included as important information for each gene. The genomic reference and exon sequences were then archived for future reference. A master assembly that included all splice variants, exons and the genomic structure was used in subsequent analyses (i.e., analysis of polymorphisms).
  • the UniGene database contains information regarding the tissue source for ESTs and cDNAs contributing to individual clusters. This information was extracted and summarized to provide an indication in which tissues the gene was expressed. Particular emphasis was placed on annotating the tissue source for bona fide ESTs, since many ESTs mapped to Unigene clusters are artifactual.
  • SAGE and microarray data also curated at NCBI (Gene Expression Omnibus), provided information on expression profiles for individual genes. Particular emphasis was placed on identifying genes that were expressed in tissues known to be involved in the pathophysiology of baldness.
  • Polymorphisms identified in candidate genes are evaluated for potential function. Initially, polymorphisms are examined for potential impact upon encoded proteins. If the protein is a member of a gene family with reported 3- dimensional structural information, this information is used to predict the location of the polymorphism with respect to protein structure. This information provided insight into the potential role of polymorphisms in altering protein or ligand interactions, as well as suitability as a drug target. In a second phase of analysis we evaluate the potential role of polymorphisms in other biological phenomena, including regulation of transcription, splicing and mRNA stability, etc. There are many examples of the functional involvement of naturally occurring polymorphisms in these processes. As part of this analysis, polymorphisms located in promoter or other regulatory elements, canonical splice sites, exonic and intronic splice enhancers and repressors, conserved noncoding sequences and UTRs are localized.
  • Candidate genes and regions are selected for sequencing in order to identify all polymorphisms. In cases where the critical interval, identified by fine mapping, was relatively small (-50 kb), the entire region, including all introns, is sequenced to identify polymorphisms, in situations where the region is large (>50 kb), candidate genes are prioritized for sequencing, and/or only functional gene elements (promoters, exons and splice sites) are sequenced.
  • the samples to be sequenced are selected according to which haplotypes contribute to the association signal observed in the region.
  • the purpose is to select a set of samples that covered all the major haplotypes in the given region. Each major haplotype must be present in a few copies.
  • the first step therefore consisted of determining the major haplotypes in the region to be sequenced.
  • each major haplotype and haplotype subset are represented in at least two to four copies
  • the protocol includes the following steps, once a region is delimited
  • the design of the primers is performed using a proprietary primer design tool A primer quality control is included in the primer design process Primers that successfully passed the control quality process were synthesized by Integrated DNA Technologies (IDT)
  • IDT Integrated DNA Technologies
  • the sense and anti-sense ohgos are separated such that the sense oligos are placed on one plate in the same position as their anti-sense counterparts are on another plate
  • Two additional plates are created from each storage plate, one for use in PCR and the other for sequencing
  • the sense and anti-sense oligos of the same pair are combined in the same well to achieve a final concentration of 1 5 ⁇ M for each oligonucleotide
  • PCR conditions are optimized by testing a variety of conditions that included varying salt concentrations and temperatures, as well as including various additives PCR products are checked for robust amplification and minimal background by agarose gel electrophoresis
  • PCR products to be used for sequencing are amplified using the conditions chosen during optimization
  • the PCR products are purified free of salts, dNTPs and unincorporated primers by use of a MultiScreen PCR384 filter plate manufactured by Milhpore Following PCR, the amplicons are quantified by use of a lambda/Hind III standard curve This is done to ensure that the quantity of PCR product required for sequencing had been generated.
  • the raw data was measured against the standard curve data in Excel by use of a macro.
  • Sequencing of PCR products is performed by DNA Landmarks using ABI 3730 capillary sequencing instruments.
  • the ABI Prism SeqScape software (Applied Biosystems) is used for SNP identification.
  • the chromatogram trace files were imported into a SeqScape sequencing project and the base calling is automatically performed. Sequences are then aligned and compared to each other using the SeqScape program.
  • the base calling is checked manually, base by base; editing was performed if needed.
  • genotyping assays may need to be utilized based on the type of polymorphism identified (i.e., SNP, indel, microsatellite).
  • the assay type can be, but is not restricted to, Sentrix Assay Matrix on lllumina BeadStations, microsatellite on MegaBACE, SNP on ABI or Orchid.
  • the frequencies of genotypes and haplotypes in cases and controls are analyzed in a similar manner as the GWS and fine mapping data.
  • polymorphisms are identified that increase an individual's susceptibility to baldness.
  • the goal of ultra-fine mapping is to identify the polymorphism that is most associated with disorder phenotype as part of the search for the actual DNA polymorphism that confers susceptibility to disorder. This statistical identification may need to be corroborated by functional studies.
  • Example 8 Confirmation of Candidate regions and genes in a general population
  • Example 7 The confirmation of any putative associations described in Example 7 is performed in an independent general population patient sample. These DNA samples consist of at least 400 male controls and 400 male patients with baldness.

Abstract

Abstract The present invention relates to the selection of a set of SNP markers for use in genome wide association studies based on linkage disequilibrium mapping. In particular, the invention relates to the fields of pharmacogenomics, diagnostics, patient therapy and the use of genetic haplotype information to predict an individual's susceptibility to Male Pattern Baldness and/or their response to a particular drug or drugs.

Description

GENEMAP OF THE HUMAN GENES ASSOCIATED WITH MALE PATTERN
BALDNESS
INVENTORS: Abdelmajid Belouchi, John Verner Raelson, Walter Edward Bradley, Bruno Paquin, Quynh Nguyen-Huu, Pascal Croteau, Rene Allard, Sophie Debrus, Randall David Little, Paul Van Eerdewegh, Jonathan Segal and Tim Keith.
PRIORITY
This application claims priority to US Provisional Application No. 60/855,415 filed October 31 , 2006, which is hereby incorporated by reference in its entirety.
FIELD OF THE INVENTION
The invention relates to the field of genomics and genetics, including genome analysis and the study of DNA variations. In particular, the invention relates to the fields of pharmacogenomics, diagnostics, patient therapy and the use of genetic haplotype information to predict an individual's susceptibility to baldness and/or their response to a particular drug or drugs, so that drugs tailored to genetic differences of population groups may be developed and/or administered to the appropriate population.
The invention also relates to a GeneMap for baldness, which links variations in DNA (including both genie and non-genic regions) to an individual's susceptibility to baldness and/or response to a particular drug or drugs. The invention further relates to the genes disclosed in the GeneMap (see Tables 2-4), which is related to methods and reagents for detection of an individual's increased or decreased risk for baldness by identifying at least one polymorphism in one or a combination of the genes from the GeneMap. Also related are the candidate regions identified in Table 1 , which are associated with baldness. In addition, the invention further relates to nucleotide sequences of those genes including genomic DNA sequences, cDNA sequences, single nucleotide polymorphisms (SNPs), alleles and haplotypes (see Sequence Listing and Tables 5, 6.1 and 6.2).
The invention further relates to isolated nucleic acids comprising these nucleotide sequences and isolated polypeptides or peptides encoded thereby. Also related are expression vectors and host cells comprising the disclosed nucleic acids or fragments thereof, as well as antibodies that bind to the encoded polypeptides or peptides. The present invention further relates to ligands that modulate the activity of the disclosed genes or gene products. In addition, the invention relates to diagnostics and therapeutics for baldness, utilizing the disclosed nucleic acids, SNPs, chromosomal regions, gene maps, polypeptides or peptides, antibodies and/or ligands and small molecules that activate or repress relevant signaling events.
BACKGROUND OF THE INVENTION
With age, both men and women lose hair density and this gradual thinning of the hair results in baldness in a number of cases. Baldness affects a large proportion of the population; about 35% of men begin to bald by the time they are 35 years old, and about two-thirds are either bald or have a balding pattern by age 60. Although inherited baldness affects more men than women, the incidence of baldness in women is significant, since it amounts to a third or half of that in men before menopause, and increases greatly after that. Baldness is neither physically disabling nor a life-threatening disease, and is only of cosmetic importance, but it may profoundly affect self-esteem and/or cause psychological stress and anxiety.
Changes that contribute to the development of baldness include alterations in the growth cycle of hair. Hair typically progresses through cycles comprising three phases: anagen (active hair growth), catagen (transition phase), and telogen (resting phase during which the hair shaft is shed prior to new growth). As baldness progresses, there is a shift in the percentages of hair follicles in each phase, with the majority shifting from anagen to telogen. The size of hair follicles is also known to decrease while the total number remains relatively constant. Baldness results when the oid shaft is no longer replaced. In most cases, the hair follicle remains alive and the potential for hair re-growth is preserved.
Since there is no clear explanation to the development of baldness, effective treatments for baldness has thus been limited. Current treatments include the administration of 1 ) nitroxides, such as Minoxidil and Nicorandil 2) antiandrogens, such as Proscar, Cyoctal and spironolactone and 3) superoxide dismutase mimetics. While progress has been made in the stimulation of hair-growth by drug treatment, none of the available treatments is completely satisfactory and most of them have undesirable associated side effects, such as hair growth in undesirable places. Most of the treatments are also temporary and hair loss resumes if the treatment is stopped. Alternative solutions include invasive procedures such as hair transplantation and scalp reduction, which are expensive, time-consuming and may be very painful. Invasive procedures are also not an option for certain baldness cases.
Despite many efforts in past studies, the role of hormones in regulating the hair cycle is not yet thoroughly understood. The role of androgens and other intrinsic factors is not yet elucidated. Despite a preponderance of evidence showing inheritance of a risk for baldness through epidemiological studies and linkage and association analyses, the genes affecting baldness have yet to be discovered. There is a need in the art for identifying specific genes related to baldness in order to enable the development of therapeutics that address the causes of the trait. The failure in past studies to identify causative genes in complex genetic traits, such as baldness, has been due to the lack of appropriate methods to detect a sufficient number of variations in genomic DNA samples (markers), the insufficient quantity of necessary markers available, and the number of needed individuals to enable such a study. The present invention addresses these issues.
The DNA sequences between two human genomes are 99.9% identical. The variations in DNA sequence between individuals can be as an example, deletions of small or large stretches of DNA, insertions of stretches of DNA, variations in the number of repetitive DNA elements, and changes in single base positions in the genome called "single nucleotide polymorphisms" (SNPs). Human DNA sequence variation accounts for a large fraction of observed differences between individuals, including susceptibility to disorder.
Many genetic traits, like baldness, are complex genetic traits and are believed to involve several genes rather than single genes, as observed in rare disorders. This makes detection of any particular gene substantially more difficult than in a rare disorder, where a single gene mutation that segregates according to a Mendelian inheritance pattern is the causative mutation. Any one of the multiple interacting gene mutations involved in the etiology of a complex and common disorder will impart a lower relative risk for the disorder than will the single gene mutation involved in a simple genetic disorder. Low relative risk alleles are more difficult to detect and, as a result, the success of positional cloning using linkage mapping that has been achieved for simple genetic disorder genes has not been repeated for complex disorders. Several approaches have been proposed to discover and characterize multiple genes in complex genetic traits. These gene discovery methods can be subdivided into hypothesis-free disorder association studies and hypothesis-driven candidate gene or region studies. The candidate gene approach relies on the analysis of a gene in patients who have a disorder in which the gene is thought to play a role. This approach is limited in utility because it only provides for the investigation of genes with known functions. Although variant sequences of candidate genes may be identified using this approach, it is inherently limited by the fact that variant sequences in other genes that contribute to the phenotype will be necessarily missed when the technique is employed. Recently, the first genome-wide scan (GWS) has been shown to be efficient in identifying baldness susceptibility markers, such as the androgen receptor on chromosome X (Hillmer et al., 2005). In contrast to the candidate gene approach, a GWS searches throughout the genome without any a priori hypothesis and consequently can identify genes that are not obvious candidates for the disorder as well as genes that are relevant candidates for the disorder, as well as chromosomal regions that are structurally important where an "associated allele", a "particular allele of a polymorphic locus", or the likes can influence the expression of specific genes.
Family-based linkage mapping methods were initially used for disorder locus identification. This technique locates genes based on the relatively limited number of genetic recombination events within the families used in the study, and results in large chromosomal regions containing hundreds of genes, any one of which could be the disorder-causing gene. Population-based, or linkage disequilibrium (LD) mapping is based on the premise that regions adjacent to a gene of interest are co-transmitted through the generations along with the gene. As a result, LD extends over shorter genetic regions than does linkage (Hewett et al., 2002), and can facilitate detection of genes with lower relative risk than family linkage mapping approaches. It also defines much smaller candidate regions which may contain only a few genes, making the identification of the actual disorder gene much easier.
It has been estimated that a GWS that uses a general population and case/control association (LD) analysis would require approximately 700,000 SNP markers (Carlson et al., 2003). The cost of a GWS at this marker density for a sufficient sample size for statistical power is economically prohibitive. The use of a special founder population (genetic isolates), such as the French Canadian population of Quebec, is one solution to the problem with LD analysis. The French Canadian population in Quebec (Quebec Founder Population - QFP) provides one of the best resources in the world for gene discovery based on its high levels of genetic sharing and genetic homogeneity. By combining DNA collected from the QFP, high throughput genotyping capabilities and proprietary algorithms for genetic analysis, a comprehensive genome-wide association study was facilitated. The present invention relates specifically to a set of baldness- causing genes (GeneMap) and targets which present attractive points of therapeutic intervention.
In view of the foregoing, identifying susceptibility genes associated with baldness and their respective biochemical pathways will facilitate the development of effective treatments for baldness and hair loss, and will permit the identification of diagnostic markers which will predict the propensity for balding and permit therapeutic intervention before baldness can occur. The identification of those genetic markers would provide the basis for novel genetic tests and eliminate or reduce the therapeutic methods currently used. The identification of those genetic markers will also provide the development of effective therapeutic intervention for a much greater proportion of the individuals affected by baldness than the current methods, which are effective only in a small number of baldness cases. The present invention satisfies this need and provides related advantages as well.
DESCRIPTION OF THE FILES CONTAINED ON THE CD-R
The sequence listing submitted concurrently herewith on compact disc under 37 C. F. R. §§1.821 (c) and 1.821(e) is herein incorporated by reference in its entirety. Two duplicate copies of the CD-R labeled "copy 1" and "copy 2" are submitted herewith. The material on each of the duplicate CD-R is identical. Thus, descriptions or references herein to the CD-R labeled "GeneMap of the human gene associated with Male Pattern Baldness" and the files contained thereon apply to both "copy 1" and "copy 2", and both are herein incorporated by reference in their entirety.
The CD-R labeled "GeneMap of the human gene associated with Male Pattern Baldness" contains the following one file of sequence listing. DEFINITIONS
Throughout the description of the present invention, several terms are used that are specific to the science of this field. For the sake of clarity and to avoid any misunderstanding, these definitions are provided to aid in the understanding of the specification and claims:
Allele: One of a pair, or series, of forms of a gene or non-genic region that occur at a given locus in a chromosome. Alleles are symbolized with the same basic symbol {e.g., B for dominant and b for recessive; B1 , B2, Bn for n additive alleles at a locus). In a normal diploid cell there are two alleles of any one gene (one from each parent), which occupy the same relative position (locus) on homologous chromosomes. Within a population there may be more than two alleles of a gene. See multiple alleles. SNPs also have alleles, i.e., the two (or more) nucleotides that characterize the SNP.
Amplification of nucleic acids: refers to methods such as polymerase chain reaction (PCR), ligation amplification (or ligase chain reaction, LCR) and amplification methods based on the use of Q-beta replicase. These methods are well known in the art and are described, for example, in U.S. Patent Nos. 4,683,195 and 4,683,202. Reagents and hardware for conducting PCR are commercially available. Primers useful for amplifying sequences from the disorder region are preferably complementary to, and preferably hybridize specifically to, sequences in the disorder region or in regions that flank a target region therein. Genes from Tables 2-4 generated by amplification may be sequenced directly. Alternatively, the amplified sequence(s) may be cloned prior to sequence analysis.
Antigenic component, is a moiety tnat binds to its specific aruiQody witn sufficiently high affinity to form a detectable antigen-antibody complex.
Antibodies: refer to polyclonal and/or monoclonal antibodies and fragments thereof, and immunologic binding equivalents thereof, that can bind to proteins and fragments thereof or to nucleic acid sequences from the disorder region, particularly from the disorder gene products or a portion thereof. The term antibody is used both to refer to a homogeneous molecular entity, or a mixture such as a serum product made up of a plurality of different molecular entities. Proteins may be prepared synthetically in a protein synthesizer and coupled to a carrier molecule and injected over several months into rabbits. Rabbit sera are tested for immunoreactivity to the protein or fragment. Monoclonal antibodies may be made by injecting mice with the proteins, or fragments thereof Monoclonal antibodies will be screened by ELISA and tested for specific immunoreactivity with protein or fragments thereof (Harlow et al 1988, Antibodies A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY) These antibodies will be useful in assays as well as therapeutics
Associated allele refers to an allele at a polymorphic locus that is associated with a particular phenotype of interest, e g , a predisposition to a disorder or a particular drug response
Baldness encompasses the complete or partial loss of hair and a variety of different types of alopecia (e g , alopecia areata, alopecia totalis, alopecia universalis, alopecia diffusa, alopeciapartialis and androgenic alopecia) due to intrinsic factors (e g , aging, hormone production and/or hormonal imbalances, pregnancy, etc ) or genetic factors, as well as disease- or extrinsic factors-related hair loss (e g , thyroid function imbalance, autoimmune disorders, stress, vitamin deficiency and/or other dietary imbalances, chemotherapy, radiotherapy or other treatments, etc ) Baldness of the scalp is characterized by, e g , loss of activity and/or miniaturization of hair follicles hair loss, slowing of hair growth, thinning of the hair, appearance of shorter and weaker hairs, as well as any of a number of characteristic structural and/or molecular features In the context of the present invention, baldness refers to all the stages of the process, e g , receding hairline, thinning of hair, loss of hair at the crown of the head, hair-loss in a typical M-shaped pattern which eventually results in a loss of hair over the top of the head, complete hair loss, etc In the present invention, baldness also encompasses transitional regions, which refer to those regions of the scalp of a subject experiencing ha>r loss and/or developing baldness that are at an intemediate state in the process (individuals who are developing baldness) In such transitional regions hair loss is important but hair follicles retain some activity and hair is still present (although it may be significantly thinner)
cDNA refers to complementary or copy DNA produced from an RNA template by the action of RNA-dependent DNA polymerase (reverse transcriptase) Thus, a cDNA clone means a duplex DNA sequence complementary to an RNA molecule of interest, included in a cloning vector or PCR amplified This term includes genes from which the intervening sequences have been removed cDNA library: refers to a collection of recombinant DNA molecules containing cDNA inserts that together comprise essentially all of the expressed genes of an organism or tissue. A cDNA library can be prepared by methods known to one skilled in the art (see, e.g., Cowell and Austin, 1997, "DNA Library Protocols," Methods in Molecular Biology). Generally, RNA is first isolated from the cells of the desired organism, and the RNA is used to prepare cDNA molecules.
Cloning: refers to the use of recombinant DNA techniques to insert a particular gene or other DNA sequence into a vector molecule. In order to successfully clone a desired gene, it is necessary to use methods for generating DNA fragments, for joining the fragments to vector molecules, for introducing the composite DNA molecule into a host cell in which it can replicate, and for selecting the clone having the target gene from amongst the recipient host cells.
Cloning vector: refers to a plasmid or phage DNA or other DNA molecule that is able to replicate in a host cell. The cloning vector is typically characterized by one or more endonuclease recognition sites at which such DNA sequences may be cleaved in a determinable fashion without loss of an essential biological function of the DNA, and which may contain a selectable marker suitable for use in the identification of cells containing the vector.
Coding sequence or a protein-coding sequence: is a polynucleotide sequence capable of being transcribed into mRNA and/or capable of being translated into a polypeptide or peptide. The boundaries of the coding sequence are typically determined by a translation start codon at the 5'-terminus and a translation stop codon at the 3'-terminus.
Complement of a nucleic acid sequence: refers to the antisense sequence that participates in Watson-Crick base-pairing with the original sequence.
Disorder region (or Trait region): refers to the portions of the human chromosomes displayed in Table 1 bounded by the markers from Tables 2-5, 6.1 and 6.2.
Disorder-associated nucleic acid or polypeptide sequence (or Trait-associated nucleic acid or polypeptide sequence): refers to a nucleic acid sequence that maps to region of Table 1 or the polypeptides encoded therein (Tables 2-5, 6.1 and 6.2 SNPs, nucleic acids, and polypeptides). For nucleic acids, this encompasses sequences that are identical or complementary to the gene sequences from Tables 2-4, as well as sequence-conservative, function-conservative, and non-conservative variants thereof. For polypeptides, this encompasses sequences that are identical to the polypeptide, as well as function-conservative and non-conservative variants thereof. Included are the alleles of naturally-occurring polymorphisms causative of baldness such as, but not limited to, alleles that cause altered expression of genes of Tables 2-4 and alleles that cause altered protein levels or stability (e.g., decreased levels, increased levels, expression in an inappropriate tissue type, increased stability, and decreased stability.
Expression vector: refers to a vehicle or plasmid that is capable of expressing a gene that has been cloned into it, after transformation or integration in a host cell. The cloned gene is usually placed under the control of (i.e., operably linked to) a regulatory sequence.
Function-conservative variants: are those in which a change in one or more nucleotides in a given codon position results in a polypeptide sequence in which a given amino acid residue in the polypeptide has been replaced by a conservative amino acid substitution. Function-conservative variants also include analogs of a given polypeptide and any polypeptides that have the ability to elicit antibodies specific to a designated polypeptide.
Founder population: Also called a population isolate, this is a large number of people who have mostly descended, in genetic isolation from other populations, from a much smaller number of people who lived many generations ago.
Gene: Refers to a DNA sequence that encodes through its template or messenger RNA a sequence of amino acids characteristic of a specific peptide, polypeptide, or protein. The term "gene" also refers to DNA sequence that encodes an RNA product. The term gene as used herein with reference to genomic DNA includes intervening, non-coding regions, as well as regulatory regions, and can include 5' and 3' ends. A gene sequence is wild-type if such sequence is usually found in individuals unaffected by the disorder or condition of interest. However, environmental factors and other genes can also play an important role in the ultimate determination of the disorder. In the context of complex disorders involving multiple genes (oligogenic disorder), the wild type, or normal sequence can also be associated with a measurable risk or susceptibility, receiving its reference status based on its frequency in the general population.
GeneMaps are defined as groups of gene(s) that are directly or indirectly involved in at least one phenotype of a disorder. As such, GeneMaps enable the development of synergistic diagnostic products, creating "theranostics". Genotype: Set of alleles at a specified locus or loci.
Haplotype: The allelic pattern of a group of (usually contiguous) DNA markers or other polymorphic loci along an individual chromosome or double helical DNA segment. Haplotypes identify individual chromosomes or chromosome segments. The presence of shared haplotype patterns among a group of individuals implies that the locus defined by the haplotype has been inherited, identical by descent (IBD), from a common ancestor. Detection of identical by descent haplotypes is the basis of linkage disequilibrium (LD) mapping. Haplotypes are broken down through the generations by recombination and mutation. In some instances, a specific allele or haplotype may be associated with susceptibility to a disorder or condition of interest, e.g., baldness. In other instances, an allele or haplotype may be associated with a decrease in susceptibility to a disorder or condition of interest, i.e., a protective sequence.
Host: includes prokaryotes and eukaryotes. The term includes an organism or cell that is the recipient of an expression vector (e.g., autonomously replicating or integrating vector).
Hybridizable: nucleic acids are hybridizable to each other when at least one strand of the nucleic acid can anneal to another nucleic acid strand under defined stringency conditions. In some embodiments, hybridization requires that the two nucleic acids contain at least 10 substantially complementary nucleotides; depending on the stringency of hybridization, however, mismatches may be tolerated. The appropriate stringency for hybridizing nucleic acids depends on the length of the nucleic acids and the degree of complementarity, and can be determined in accordance with the methods described herein.
Identity by descent (IBD): Identity among DNA sequences for different individuals that is due to the fact that they have all been inherited from a common ancestor. LD mapping identifies IBD haplotypes as the likely location of disorder genes shared by a group of patients.
Identity: as known in the art, is a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, as determined by comparing the sequences. In the art, identity also means the degree of sequence relatedness between polypeptide or polynucleotide sequences, as the case may be, as determined by the match between strings of such sequences. Identity and similarity can be readily calculated by known methods, including but not limited to those described in A.M. Lesk (ed), 1988, Computational Molecular Biology, Oxford University Press, NY; D.W. Smith (ed), 1993, Biocomputing. Informatics and Genome Projects, Academic Press, NY; A.M. Griffin and H. G. Griffin, H. G (eds), 1994, ComputerAnalysis of Sequence Data, Part 1 , Humana Press, NJ; G. von Heinje, 1987, Sequence Analysis in Molecular Biology, Academic Press; and M. Gribskov and J. Devereux (eds), 1991 , Sequence Analysis Primer, M Stockton Press, NY; H. Carillo and D. Lipman, 1988, SIAM J. Applied Math., 48:1073.
Immunogenic component: is a moiety that is capable of eliciting a humoral and/or cellular immune response in a host animal.
Isolated nucleic acids: are nucleic acids separated away from other components (e.g., DNA, RNA, and protein) with which they are associated (e.g., as obtained from cells, chemical synthesis systems, or phage or nucleic acid libraries). Isolated nucleic acids are at least 60% free, preferably 75% free, and most preferably 90% free from other associated components. In accordance with the present invention, isolated nucleic acids can be obtained by methods described herein, or other established methods, including isolation from natural sources (e.g., cells, tissues, or organs), chemical synthesis, recombinant methods, combinations of recombinant and chemical methods, and library screening methods.
Isolated polypeptides or peptides: are those that are separated from other components (e.g., DNA, RNA, and other polypeptides or peptides) with which they are associated (e.g., as obtained from cells, translation systems, or chemical synthesis systems). In a preferred embodiment, isolated polypeptides or peptides are at least 10% pure; more preferably, 80% or 90% pure. Isolated polypeptides and peptides include those obtained by methods described herein, or other established methods, including isolation from natural sources (e.g., cells, tissues, or organs), chemical synthesis, recombinant methods, or combinations of recombinant and chemical methods. Proteins or polypeptides referred to herein as recombinant are proteins or polypeptides produced by the expression of recombinant nucleic acids. A portion as used herein with regard to a protein or polypeptide, refers to fragments of that protein or polypeptide. The fragments can range in size from 5 amino acid residues to all but one residue of the entire protein sequence. Thus, a portion or fragment can be at least 5, 5-50, 50-100, I00-200, 200-400, 400-800, or more consecutive amino acid residues of a protein or polypeptide. Linkage disequilibrium (LD): the situation in which the alleles for two or more loci do not occur together in individuals sampled from a population at frequencies predicted by the product of their individual allele frequencies. In other words, markers that are in LD do not follow Mendel's second law of independent random segregation. LD can be caused by any of several demographic or population artifacts as well as by the presence of genetic linkage between markers. However, when these artifacts are controlled and eliminated as sources of LD, then LD results directly from the fact that the loci involved are located close to each other on the same chromosome so that specific combinations of alleles for different markers (haplotypes) are inherited together. Markers that are in high LD can be assumed to be located near each other and a marker or haplotype that is in high LD with a genetic trait can be assumed to be located near the gene that affects that trait. The physical proximity of markers can be measured in family studies where it is called linkage or in population studies where it is called linkage disequilibrium.
LD mapping: population based gene mapping, which locates disorder genes by identifying regions of the genome where haplotypes or marker variation patterns are shared statistically more frequently among disorder patients compared to healthy controls. This method is based upon the assumption that many of the patients will have inherited an allele associated with the disorder from a common ancestor (IBD), and that this allele will be in LD with the disorder gene.
Locus: a specific position along a chromosome or DNA sequence. Depending upon context, a locus could be a gene, a marker, a chromosomal band or a specific sequence of one or more nucleotides.
Minor allele frequency (MAF): the population frequency of one of the alleles for a given polymorphism, which is equal or less than 50%. The sum of the MAF and the Major allele frequency equals one.
Markers: an identifiable DNA sequence that is variable (polymorphic) for different individuals within a population. These sequences facilitate the study of inheritance of a trait or a gene. Such markers are used in mapping the order of genes along chromosomes and in following the inheritance of particular genes; genes closely linked to the marker or in LD with the marker will generally be inherited with it. Two types of markers are commonly used in genetic analysis, microsatellites and SNPs. Microsatellite: DNA of eukaryotic cells comprising a repetitive, short sequence of DNA that is present as tandem repeats and in highly variable copy number, flanked by sequences unique to that locus.
Mutant sequence: if it differs from one or more wild-type sequences. For example, a nucleic acid from a gene listed in Tables 2-4 containing a particular allele of a single nucleotide polymorphism may be a mutant sequence. In some cases, the individual carrying this allele has increased susceptibility toward the disorder/trait or condition of interest. In other cases, the mutant sequence might also refer to an allele that decreases the susceptibility toward a disorder/trait or condition of interest and thus acts in a protective manner. The term mutation may also be used to describe a specific allele of a polymorphic locus.
Non-conservative variants: are those in which a change in one or more nucleotides in a given codon position results in a polypeptide sequence in which a given amino acid residue in a polypeptide has been replaced by a non-conservative amino acid substitution. Non-conservative variants also include polypeptides comprising non- conservative amino acid substitutions.
Nucleic acid or polynucleotide: purine- and pyrimidine-containing polymers of any length, either polyribonucleotides or polydeoxyribonucleotide or mixed polyribo polydeoxyribonucleotides. This includes single-and double-stranded molecules, i.e., DNA-DNA, DNA-RNA and RNA-RNA hybrids, as well as protein nucleic acids (PNA) formed by conjugating bases to an amino acid backbone. This also includes nucleic acids containing modified bases.
Nucleotide: a nucleotide, the unit of a DNA molecule, is composed of a base, a 2 - deoxyribose and phosphate ester(s) attached at the 5' carbon of the deoxyribose. For its incorporation in DNA, the nucleotide needs to possess three phosphate esters but it is converted into a monoester in the process.
Operably linked: means that the promoter controls the initiation of expression of the gene. A promoter is operably linked to a sequence of proximal DNA if upon introduction into a host cell the promoter determines the transcription of the proximal DNA sequence(s) into one or more species of RNA. A promoter is operably linked to a DNA sequence if the promoter is capable of initiating transcription of that DNA sequence. Ortholog: denotes a gene or polypeptide obtained from one species that has homology to an analogous gene or polypeptide from a different species.
Paralog: denotes a gene or polypeptide obtained from a given species that has homology to a distinct gene or polypeptide from that same species.
Phenotype: any visible, detectable or otherwise measurable property of an organism such as symptoms of, or susceptibility to, a disorder.
Polymorphism: occurrence of two or more alternative genomic sequences or alleles between or among different genomes or individuals at a single locus. A polymorphic site thus refers specifically to the locus at which the variation occurs. In some cases, an individual carrying a particular allele of a polymorphism has an increased or decreased susceptibility toward a disorder or condition of interest.
Portion and fragment: are synonymous. A portion as used with regard to a nucleic acid or polynucleotide refers to fragments of that nucleic acid or polynucleotide. The fragments can range in size from 8 nucleotides to all but one nucleotide of the entire gene sequence. Preferably, the fragments are at least about 8 to about 10 nucleotides in length; at least about 12 nucleotides in length; at least about 15 to about 20 nucleotides in length; at least about 25 nucleotides in length; or at least about 35 to about 55 nucleotides in length.
Probe or primer: refers to a nucleic acid or oligonucleotide that forms a hybrid structure with a sequence in a target region of a nucleic acid due to complementarity of the probe or primer sequence to at least one portion of the target region sequence.
Protein and polypeptide: are synonymous. Peptides are defined as fragments or portions of polypeptides, preferably fragments or portions having at least one functional activity (e.g., proteolysis, adhesion, fusion, antigenic, or intracellular activity) as the complete polypeptide sequence.
Recombinant nucleic acids: nuclei acids which have been produced by recombinant DNA methodology, including those nucleic acids that are generated by procedures which rely upon a method of artificial replication, such as the polymerase chain reaction (PCR) and/or cloning into a vector using restriction enzymes. Portions of recombinant nucleic acids which code for polypeptides can be identified and isolated by, for example, the method of M. Jasin et al., U.S. Patent No. 4,952,501. Regulatory sequence: refers to a nucleic acid sequence that controls or regulates expression of structural genes when operably linked to those genes. These include, for example, the lac systems, the trp system, major operator and promoter regions of the phage lambda, the control region of fd coat protein and other sequences known to control the expression of genes in prokaryotic or eukaryotic cells. Regulatory sequences will vary depending on whether the vector is designed to express the operably linked gene in a prokaryotic or eukaryotic host, and may contain transcriptional elements such as enhancer elements, termination sequences, tissue-specificity elements and/or translational initiation and termination sites.
Sample: as used herein refers to a biological sample, such as, for example, tissue or fluid isolated from an individual or animal (including, without limitation, plasma, serum, cerebrospinal fluid, lymph, tears, nails, hair, saliva, milk, pus, and tissue exudates and secretions) or from in vitro cell culture-constituents, as well as samples obtained from, for example, a laboratory procedure.
Single nucleotide polymorphism (SNP): variation of a single nucleotide. This includes the replacement of one nucleotide by another and deletion or insertion of a single nucleotide. Typically, SNPs are biallelic markers although tri- and tetra-allelic markers also exist. For example, SNP A\C may comprise allele C or allele A (Tables 5, 6.1 and 6.2). Thus, a nucleic acid molecule comprising SNP A\C may include a C or A at the polymorphic position. For a combination of SNPs, the term "haplotype" is used, e.g. the genotype of the SNPs in a single DNA strand that are linked to one another. In certain embodiments, the term "haplotype" is used to describe a combination of SNP alleles, e.g., the alleles of the SNPs found together on a single DNA molecule. In specific embodiments, the SNPs in a haplotype are in linkage disequilibrium with one another.
Sequence-conservative: variants are those in which a change of one or more nucleotides in a given codon position results in no alteration in the amino acid encoded at that position (Ae., silent mutation).
Substantially homologous: a nucleic acid or fragment thereof is substantially homologous to another if, when optimally aligned (with appropriate nucleotide insertions and/or deletions) with the other nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least 60% of the nucleotide bases, usually at least 70%, more usually at least 80%, preferably at least 90%, and more preferably at least 95-98% of the nucleotide bases. Alternatively, substantial homology exists when a nucleic acid or fragment thereof will hybridize, under selective hybridization conditions, to another nucleic acid (or a complementary strand thereof). Selectivity of hybridization exists when hybridization which is substantially more selective than total lack of specificity occurs. Typically, selective hybridization will occur when there is at least about 55% sequence identity over a stretch of at least about nine or more nucleotides, preferably at least about 65%, more preferably at least about 75%, and most preferably at least about 90% (M. Kanehisa, 1984, NucL Acids Res. 11 :203-213). The length of homology comparison, as described, may be over longer stretches, and in certain embodiments will often be over a stretch of at least 14 nucleotides, usually at least 20 nucleotides, more usually at least 24 nucleotides, typically at least 28 nucleotides, more typically at least 32 nucleotides, and preferably at least 36 or more nucleotides.
Wild-type gene from Tables 2-4: refers to the reference sequence. The wild-type gene sequences from Tables 2-4 used to identify the variants (single nucleotide polymorphisms, alleles, and haplotypes) described in detail herein.
Technical and scientific terms used herein have the meanings commonly understood by one of ordinary skill in the art to which the present invention pertains, unless otherwise defined. Reference is made herein to various methodologies known to those of skill in the art. Publications and other materials setting forth such known methodologies to which reference is made are incorporated herein by reference in their entireties as though set forth in full. Standard reference works setting forth the general principles of recombinant DNA technology include J. Sambrook et a., 1989, Molecular Cloning: A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY; P. B. Kaufman ef a/., (eds), 1995, Handbook of Molecular and Cellular Methods in Biology and Medicine, CRC Press, Boca Raton, M.J McPherson (ed), 1991 , Directed Mutagenesis: A Practical Approach, IRL Press, Oxford; J. Jones, 1992, Amino Acid and Peptide Synthesis, Oxford Science Publications, Oxford; B. M. Austen and O. M. R. Westwood, 1991 , Protein Targeting and Secretion, IRL Press, Oxford; D.N Glover (ed), 1985, DNA Cloning, Volumes I and 11 ; M.J. Gait (ed), 1984, Oligonucleotide Synthesis; B. D. Hames and SJ. Higgins (eds), 1984, Nucleic Acid Hybridization; Quirke and Taylor (eds), 1991 , PCR-A Practical Approach; Harries and Higgins (eds), 1984, Transcription and Translation; R.I. Freshney (ed), 1986, Animal Cell Culture; Immobilized Cells and Enzymes, 1986, IRL Press; Perbal, 1984, A Practical Guide to Molecular Cloning, J. H. Miller and M. P. Calos (eds), 1987, Gene Transfer Vectors for Mammalian Cells, Cold Spring Harbor Laboratory Press; M.J. Bishop (ed), 1998, Guide to Human Genome Computing, 2d Ed., Academic Press, San Diego, CA; L. F. Peruski and A.H. Peruski, 1997, The Internet and the New Biology. Tools for Genomic and Molecular Research, American Society for Microbiology, Washington, D. C. Standard reference works setting forth the general principles of immunology include S. Sell, 1996, Immunology, lmmunopathology & Immunity, 5th Ed., Appleton & Lange, Publ., Stamford, CT; D. Male et ai, 1996, Advanced Immunology, 3d Ed., Times Mirror Int'l Publishers Ltd., Publ., London; D. P. Stites and A.L Terr, 1991 , Basic and Clinical Immunology, 7th Ed., Appleton & Lange, Publ., Norwalk, CT; and A.K. Abbas ef a/., 1991 , Cellular and Molecular Immunology, W. B. Saunders Co., Publ., Philadelphia, PA. Any suitable materials and/or methods known to those of skill can be utilized in carrying out the present invention; however, preferred materials and/or methods are described. Materials, reagents, and the like to which reference is made in the following description and examples are generally obtainable from commercial sources, and specific vendors are cited herein.
DETAILED DESCRIPTION OF THE INVENTION
General Description of baldness trait
One of the most prevalent types of baldness is male pattern baldness (MPB) also known as androgenic alopecia. This condition is widespread, affecting two of every three men. Androgenic alopecia, which is inherited as an autosomal dominant trait with partial penetrance, is known to be androgen-dependent. Androgens are steroid hormones found circulating at varying levels in both men and women. They are essential in sex differentiation, development, and reproductive function. This is evidenced in the fact tnat castrated males do not develop baldness.
Hair follicles initially appear in utero. No new follicles are created after birth, and it is believed that none are lost in adult life. However, in androgenic alopecia, hair follicles do become progressively smaller (miniaturized). Hair follicles exhibit cyclic activity. Each period of active growth of hair (anagen) alternates with a resting period (telogen), separated by a relatively short transition phase (catagen). Hair growth on the human scalp is a mosaic of follicular activity with each follicle at a stage independent of its neighbors. In the human scalp, anagen may occupy three years or more; however, the percentage of follicles in telogen increases with age, resulting in a gradual thinning. In androgenic alopecia, the ratio of telogen to anagen is increased still further. Also, in androgenic alopecia the hairs in affected areas become steadily shorter and finer, and ultimately may be reduced to the short, fine, unpigmented hair known as vellus hair.
Although the endocrine system does not directly initiate or curtail the activity of the hair follicle, androgens do accelerate or retard the normal cyclic activity of hair growth described above. Testosterone is the major circulating androgen. Because circulating testosterone is largely bound to sex hormone binding globulin (SHBG), the availability of testosterone depends not only on its total concentration, but also on the level of SHBG. Past studies have indicated that plasma testosterone levels in androgenic alopecia appear to be normal, but SHBG levels tend to be low. Also, others have studied the implication of DHT, 5-alpha-dihydrotestosterone, a much more active metabolite formed in the cytoplasm of hair follicle cells after reduction of testosterone by the enzyme 5- alpha-reductase, which is thought to have increased activity in the hair follicles and skin of the frontal scalp in individuals affected by baldness.
Androgens, such as DHT, have been shown to bind specifically to the androgen receptors, which belong to a subfamily of steroid/thyroid hormone/retinoic acid receptors, whose activity is controlled by the tight and specific binding of the cognate ligand. Recent studies have demonstrated that antiandrogens (spironolactone, cyproterone acetate, flutamide and cimetidine) can alleviate androgenic alopecia, by binding to androgen receptors and competing with ligands such as DHT.
Androgenic alopecia, is a strongly heritable condition and is inherited as a complex trait. Familial aggregation, twin studies and consistent ethnic differences in trait frequency have strongly supported the important role of genetic factors in the cause of baldness. Niermann et al., (1964) and Hayakawa et al., (1992) reported the concordance of androgenic alopecia in all the monozygotic pairs and half of the dizygotics pairs they studied, suggesting a very high heritability and an autosomal dominant inheritance pattern. Several baldness-susceptibility loci have been reported on several chromosomes (chromosomes 2, 5 and X), but only recently Hillmer ef al., (2005) confirmed results from a linkage study to markers present on the X chromosome, and found that mutations in the androgen receptor (AR) were a major determinant of early- onset androgenic alopecia. To date, only a few specific candidate genes have been studied in association with androgenic alopecia Genes that are involved in androgen biosynthesis and metabolism were selected as good candidates for involvement in androgenic alopecia However no association between sequence variations in the SRD5A1 and SRD5A2 enzymes (Ellis ef al , 1998 and Ha ef al , 2003), nor 17 alpha-hydroxylase (CYP17) (Carey 1994) could be evidenced to date Only polymorphisms in the androgen receptor were positively associated with Androgenic alopecia in 3 different studies (Ellis ef al , 2001 , Hayes ef al , 2005 and Hillmer ef al , 2005) Insulin was also suggested to be involved in baldness, since it is present in hair follicles and may play a role in the regulation of androgen metabolism and the hair growth cycle A recent study examined the role of this gene in androgenic alopecia but no significant differences between cases and controls in allele, genotype or haplotype frequencies were identified (Ellis ef a/ , 1999) In addition, mutations in the hairless (HR) gene have been reported in families with autosomal recessive alopecia universalis congenitalis and atrichia with papular lesion The main feature of these disorders is persistent and complete absence of hair at or shortly after birth However, the HR gene was not found associated with androgenic alopecia (Hillmer ef a/ , 2001 , and 2002) Also recently, an association has been evidenced between androgenic alopecia and polymorphisms in the ornithine decarboxylase (ODC) gene which has been suggested to play a role in the hair growth cycle (Garton ef al , 2005) More genes and genetic markers associated with baldness need to be identified for a better understanding of the trait
In the past, baldness was treated only with surgical procedures, such as hair transplants and scalp reduction Recently, however, there have been some advances in medical treatment of baldness The most publicized of these is minoxidil (Rogaine) Minoxidil is a potent vasodilator wnich has been used as a treatment for hypertension A noted side effect of this treatment was the growth of hair on parts of the body Research indicates that minoxidil did not help the majority of balding men, although it did help a specific population of minimally balding young men, who had begun to bald within five years The reason for the effectiveness of minoxidil is not known, but is suggested to be due to an increase in blood flow caused by the vasodilating effect of the drug The longterm effects of minoxidil treatment are also not known Another treatment is the topical use of progesterone, a molecule similar to testosterone, which competes with testosterone for binding 5-alpha-reductase, the enzyme that converts testosterone to DHT, and results in lowering the production of DHT from testosterone The side effects of this treatment are not known and again this treatment has not proven to be efficient on a vast majority of balding individuals.
Thus, while there are a number of treatments for baldness currently available, they all are accompanied by various side effects, high costs, and long complicated treatment protocols, which are often not available and effective in a large number of balding individuals. Accordingly, there remains a need in the art for more effective and otherwise improved methods for treating and preventing baldness. Thus, there is a continuing need in the medical arts for genetic markers of baldness and guidance for the use of such markers. The present invention fulfills this need and provides further related advantages.
Genome wide association study to construct a GeneMap for baldness
The present invention is based on the discovery of genes associated with baldness. In the preferred embodiment, disorder-associated loci (candidate regions listed in Table 1 and genes listed in Tables 2-4) are therefore identified by the statistically significant differences in allele frequencies between the cases and the controls. For the purpose of the present invention, 65 candidate regions showing a difference with a -Iog10 P value of 3.0 or higher are identified along with previously known region that has been previously reported to be associated with baldness (example: the androgen receptor on Chromosome X).
The invention provides a method for the discovery of genes associated with baldness and the construction of a GeneMap for baldness in a human population, comprising the following steps (see Example section herein):
Step 1 : Recruit patients (cases) and controls
In the preferred embodiment, 500 patients diagnosed for baldness along with 500 control individuals are recruited from the Quebec Founder Population (QFP).
In another embodiment, less than 500 male patients diagnosed for baldness along with less than 500 male control individuals are recruited from the Quebec Founder Population (QFP).
In another embodiment, more than 500 male patients diagnosed for baldness along with more than 500 male control individuals are recruited from the Quebec Founder Population (QFP). In another embodiment, the present invention is performed as a whole or partially with DNA samples from individuals of another founder population than the Quebec population or from the general population.
Step 2: DNA extraction and dosage
Any sample comprising cells or nucleic acids from patients or controls may be used. Preferred samples are those easily obtained from the patient or control. Such samples include, but are not limited to blood, peripheral lymphocytes, buccal swabs, epithelial cell swabs, nails, hair, bronchoalveolar lavage fluid, sputum, or other body fluid or tissue obtained from an individual.
In the preferred embodiment, DNA is extracted from such samples in the quantity and quality necessary to perform the invention using conventional DNA extraction and dosage techniques. The present invention is not linked to any DNA extraction or dosage platform in particular.
The extracted DNA from case and control samples from recruited individuals is pooled together in various pools. Pools are designed to segregate probands (cases) from controls.
In the preferred embodiment, proband pools, consisting of DNA extracted from recruited individuals, are separated according to level of baldness as determined on the Norwood/Hamilton scale (Norwood 1975), and further separated by age group. Control pools, consisting of DNA extracted from control individuals, but is not limited to pools separated as determined by Norwood/Hamilton scales. The preferred pools distribution is as follows- six proband pools consisted of Norwood/Hamilton type Vl and 2 proband pools consisted of Norwood/Hamilton type VII. The control pools consisted of 5 pools of 57 samples with Norwood/Hamilton type I, and 3 pools of 60, 60, or 65 samples with Norwood/Hamilton type II.
In the preferred embodiment proband samples of 468 male subjects are used to construct the case pools and 470 male controls are used to construct the control pools.
In another embodiment, probant samples of less or more than 500 male subjects are used to construct the case pools and less or more than 500 male controls are used to construct the control pools. Step 3: Genotype the proband and control pools
In the preferred embodiment, assay specific and/or locus-specific and/or allele-specific oligonucleotides for every SNP marker of the present invention (Tables 5, 6.1 and 6.2) are organized onto one or more arrays. The genotype at each SNP locus is revealed by hybridizing short PCR fragments comprising each SNP locus onto these arrays. The arrays permit a high-throughput genome wide association study using DNA samples from individuals of the Quebec founder population. Such assay-specific and/or locus- specific and/or allele-specific oligonucleotides necessary for scoring each SNP of the present invention are preferably organized onto a solid support. Such supports can be arrayed on wafers, glass slides, beads or any other type of solid support.
In another embodiment, the assay-specific and/or locus-specific and/or allele-specific oligonucleotides are not organized onto a solid support but are still used as a whole, in panels or one by one. The present invention is therefore not linked to any genotyping platform in particular.
In another embodiment, one or more portions of the SNPs maps (publicly available maps, proprietary maps from Perlegen Sciences, Inc. (Mountain View, CA, USA), and our own proprietary QLDM map are used to screen the whole genome, a subset of chromosomes, a chromosome, a subset of genomic regions or a single genomic region.
Step 4: Exclude the markers that did not pass the quality control of the assay.
Preferably, the quality controls consist of, but are not limited to, the following criteria: eliminate SNPs that are non-polymorphic in the Quebec founder population or have ≤ 10% minor allele frequency (MAF).
Step 5: Perform the genetic analysis on the results obtained
In the preferred embodiment, genetic analysis is performed on all the genotypes from step 3.
In another embodiment, genetic analysis is performed on a total of 248,535 SNPs.
In one embodiment, the data analysis compares the relative fluorescence intensities of features corresponding to the reference allele of a given SNP with those corresponding to the alternate allele, to calculate a p-hat value. The latter is proportional to the fluorescence signal from perfect match features for the reference allele divided by the sum of fluorescence signals from perfect match features for the reference plus the alternate alleles. P-hat assumes values close to 1 (typically 0.9) for pure reference samples and close to 0 (typically 0.1 ) for pure alternate samples, and can be used as a measured estimate of the reference allele frequency of a SNP in a DNA pool. The difference between case and control pools, delta p-hat, is calculated using the weighted average of case and control p-hats. Delta p-hat is a reliable estimate of the allele frequency difference between the cases and controls.
In yet another embodiment, the data is analyzed according to the p-hat value obtained from the previous embodiment on each pool. Single marker P values are calculated for all markers within the genome wide scan map as described in Example 3 herein, using the p-hat value.
In another embodiment, the combined P values across multi-marker sliding windows are calculated after the method of Fisher (described in Example 3 herein).
Step 6: Fine Mapping
In this step, the candidate regions that were identified by step 5 are further mapped and confirmed for the purpose of refinement and validation.
In the preferred embodiment, the cases and controls are individually genotyped to confirm the candidate regions. The confirmed candidate regions are processed by fine mapping to refine the candidate regions.
In the preferred embodiment, this fine mapping is performed with a density of genetic markers higher than in the genome wide scan (step 3) using any genotyping platform available in the art. Such fine mapping can be, but is not limited to, typing the allele via an allele-specific elongation assay that is then ligated to a locus-specific oligonucleotide. Such assays can be performed directly on the genomic DNA at a highly multiplex level and the products can be amplified using universal oligonucleotides. For each candidate region, the density of genetic markers can be, but is not limited to, a set of SNP markers with an average inter-marker distance of 1 -4 Kb distributed over about 400 Kb to 1 Mb, roughly centered at the highest point of the GWS association. The preferred samples are those obtained from baldness samples including the ones used for the GWS.
In another embodiment, samples used for the GWS can be supplemented with other samples to increase total cases and controls. In another embodiment, 650 cases and 650 controls samples can be used for the fine mapping genotyping analysis.
In the preferred embodiment, the genetic analysis of the results obtained using haplotype information (available after confirmatory mapping individual samples, see Example section herein) as well as single-marker association (as performed as in step 5, described herein) are performed as described herein (step 5 and Example section). The candidate regions that are validated and confirmed after this analysis proceed to a gene mining step described in Example 5, herein, to characterize their marker and genetic content.
Step 7: SNP and DNA polymorphism discovery
In the preferred embodiment, all the candidate genes and regions identified in step 6 are sequenced for polymorphism identification.
In another embodiment, the entire region, including all introns, is sequenced to identify all polymorphisms.
In yet another embodiment, the candidate genes are prioritized for sequencing, and only functional gene elements (promoters, exons and splice sites) are sequenced.
In yet another embodiment, previously identified polymorphisms in the candidate regions can also be used. For example, SNPs from dbSNP, Perlegen Sciences, Inc., or others can also be used rather than resequencing the candidate regions to identify polymorphisms.
The discovery of SNPs and DNA polymorphisms generally compi ises a sιeρ consisting of determining the major haplotypes in the region to be sequenced. The preferred samples are selected according to which haplotypes contribute to the association signal observed in the region to be sequenced. The purpose is to select a set of samples that cover all the major haplotypes in the given region. Each major haplotype is preferably analyzed in at least a few individuals.
Any analytical procedure may be used to detect the presence or absence of variant nucleotides at one or more polymorphic positions of the invention. In general, the detection of allelic variation requires a mutation discrimination technique, optionally an amplification reaction and optionally a signal generation system. Any means of mutation detection or discrimination may be used. For instance, DNA sequencing, scanning methods, hybridization, extension based methods, incorporation based methods, restriction enzyme-based methods and ligation-based methods may be used in the methods of the invention.
Sequencing methods include, but are not limited to, direct sequencing, and sequencing by hybridization. Scanning methods include, but are not limited to, protein truncation test (PTT), single-strand conformation polymorphism analysis (SSCP), denaturing gradient gel electrophoresis (DGGE), temperature gradient gel electrophoresis (TGGE), cleavage, heteroduplex analysis, chemical mismatch cleavage (CMC), and enzymatic mismatch cleavage. Hybridization-based methods of detection include, but are not limited to, solid phase hybridization such as dot blots, multiple allele specific diagnostic assay (MASDA), reverse dot blots, and oligonucleotide arrays (DNA Chips). Solution phase hybridization amplification methods may also be used, such as Taqman. Extension based methods include, but are not limited to, amplification refraction mutation systems (ARMS), amplification refractory mutation systems (ALEX), and competitive oligonucleotide priming systems (COPS). Incorporation based methods include, but are not limited to, mini-sequencing and arrayed primer extension (APEX). Restriction enzyme-based detection systems include, but are not limited to, restriction site generating PCR. Lastly, ligation based detection methods include, but are not limited to, oligonucleotide ligation assays (OLA). Signal generation or detection systems that may be used in the methods of the invention include, but are not limited to, fluorescence methods such as fluorescence resonance energy transfer (FRET), fluorescence quenching, fluorescence polarization as well as other chemiluminescence, electrochemiluminescence, Raman, radioactivity, colometric methods, hybridization protection assays and mass spectrometry methods. Further amplification methods include, but are not limited to self sustained replication (SSR), nucleic acid sequence based amplification (NASBA), ligase chain reaction (LCR), strand displacement amplification (SDA) and branched DNA (B-DNA).
Step 8: Ultrafine Mapping
This step further maps the candidate regions and genes confirmed in the previous step to identify and validate the responsible polymorphisms associated with baldness in the human population. In a preferred embodiment, the discovered SNPs and polymorphisms of step 7 are ultrafine mapped at a higher density of markers than the fine mapping described herein using the same technology described in step 6
Step 9 GeneMap construction
The confirmed variations in DNA (including both genie and non-genic regions) are used to build a GeneMap for baldness The gene content of this GeneMap is described in more detail below Such GeneMap can be used for other methods of the invention comprising the diagnostic methods described herein, the susceptibility to baldness, the response to a particular drug, the efficacy of a particular drug, the screening methods described herein and the treatment methods described herein
As is evident to one of ordinary skill in the art, all of the above steps do not need to be performed, or performed in a given order to practice or use the SNPs, genomic regions, genes, proteins, etc in the methods of the invention
Genes from the GeneMap
In the preferred embodiment the GeneMap consists of genes and targets, in a variety of combinations, identified from the candidate regions listed in Table 1 In the preferred embodiment, all genes from Tables 2-4 are present in the GeneMap
For additional clarity, some known functions and characteristics of the genes identified in the WGAS on pooled samples and confirmed in a fine-mapping study for male pattern baldness (MPB) are evaluated using the Ingenuity Pathway Analysis application (IPA, Ingenuity systems) in order to identify direct biological interactions between these genes, and also to identify molecular regulators acting on those genes (indirect interactions) that can be involved in MPB The purpose of this effort is to decipher the molecules involved in contributing to MPB susceptibility These gene interaction networks are very valuable tools in the sense that they facilitate extension of the map of gene products that could represent potential drug targets for MPB
Based on the results of the genetic analyses described herein, a selection of 4 genes is used for the development of potential protein interaction networks involved in MPB These genes are imported in the IPA software and are presented in Table A, below Table A. Input list of genes
Region ID Entrez Gene ID Gene Symbol
63 60401 EDA2R
63 367 AR
20 1879 EBF1
65 1896 EDA
In a first step, the analysis is performed by looking for direct interactions only. From this analysis 44 genes are mapped to the Ingenuity database and assigned to 1 network as defined by IPA (Figure 1 below). These networks are based on functional relationships between gene products using known interactions in the literature. For each individual network, some nodes are manually extended to include good candidate genes that could play a role in the biochemical pathways of MPB. Table B contains information about the gene content of the network, as well as the top functions assigned to those biochemical pathways.
Table B. Genetic networks associated with genes confirmed by the fine-mapping on MPB. Direct interactions only. Bold genes are those identified by the fine-mapping
Genes manually
Network Genes in Ingenuity network Top functions added to the networks
ACTA1. AKR1 B7, AR1 CCNE1 , CD79B, CDC25B, CITED2, CYP17A1. DHFR, EBF1. EDA, EDA2R, ELF3,
Cell death, Cancer, CD19, CD79A, EDAR,
EP300, GATA5, GTF2F1 ,
Cellular growth and PAX5, PPARA,
HSPA1 B, KLKBI 1 LHB, proliferation, Tissue SLC2A4, TCF3,
MEF2D, MYH11. NCOA6, morphology TRAF3, TRAF6
PARP1 , PEM, PIAS3,
PLAGL1 , PTGS2, RNF4,
SMARCC1. SOX9, SP1 ,
TAL1. TFF1 , UXT, ZNF148
Network 1 contains 44 nodes (35 original and 9 manual additions) and includes the 4 input genes (Figure 1 ). A short description of these 4 genes follows. By virtue of their role in hair follicle morphogenesis and/or androgen signaling, several genes from this network are very good candidates for involvement in the pathophysiology of MPB.
EDA2R: EDA2R gene is also known as XEDAR, EDAA2R, EDA-A2R and TNFRSF27. This protein is a type III transmembrane protein of the TNFR (tumor necrosis factor receptor) superfamily, and contains 3 cysteine-rich repeats and a single transmembrane domain but lacks an N-terminal signal peptide (Yan et al 2000). Multiple alternatively spliced transcript variants have been found for this gene, but some variants lack sufficient support. This receptor specifically binds EDA-A2, an isoform of the anhidrotic ectodermal dysplasia (EDA) gene (see below). EDA-A2 binding to EDA2R leads to interaction of its intracellular domain with TRAF3 and TRAF6 and activation of both NF- KB and JNK pathways (Botchkarev and Fessing 2005). In developing mouse skin, by E16 and E17 XEDAR expression was expressed in large amounts in maturing follicles. By postnatal day 1 , the pattern of expression was confined to hair follicles (Yan et al 2000).
AR: The androgen receptor gene (AR) is more than 90 kb long and codes for a protein that has 3 major functional domains: the N-terminal domain, DNA-binding domain, and androgen-binding domain. The protein functions as a steroid-hormone activated transcription factor. Upon binding the hormone ligand, the receptor dissociates from accessory proteins, translocates into the nucleus, dimerizes, and then stimulates transcription of androgen responsive genes. This gene contains 2 polymorphic trinucleotide repeat segments that encode polyglutamine and polyglycine tracts in the N- terminal transactivation domain of its protein. The involvement of androgen signaling in the development of MPB is well known. To date, the androgen receptor is the only gene reported to be positively associated with MPB by several independent studies (Ellis et al 2001 ; Hayes et al 2005; Hillmer et al 2005; Levy-Nissenbaum et al 2005).
EBF1 : the gene early B cell factor 1 encodes a transcription which plays an essential role in B-cell develoment. A role of the immune system on hair follicle cycling has been hypothesized (Stenn and Paus 2001 ). Interestingly, TCF family members LEF1 and TCF1 were first characterized by their role in T- and B-cell development (Staal and Clevers 2000), and were shown subsequently to be involved in hair morphogenesis (Fuchs et al 2001 , Schmidt-Ullrich and Paus 2005) In add:tion, expression of LEF1 is decreased in Ebf+/- E2a+/- mice pro-B cells (O'Riordan and Grosschedl 1999). Finally, EBF1 interacts with the transcription factor RUNX1 (Maier et al 2004) which has a role in hair morphogenesis (Raveh et al 2006). Those evidence make the EBF1 gene a very relevant candidate to play a role in hair follicle morphogenesis, and hence MPB.
EDA: The anhidrotic ectodermal dysplasia (EDA) gene encodes various isoforms of ectodysplasin. EDA-A1 and EDA-A2 are two of the 8 isoforms of EDA. EDA-A1 is the longest form and encodes a 391 -residue protein with a domain similar to TNF at the COOH terminus. EDA-A1 binds specifically EDA1 R receptor. Except for a two-residue deletion (Glu308 and Val309 in this domain), EDA-A2 is identical to EDA-A1. EDA-A2 is generated through the use of an alternative internal splice donor site. EDA-A1 and EDA-A2 are 2 trimeric type Il membrane proteins both containing a short intracellular domain, transmembrane region, and extracellular portion with collagenous domain and a TNF ligand motif in the C terminal region. Cleavage of EDA-A1/2 by the furin-like enzyme leads to formation of soluble extracellular molecule which is able to interact with corresponding receptors and mediate the signal. Mutations in EDA give rise to a clinical syndrome "hypohydrotic ectodermal dysplasia" characterized by loss of hair, sweat glands, and teeth (OMIM 300451 ). Mutations in EDA are responsible for most of the clinical cases studied to date. In a minority of patients, this phenotype is due to mutations in a distinct gene termed downless in mice and EDAR in humans (or EDA1 R) (OMIM 604095).
In a second step, the analysis is performed by looking for direct and indirect interactions. From this analysis 35 genes are mapped to the Ingenuity database and assigned to 1 genetic network as defined by IPA (Figure 2). Table C contains information about the gene content of the network, as well as the top functions assigned to those biochemical pathways.
Network 1 b contains 35 original nodes and includes the 4 input genes (Figure 2). For descriptions of the EDA2R, AR, EBF1 and EDA gene, please refer to text above about networks from direct analysis only. Figure 3 below lists the symbols used in both figures 1 and 2.
Figure imgf000031_0001
Nucleic acid sequences The nucleic acid sequences of the present invention may be derived from a variety of sources including DNA, miRNA, cDNA, synthetic DNA, synthetic RNA, derivatives, mimetics or combinations thereof. Such sequences may comprise genomic DNA, which may or may not include naturally occurring introns, genie regions, nongenic regions, and regulatory regions. Moreover, such genomic DNA may be obtained in association with promoter regions or poly (A) sequences. The sequences, genomic DNA, or cDNA may be obtained in any of several ways. Genomic DNA can be extracted and purified from suitable cells by means well known in the art. Alternatively, mRNA can be isolated from a cell and used to produce cDNA by reverse transcription or other means. The nucleic acids described herein are used in certain embodiments of the methods of the present invention for production of RNA, proteins or polypeptides, through incorporation into cells, tissues, or organisms. In one embodiment, DNA containing all or part of the coding sequence for the genes described in Tables 2-4, or the SNP markers described in Tables 5, 6.1 and 6.2, is incorporated into a vector for expression of the encoded polypeptide in suitable host cells. The invention also comprises the use of the nucleotide
1 sequence of the nucleic acids of this invention to identify DNA probes for the genes described in Tables 2-4 or the SNP markers described in Tables 5, 6.1 and 6.2, PCR primers to amplify the genes described in Tables 2-4 or the SNP markers described in Tables 5, 6.1 and 6.2, nucleotide polymorphisms in the genes described in Tables 2-4, and regulatory elements of the genes described in Tables 2-4. The nucleic acids of the present invention find use as primers and templates for the recombinant production of baldness-associated peptides or polypeptides, for chromosome and gene mapping, to provide antisense sequences, for tissue distribution studies, to locate and obtain full length genes, to identify and obtain homologous sequences (wild-type and mutants), and in diagnostic applications.
Antisense oligonucleotides
In a particular embodiment of the invention, an antisense nucleic acid or oligonucleotide is wholly or partially complementary to, and can hybridize with, a target nucleic acid (either DNA or RNA) having the sequence of SEQ ID NO:1 , NO:3 or any SEQ ID from Tables 2-5, 6.1 and 6.2. For example, an antisense nucleic acid or oligonucleotide comprising 16 nucleotides can be sufficient to inhibit expression of at least one gene from Tables 2-4. Alternatively, an antisense nucleic acid or oligonucleotide can be complementary to 51 or 3' untranslated regions, or can overlap the translation initiation codon (51 untranslated and translated regions) of at least one gene from Tables 2-4, or its functional equivalent. In another embodiment, the antisense nucleic acid is wholly or partially complementary to, and can hybridize with, a target nucleic acid that encodes a polypeptide from a gene described in Tables 2-4.
In addition, oligonucleotides can be constructed which will bind to duplex nucleic acid (i.e., DNA:DNA or DNA:RNA), to form a stable triple helix containing or triplex nucleic acid. Such triplex oligonucleotides can inhibit transcription and/or expression of a gene from Tables 2-4, or its functional equivalent (M. D. Frank-Kamenetskii et a/., 1995). Triplex oligonucleotides are constructed using the basepairing rules of triple helix formation and the nucleotide sequence of the genes described in Tables 2-4.
The present invention encompasses methods of using oligonucleotides in antisense inhibition of the function of the genes from Tables 2-4. In the context of this invention, the term "oligonucleotide" refers to naturally-occurring species or synthetic species formed from naturally-occurring subunits or their close homologs. The term may also refer to moieties that function similarly to oligonucleotides, but have non-naturally-occurring portions. Thus, oligonucleotides may have altered sugar moieties or inter-sugar linkages. Exemplary among these are phosphorothioate and other sulfur containing species which are known in the art. In preferred embodiments, at least one of the phosphodiester bonds of the oligonucleotide has been substituted with a structure that functions to enhance the ability of the compositions to penetrate into the region of cells where the RNA whose activity is to be modulated is located. It is preferred that such substitutions comprise phosphorothioate bonds, methyl phosphonate bonds, or short chain alkyl or cycloalkyl structures. In accordance with other preferred embodiments, the phosphodiester bonds are substituted with structures which are, at once, substantially non-ionic and πon chira!, or with structures which are chira' and enantiomerically specific. Persons of ordinary skill in the art will be able to select other linkages for use in the practice of the invention. Oligonucleotides may also include species that include at least some modified base forms. Thus, purines and pyrimidines other than those normally found in nature may be so employed. Similarly, modifications on the furanosyl portions of the nucleotide subunits may also be effected, as long as the essential tenets of this invention are adhered to. Examples of such modifications are 2'-O-alkyl- and T- halogen-substituted nucleotides. Some non-limiting examples of modifications at the 2' position of sugar moieties which are useful in the present invention include OH, SH, SCH3, F, OCH3, OCN, O(CH2), NH2 and O(CH2)n CH3, where n is from 1 to about 10. Such oligonucleotides are functionally interchangeable with natural oligonucleotides or synthesized oligonucleotides, which have one or more differences from the natural structure All such analogs are comprehended by this invention so long as they function effectively to hybridize with at least one gene from Tables 2-4 DNA or RNA to inhibit the function thereof
The oligonucleotides in accordance with this invention preferably comprise from about 3 to about 50 subunits It is more preferred that such oligonucleotides and analogs comprise from about 8 to about 25 subunits and still more preferred to have from about 12 to about 20 subunits As defined herein, a "subunit" is a base and sugar combination suitably bound to adjacent subunits through phosphodiester or other bonds Antisense nucleic acids or oligonucleotides can be produced by standard techniques (see, e g , Shewmaker et al , U S Patent No 6,107,065) The oligonucleotides used in accordance with this invention may be conveniently and routinely made through the well-known technique of solid phase synthesis Any other means for such synthesis may also be employed, however, the actual synthesis of the oligonucleotides is well within the abilities of the practitioner It is also well known to prepare other oligonucleotide such as phosphorothioates and alkylated derivatives
The oligonucleotides of this invention are designed to be hybπdizable with RNA (e g , mRNA) or DNA from genes described in Tables 2-4 For example, an oligonucleotide (e g , DNA oligonucleotide) that hybridizes to mRNA from a gene described in Tables 2-4 can be used to target the mRNA for RnaseH digestion Alternatively, an oligonucleotide that can hybridize to the translation initiation site of the mRNA of a gene described in Tables 2-4 can be used to prevent translation of the mRNA In another approach, oligonucleotides that bind to the double-stranded DNA of a gene from Tables 2-4 can be admιnιsfered Such oligonucleotides can form a tπo'ev COiSt-Uc+ and nh'bit the transcription of the DNA encoding polypeptides of the genes described in Tables 2-4 Triple helix pairing prevents the double helix from opening sufficiently to allow the binding of polymerases, transcription factors, or regulatory molecules Recent therapeutic advances using triplex DNA have been described (see, e g , J E Gee ef al , 1994, Molecular and Immunologic Approaches, Futura Publishing Co , Mt Kisco, NY)
As non-limiting examples, antisense oligonucleotides may be targeted to hybridize to the following regions mRNA cap region, translation initiation site, translational termination site, transcription initiation site, transcription termination site, polyadenylation signal, 31 untranslated region, 5' untranslated region, 5'codιng region, mid coding region, and 3' coding region Preferably, the complementary oligonucleotide is designed to hybridize to the most unique 5' sequence of a gene described in Tables 2-4, including any of about 15-35 nucleotides spanning the 5' coding sequence. In accordance with the present invention, the antisense oligonucleotide can be synthesized, formulated as a pharmaceutical composition, and administered to a subject. The synthesis and utilization of antisense and triplex oligonucleotides have been previously described (e.g., Simon et al., 1999; Barre et al., 2000; Elez et al., 2000; Sauter et a/., 2000).
Alternatively, expression vectors derived from retroviruses, adenovirus, herpes or vaccinia viruses, or from various bacterial plasmids may be used for delivery of nucleotide sequences to the targeted organ, tissue or cell population. Methods which are well known to those skilled in the art can be used to construct recombinant vectors which will express nucleic acid sequence that is complementary to the nucleic acid sequence encoding a polypeptide from the genes described in Tables 2-4. These techniques are described both in Sambrook et al., 1989 and in Ausubel et al., 1992. For example, expression of at least one gene from Tables 2-4 can be inhibited by transforming a cell or tissue with an expression vector that expresses high levels of untranslatable sense or antisense sequences. Even in the absence of integration into the DNA, such vectors may continue to transcribe RNA molecules until they are disabled by endogenous nucleases. Transient expression may last for a month or more with a nonreplicating vector, and even longer if appropriate replication elements are included in the vector system. Various assays may be used to test the ability of gene-specific antisense oligonucleotides to inhibit the expression of at least one gene from Tables 2-4. For example, mRNA levels of the genes described in Tables 2-4 can be assessed by Northern blot analysis (Sambrook et al., 1989; Ausubel et al., 1992; J. C. Alwine et al. 1977; I. M. Bird, 1998), quantitative or semi-quantitative RT-PCR analysis (see, e.g., W.M. Freeman et al., 1999; Ren et al., 1998; J. M. CaIe er al., 1998), or in situ hybridization (reviewed by A.K. Raap, 1998). Alternatively, antisense oligonucleotides may be assessed by measuring levels of the polypeptide from the genes described in Tables 2-4, e.g., by western blot analysis, indirect immunofluorescence and immunoprecipitation techniques (see, e.g., J. M. Walker, 1998, Protein Protocols on CD-ROM, Humana Press, Totowa, NJ). Any other means for such detection may also be employed, and is well within the abilities of the practitioner. Mapping Technologies
The present invention includes various methods which employ mapping technologies to map SNPs and polymorphisms. For purpose of clarity, this section comprises, but is not limited to, the description of mapping technologies that can be utilized to achieve the embodiments described herein. Mapping technologies may be based on amplification methods, restriction enzyme cleavage methods, hybridization methods, sequencing methods, and cleavage methods using agents.
Amplification methods include: self sustained sequence replication (Guatelli et al., 1990), transcriptional amplification system (Kwoh et al., 1989), Q-Beta Replicase (Lizardi et al., 1988), isothermal amplification (e.g. Dean et al., 2002; and Hafner et al., 2001 ), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of ordinary skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low number.
Restriction enzyme cleavage methods include: isolating sample and control DNA, amplification (optional), digestion with one or more restriction endonucleases, determination of fragment length sizes by gel electrophoresis and comparing samples and controls. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA. Moreover, sequence specific ribozymes (see, e.g., U.S. Pat. No. 5,498,531 ) or DNAzyme (e.g. U.S. Pat. No. 5,807,718) can be used to score for the presence of specific mutations by development or loss of a ribozyme or DNAzyme cleavage site.
SNPs and SNP maps of the invention can be identified or generateα Dy hybridizing sample nucleic acids, e.g., DNA or RNA, to high density arrays or bead arrays containing oligonucleotide probes corresponding to the SNPS of Tables 5, 6.1 and 6.2 (see the Affymetrix arrays and lllumina bead sets at www.affymetrix.com and www.illumina.com and see Cronin et al., 1996; or Kozal et al., 1996).
A variety of sequencing reactions known in the art can be used to directly sequence nucleic acids for the presence or the absence of one or more SNPs of Tables 5, 6.1 and 6.2. Examples of sequencing reactions include those based on techniques developed by Maxam and Gilbert (1977) or Sanger (1977). It is also contemplated that any of a variety of automated sequencing procedures can be utilized, including sequencing by mass spectrometry (see, e.g. PCT International Publication No. WO 94/16101 ; Cohen et al., 1996; and Griffin et a/., 1993), real-time pyrophosphate sequencing method (Ronaghi et a/., 1998; and Permutt et al., 2001 ) and sequencing by hybridization (see e.g. Drmanac et al., 2002).
Other methods of detecting SNPs include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA, DNA/DNA or RNA/DNA heteroduplexes (Myers et al., 1985). In general, the technique of "mismatch cleavage" starts by providing heteroduplexes formed by hybridizing (labeled) RNA or DNA containing a wild-type sequence with potentially mutant RNA or DNA obtained from a sample. The double-stranded duplexes are treated with an agent who cleaves single- stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with S1 nuclease to enzymatically digest the mismatched regions. In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of a mutation or SNP (see, for example, Cotton et al., 1988; and Saleeba et al., 1992). In a preferred embodiment, the control DNA or RNA can be labeled for detection.
In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping SNPs. For example, the mutY enzyme of E. coli c'eaves A at G/A mismatches (Hsu et a/ , 1994) Other examples include, but are not limited to, the MutHLS enzyme complex of E. coli (Smith and Modrich Proc. 1996) and CeI 1 from the celery (Kulinski et al., 2000) both cleave the DNA at various mismatches. According to an exemplary embodiment, a probe based on a polymorphic site corresponding to a SNP of Tables 5, 6.1 and 6.2 is hybridized to a cDNA or other DNA product from a test cell or cells. The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like (see, for example, U.S. Pat. No. 5,459,039). Alternatively, the screen can be performed in vivo following the insertion of the heteroduplexes in an appropriate vector. The whole procedure is known to those ordinary skilled in the art and is referred to as mismatch repair detection (see e.g. Fakhrai-Rad et al., 2004).
In other embodiments, alterations in electrophoretic mobility can be used to identify SNPs in a sample. For example, single strand conformation polymorphism (SSCP) can be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids (Orita et al., 1989; Cotton et al., 1993; and Hayashi 1992). Single-stranded DNA fragments of case and control nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence. The resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In a preferred embodiment, the method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Kee et al., 1991 ).
In yet another embodiment, the movement of mutant or wild-type fragments in a polyacrylamide gel containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al., 1985). When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum et al., 1987). In another embodiment, the mutant fragment is detected using denaturing HPLC (see e.g. Hoogendoorn et a/ . 2000)
Examples of other techniques for detecting SNPs include, but are not limited to, selective oligonucleotide hybridization, selective amplification, selective primer extension, selective ligation, single-base extension, selective termination of extension or invasive cleavage assay. For example, oligonucleotide primers may be prepared in which the SNP is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al., 1986; Saiki et al., 1989). Such oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA. Alternatively, the amplification, the allele-specific hybridization and the detection can be done in a single assay following the principle of the 5' nuclease assay (e.g. see Livak et al., 1995). For example, the associated allele, a particular allele of a polymorphic locus, or the like is amplified by PCR in the presence of both allele-specific oligonucleotides, each specific for one or the other allele. Each probe has a different fluorescent dye at the 5' end and a quencher at the 3' end. During PCR, if one or the other or both allele-specific oligonucleotides are hybridized to the template, the Taq polymerase via its 5' exonuclease activity will release the corresponding dyes. The latter will thus reveal the genotype of the amplified product.
Hybridization assays may also be carried out with a temperature gradient following the principle of dynamic allele-specific hybridization or like e.g. Jobs et al., (2003); and Bourgeois and Labuda, (2004). For example, the hybridization is done using one of the two allele-specific oligonucleotides labeled with a fluorescent dye, an intercalating quencher under a gradually increasing temperature. At low temperature, the probe is hybridized to both the mismatched and full-matched template. The probe melts at a lower temperature when hybridized to the template with a mismatch. The release of the probe is captured by an emission of the fluorescent dye, away from the quencher. The probe melts at a higher temperature when hybridized to the template with no mismatch. The temperature-dependent fluorescence signals therefore indicate the absence or presence of an associated allele, a particular allele of a polymorphic locus, or the like (e.g. Jobs et al., 2003). Alternatively, the hybridization is done under a gradually decreasing temperature. In this case, both allele-specific oligonucleotides are hybridized to the template competitively. At high temperature none of the two probes are hybridized. Once the optimal temperature of the full-matched probe is reached, it hybridizes and leaves no target for the mismatched probe (e.g. Bourgeois and Labuda, 2004). In the latter case, if the allele-specific probes are differently labeled, then they are hybridized to a single PCR-amphfied target. If the probes are labeled with the same dye, then the probe cocktail is hybridized to twice to identical templates with only one labeled probes, different in the two cocktails, in the presence of the unlabeled competitive probe.
Alternatively, allele specific amplification technology that depends on selective PCR amplification may be used in conjunction with the present invention. Oligonucleotides used as primers for specific amplification may carry the associated allele, a particular allele of a polymorphic locus, or the like, also referred to as "mutation" of interest in the center of the molecule, so that amplification depends on differential hybridization (Gibbs et al., 1989) or at the extreme 3' end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner, 1993). In addition it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection (Gasparini et al., 1992). It is anticipated that in certain embodiments, amplification may also be performed using Taq ligase for amplification (Barany, 1991 ). In such cases, ligation will occur only if there is a perfect match at the 3' end of the 5' sequence making it possible to detect the presence of a known associated allele, a particular allele of a polymorphic locus, or the like at a specific site by looking for the presence or absence of amplification. The products of such an oligonucleotide ligation assay can also be detected by means of gel electrophoresis. Furthermore, the oligonucleotides may contain universal tags used in PCR amplification and zip code tags that are different for each allele. The zip code tags are used to isolate a specific, labeled oligonucleotide that may contain a mobility modifier (e.g. Grossman et al., 1994).
In yet another alternative, allele-specific elongation followed by ligation will form a template for PCR amplification. In such cases, elongation will occur only if there is a perfect match at the 3' end of the allele-specific oligonucleotide using a DNA polymerase. This reaction is performed directly on the genomic DNA and the extension/ligation products are amplified by PCR. To this end, the oligonucleotides contain universal tags allowing amplification at a high multiplex level and a zip code for SNP identification. The PCR tags are designed in such a way that the two alleles of a SNP are amplified by different forward primers, each having a different dye. The zip code tags are the same for both alleles of a given SNPs and they are used for hybridization of the PCR-amplified products to oligonucleotides bound to a solid support, chip, bead array or like. For an example of the procedure, see Fan et al. (Cold Spring Harbor Symposia on Quantitative Biology, Vol. LXVIII, pp. 69-78 2003).
Another alternative includes the single-bass extension'ligat;oi assay ι,sirg s molecular inversion probe, consisting of a single, long oligonucleotide (see e.g. Hardenbol et al., 2003). In such an embodiment, the oligonucleotide hybridizes on both side of the SNP locus directly on the genomic DNA, leaving a one-base gap at the SNP locus. The gap- filling, one-base extension/ligation is performed in four tubes, each having a different dNTP. Following this reaction, the oligonucleotide is circularized whereas unreactive, linear oligonucleotides are degraded using an exonuclease such as exonuclease I of E. coli. The circular oligonucleotides are then linearized and the products are amplified and labeled using universal tags on the oligonucleotides. The original oligonucleotide also contains a SNP-specific zip code allowing hybridization to oligonucleotides bound to a solid support, chip, and bead array or like. This reaction can be performed at a high multiplexed level.
In another alternative, the associated allele, a particular allele of a polymorphic locus, or the like is scored by single-base extension (see e.g. U.S. Pat. No. 5,888,819). The template is first amplified by PCR. The extension oligonucleotide is then hybridized next to the SNP locus and the extension reaction is performed using a thermostable polymerase such as ThermoSequenase (GE Healthcare) in the presence of labeled ddNTPs. This reaction can therefore be cycled several times. The identity of the labeled ddNTP incorporated will reveal the genotype at the SNP locus. The labeled products can be detected by means of gel electrophoresis, fluorescence polarization (e.g. Chen et al., 1999) or by hybridization to oligonucleotides bound to a solid support, chip, and bead array or like. In the latter case, the extension oligonucleotide will contain a SNP-specific zip code tag.
In yet another alternative, a SNP is scored by selective termination of extension. The template is first amplified by PCR and the extension oligonucleotide hybridizes in vicinity to the SNP locus, close to but not necessarily adjacent to it. The extension reaction is carried out using a thermostable polymerase such as Thermo Sequenase (GE Healthcare) in the presence of a mix of dNTPs and at least one ddNTP. The latter has to terminate the extension at one of the allele of the interrogated SNP, but not both such that the two alleles will generate extension products of different sizes. The extension product can then be detected by means of gel electrophoresis, in which case the extension products need to be labeled, or by mass spectrometry (see e.g. Storm ef al., 2003).
In another alternative, SNPs are detected using an invasive cleavage assay (see U.S. Pat. No. 6,090,543). There are five oligonucleotides per SNP to interrogate but these are used in a two step-reaction. During the primary reaction, three of the designed oligonucleotides are first hybridized directly to the genomic DNA. One of them is locus- specific and hybridizes up to the SNP locus (the pairing of the 3' base at the SNP locus is not necessary). There are two allele-specific oligonucleotides that hybridize in tandem to the locus-specific probe but also contain a 5' flap that is specific for each allele of the SNP. Depending upon hybridization of the allele-specific oligonucleotides at the base of the SNP locus, this creates a structure that is recognized by a cleavase enzyme (U.S. Pat. No. 6,090,606) and the allele-specific flap is released. During the secondary reaction, the flap fragments hybridize to a specific cassette to recreate the same structure as above except that the cleavage will release a small DNA fragment labeled with a fluorescent dye that can be detected using regular fluorescence detector. In the cassette, the emission of the dye is inhibited by a quencher.
Methods to identify agents that modulate the expression of a nucleic acid encoding a gene involved in baldness.
The present invention provides methods for identifying agents that modulate the expression of a nucleic acid encoding a gene from Tables 2-4. Such methods may utilize any available means of monitoring for changes in the expression level of the nucleic acids of the invention. As used herein, an agent is said to modulate the expression of a nucleic acid of the invention if it is capable of up- or down- regulating expression of the nucleic acid in a cell. Such cells can be obtained from any parts of the body such as the scalp, blood, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, genitalia, vessels and endothelium. Some non-limiting examples of cells that can be used are: hair (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
In one assay format, the expression of a nucleic acid encoding a gene of the invention (see Tables 2-4) in a cell or tissue sample is monitored directly by hybridization to the nucleic acids of the invention. Cell lines or tissues are exposed to the agent to be tested under appropriate conditions and time and total RNA or mRNA is isolated by standart procedures such as those disclosed in Sambrook et al., (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press).
Probes to detect differences in RNA expression levels between cells exposed to the agent and control cells may be prepared as described above. Hybridization conditions are modified using known methods, such as those described by Sambrook et al., and Ausubel ef al., as required for each probe. Hybridization of total cellular RNA or RNA enriched for polyA RNA can be accomplished in any available format. For instance, total cellular RNA or RNA enriched for polyA RNA can be affixed to a solid support and the solid support exposed to at least one probe comprising at least one, or part of one of the sequences of the invention under conditions in which the probe will specifically hybridize. Alternatively, nucleic acid fragments comprising at least one, or part of one of the sequences of the invention can be affixed to a solid support, such as a silicon chip or a porous glass wafer. The chip or wafer can then be exposed to total cellular RNA or polyA RNA from a sample under conditions in which the affixed sequences will specifically hybridize to the RNA. By examining for the ability of a given probe to specifically hybridize to an RNA sample from an untreated cell population and from a cell population exposed to the agent, agents which up or down regulate expression are identified.
Methods to identify agents that modulate the activity of a protein encoded by a gene involved in baldness.
The present invention provides methods for identifying agents that modulate at least one activity of the proteins described in Tables 2-4. Such methods may utilize any means of monitoring or detecting the desired activity. As used herein, an agent is said to modulate the expression of a protein of the invention if it is capable of up- or down- regulating expression of the protein in a cell. Such cells can be obtained from any parts of the body such as the scalp, blood, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, genitalia, vessels and endothelium. Some non-limiting examples of cells that can be used are: hair (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
In one format, the specific activity of a protein of the invention, normalized to a standard unit, may be assayed in a cell population that has been exposed to the agent to be tested and compared to an unexposed control cell population may be assayed. Cell lines or populations are exposed to the agent to be tested under appropriate conditions and time. Cellular lysates may be prepared from the exposed cell line or population and a control, unexposed cell line or population. The cellular lysates are then analyzed with the probe.
Antibody probes can be prepared by immunizing suitable mammalian hosts utilizing appropriate immunization protocols using the proteins of the invention or antigen- containing fragments thereof. To enhance immunogenicity, these proteins or fragments can be conjugated to suitable carriers. Methods for preparing immunogenic conjugates with carriers such as BSA, KLH or other carrier proteins are well known in the art. In some circumstances, direct conjugation using, for example, carbodiimide reagents may be effective; in other instances linking reagents such as those supplied by Pierce Chemical Co. (Rockford, IL) may be desirable to provide accessibility to the hapten. The hapten peptides can be extended at either the amino or carboxy terminus with a cysteine residue or interspersed with cysteine residues, for example, to facilitate linking to a carrier. Administration of the immunogens is conducted generally by injection over a suitable time period and with use of suitable adjuvants, as is generally understood in the art. During the immunization schedule, titers of antibodies are taken to determine adequacy of antibody formation. While the polyclonal antisera produced in this way may be satisfactory for some applications, for pharmaceutical compositions, use of monoclonal preparations is preferred. Immortalized cell lines which secrete the desired monoclonal antibodies may be prepared using standard methods, see e.g., Kohler & Milstein (1992) or modifications which affect immortalization of lymphocytes or spleen cells, as is generally known. The immortalized cell lines secreting the desired antibodies can be screened by immunoassay in which the antigen is the peptide hapten, polypeptide or protein. When the appropriate immortalized cell culture secreting the desired antibody is identified, the cells can be cultured either in vitro or by production in ascites fluid. The desired monoclonal antibodies may be recovered from the culture supernatant or from the ascites supernatant. Fragments of the monoclonal antibodies or the polyclonal antisera which contain the immunologically significant portion(s) can be used as antagonists, as well as the intact antibodies. Use of immunologically reactive fragments, such as Fab or Fab' fragments, is often preferable, especially in a therapeutic context, as these fragments are generally less immunogenic than the whole immunoglobulin. The antibodies or fragments may also be produced, using current technology, by recombinant means. Antibody regions that bind specifically to the desired regions of the protein can also be produced in the context of chimeras derived from multiple species. Antibody regions that bind specifically to the desired regions of the protein can also be produced in the context of chimeras from multiple species, for instance, humanized antibodies. The antibody can therefore be a humanized antibody or a human antibody, as described in U.S. Patent 5,585,089 or Riechmann et al. (1988).
Agents that are assayed in the above method can be randomly selected or rationally selected or designed. As used herein, an agent is said to be randomly selected when the agent is chosen randomly without considering the specific sequences involved in the association of the a protein of the invention alone or with its associated substrates, binding partners, etc. An example of randomly selected agents is the use of a chemical library or a peptide combinatorial library, or a growth broth of an organism. As used herein, an agent is said to be rationally selected or designed when the agent is chosen on a non-random basis which takes into account the sequence of the target site or its conformation in connection with the agent's action Agents can be rationally selected or rationally designed by utilizing the peptide sequences that make up these sites For example, a rationally selected peptide agent can be a peptide whose ammo acid sequence is identical to or a derivative of any functional consensus site The agents of the present invention can be, as examples, oligonucleotides, antisense polynucleotides, interfering RNA, peptides, peptide mimetics, antibodies, antibody fragments, small molecules, vitamin derivatives, as well as carbohydrates Peptide agents of the invention can be prepared using standard solid phase (or solution phase) peptide synthesis methods, as is known in the art In addition, the DNA encoding these peptides may be synthesized using commercially available oligonucleotide synthesis instrumentation and produced recombinantly using standard recombinant production systems The production using solid phase peptide synthesis is necessitated if non-gene-encoded amino acids are to be included
Another class of agents of the present invention includes antibodies or fragments thereof that bind to a protein encoded by a gene in Tables 2-4 Antibody agents can be obtained by immunization of suitable mammalian subjects with peptides, containing as antigenic regions, those portions of the protein intended to be targeted by the antibodies (see section above of antibodies as probes for standard antibody preparation methodologies)
In yet another class of agents, the present invention includes peptide mimetics that mimic the three-dimensional structure of the protein encoded by a gene from Tables 2-4 Such peptide mimetics may have significant advantages over naturally occurring peptides 'ncludιng for
Figure imgf000045_0001
grea+er chemical stab'li+y enhanced pharmacological properties (half-life, absorption, potency, efficacy, etc ), altered specificity (e g , a broad-spectrum of biological activities), reduced antigenicity and others In one form, mimetics are peptide-containing molecules that mimic elements of protein secondary structure The underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions, such as those of antibody and antigen A peptide mimetic is expected to permit molecular interactions similar to the natural molecule In another form, peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template peptide These types of non-peptide compounds are also referred to as peptide mimetics or peptidomimetics (Fauchere, 1986; Veber & Freidinger, 1985; Evans et al., 1987) which are usually developed with the aid of computerized molecular modeling. Peptide mimetics that are structurally similar to therapeutically useful peptides may be used to produce an equivalent therapeutic or prophylactic effect. Generally, peptide mimetics are structurally similar to a paradigm polypeptide (i.e., a polypeptide that has a biochemical property or pharmacological activity), but have one or more peptide linkages optionally replaced by a linkage using methods known in the art. Labeling of peptide mimetics usually involves covalent attachment of one or more labels, directly or through a spacer (e.g., an amide group), to non-interfering position(s) on the peptide mimetic that are predicted by quantitative structure-activity data and molecular modeling. Such non- interfering positions generally are positions that do not form direct contacts with the macromolecule(s) to which the peptide mimetic binds to produce the therapeutic effect. Derivitization (e.g., labeling) of peptide mimetics should not substantially interfere with the desired biological or pharmacological activity of the peptide mimetic. The use of peptide mimetics can be enhanced through the use of combinatorial chemistry to create drug libraries. The design of peptide mimetics can be aided by identifying amino acid mutations that increase or decrease binding of the protein to its binding partners. Approaches that can be used include the yeast two hybrid method (see Chien et al., 1991 ) and the phage display method. The two hybrid method detects protein-protein interactions in yeast (Fields et al., 1989). The phage display method detects the interaction between an immobilized protein and a protein that is expressed on the surface of phages such as lambda and M13 (Amberg ef al., 1993; Hogrefe et al., 1993). These methods allow positive and negative selection for protein-protein interactions and the identification of the sequences that determine these interactions.
Method to diagnose baldness
The present invention also relates to methods for diagnosing baldness or a related disorder, preferably androgenic alopecia, a disposition to such disorder, predisposition to such a disorder and/or disorder progression. In some methods, the steps comprise contacting a target sample with (a) nucleic molecule(s) or fragments thereof and comparing the concentration of individual mRNA(s) with the concentration of the corresponding mRNA(s) from at least one healthy donor. An aberrant (increased or decreased) mRNA level of at least one gene from Tables 2-4, at least 5 or 10 genes from Tables 2-4, at least 20 genes from Tables 2-4, at least 50 genes from Tables 2-4 determined in the sample in comparison to the control sample is an indication of baldness or a related disorder or a disposition to such kinds of disorders For diagnosis, samples are from any parts of the body such as the scalp, blood, dermis, epidermis and other skin cells, cutaneous surfaces, intertπgious areas, genitalia, vessels and endothelium Some non-limiting examples of cells that can be used are hair (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes
For analysis of gene expression, total RNA is obtained from cells according to standard procedures and, preferably, reverse-transcribed Preferably, a DNAse treatment (in order to get rid of contaminating genomic DNA) is performed Some non-limiting examples of cells that can be used are hair (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes
The nucleic acid molecule or fragment is typically a nucleic acid probe for hybridization or a primer for PCR The person skilled in the art is in a position to design suitable nucleic acids probes based on the information provided in the Tables of the present invention The target cellular component, / e mRNA, e g , in skin, may be detected directly in situ, e g by in situ hybridization or it may be isolated from other cell components by common methods known to those skilled in the art before contacting with a probe Detection methods include Northern blot analysis, RNase protection, in situ methods e g in situ hybridization in vtm amplification r^e+hods (PCP LCR QRMA replicase or RNA-transcπption/amplification (TAS, 3SR), reverse dot blot disclosed in EP-B10237362) and other detection assays that are known to those skilled in the art Products obtained by in vitro amplification can be detected according to established methods, e g by separating the products on agarose or polyacrylamide gels and by subsequent staining with ethidium bromide Alternatively, the amplified products can be detected by using labeled primers for amplification or labeled dNTPs Preferably, detection is based on a microarray
The probes (or primers) (or, alternatively, the reverse-transcribed sample mRNAs) can be detectably labeled, for example, with a radioisotope, a bioluminescent compound, a chemiluminescent compound, a fluorescent compound, a metal chelate, or an enzyme The present invention also relates to the use of the nucleic acid molecules or fragments described above for the preparation of a diagnostic composition for the diagnosis of baldness or a disposition to such a disorder.
The present invention also relates to the use of the nucleic acid molecules of the present invention for the isolation or development of a compound which is useful for therapy of baldness. For example, the nucleic acid molecules of the invention and the data obtained using said nucleic acid molecules for diagnosis of baldness might allow for the identification of further genes which are specifically dysregulated, and thus may be considered as potential targets for therapeutic interventions.
The invention further provides prognostic assays that can be used to identify subjects having or at risk of developing baldness. In such method, a test sample is obtained from a subject and the amount and/or concentration of the nucleic acid described in Tables 2- 4 is determined; wherein the presence of an associated allele, a particular allele of a polymorphic locus, or the likes in the nucleic acids sequences of this invention (see SEQ ID from Tables 5, 6.1 and 6.2) can be diagnostic for a subject having or at risk of developing baldness. As used herein, a "test sample" refers to a biological sample obtained from a subject of interest. For example, a test sample can be a biological fluid, a cell sample, or tissue. A biological fluid can be, but is not limited to saliva, serum, mucus, urine, stools, spermatozoids, vaginal secretions, lymph, amiotic liquid, pleural liquid and tears. Cells can be, but are not limited to hair cells (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, bngeΦans cells, CD4 f and CD3+ T cells and lymphocytes
Furthermore, the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, polypeptide, nucleic acid such as antisense DNA or interfering RNA (RNAi), small molecule or other drug candidate) to treat baldness. Specifically, these assays can be used to predict whether an individual will have an efficacious response or will experience adverse events in response to such an agent. For example, such methods can be used to determine whether a subject can be effectively treated with an agent that modulates the expression and/or activity of a gene from Tables 2-4, or the nucleic acids described herein. In another example, an association study may be performed to identify polymorphisms from Tables 5, 6.1 and 6.2 that are associated with a given response to the agent, e.g., an efficacious response or the likelihood of one or more adverse events. Thus, one embodiment of the present invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with aberrant expression or activity of a gene from Tables 2-4, in which a test sample is obtained and nucleic acids or polypeptides from Tables 2-5, 6.1 and 6.2 are detected (e.g., wherein the presence of a particular level of expression of a gene from Tables 2-4 or a particular allelic variant of such gene, such as polymorphism from Tables 2-5, 6.1 and 6.2, is diagnostic for a subject that can be administered an agent to treat a disorder such as baldness). In one embodiment, the method includes obtaining a sample from a subject suspected of having baldness or an affected individual and exposing such sample to an agent. The expression and/or activity of the nucleic acids and or genes of the invention are monitored before and after treatment with such agent to assess the effect of such agent. After analysis of the expression values, one skilled in the art can determine whether such agent can effectively treat such subject. In another embodiment, the method includes obtaining a sample from a subject having or susceptible to developing baldness and determining the allelic constitution of polymorphisms from Table 1 that are associated with a particular response to an agent. After analysis of the allelic constitution of the individual at the associated polymorphisms, one skilled in the art can determine whether such agent can effectively treat such subject.
The methods of the invention can also be used to detect genetic alterations in a gene from Tables 2-4, thereby determining if a subject with the lesioned gene is at risk for a disorder associated with baldness. In preferred embodiments, the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic alteration characterized by at least one alteration linked to or affecting the integrity of a gene from Tables 2-4 encoding a polypeptide or the misexpression of such gene. For example, such genetic alterations can be detected by ascertaining the existence of at least one of: (1 ) a deletion of one or more nucleotides from a gene from Tables 2-4; (2) an addition of one or more nucleotides to a gene from Tables 2-4; (3) a substitution of one or more nucleotides of a gene from Tables 2-4; (4) a chromosomal rearrangement of a gene from Tables 2-4; (5) an alteration in the level of a messenger RNA transcript of a gene from Tables 2-4; (6) aberrant modification of a gene from Tables 2-4, such as of the methylation pattern of the genomic DNA, (7) the presence of a non-wild type splicing pattern of a messenger RNA transcript of a gene from Tables 2-4; (8) inappropriate post- translational modification of a polypeptide encoded by a gene from Tables 2-4; and (9) alternative promoter use. As described herein, there are a large number of assay techniques known in the art which can be used for detecting alterations in a gene from Tables 2-4. A preferred biological sample is a peripheral blood sample obtained by conventional means from a subject. Another preferred biological sample is a buccal swab. Other biological samples can be, but is not limited to, urine, stools, spermatozoids, vaginal secretions, lymph, amiotic liquid, pleural liquid and tears.
In certain embodiments, detection of the alteration involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Pat. Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et a/., 1988; and Nakazawa et al., 1994), the latter of which can be particularly useful for detecting point mutations in a gene from Tables 2-4 (see Abavaya et al., 1995). This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic DNA, mRNA, or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers which specifically hybridize to a gene from Tables 2-4 under conditions such that hybridization and amplification of the nucleic acid from Tables 2-4 (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with some of the techniques used for detecting a mutation, an associated allele, a particular allele of a polymorphic locus, or the like described herein.
Alternative amplification methods include: self sustained sequence replication (Guatelli et al., 1990), transcriptional amplification system (Kwoh et al., 1989), Q-Beta Replicase (Lizardi et al., 1988), isothermal amplification (e.g. Dean ef a/., 2002); and Hafner et al.,
2001 ), cr any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of ordinary skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low number.
In an alternative embodiment, alterations in a gene from Tables 2-4, from a sample cell can be identified by identifying changes in a restriction enzyme cleavage pattern. For example, sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates a mutation(s), an associated allele, a particular allele of a polymorphic locus, or the like in the sample DNA. Moreover, sequence specific ribozymes (see, e.g., U.S. Pat. No. 5,498,531 or DNAzyme e.g. U.S. Pat. No. 5,807,718) can be used to score for the presence of specific associated allele, a particular allele of a polymorphic locus, or the likes by development or loss of a ribozyme or DNAzyme cleavage site.
The present invention also relates to further methods for diagnosing baldness or a related disorder, preferably androgenic alopecia, a disposition to such disorder, predisposition to such a disorder and/or disorder progression. In some methods, the steps comprise contacting a target sample with (a) nucleic molecule(s) or fragments thereof and determining the presence or absence of a particular allele of a polymorphism that confers a disorder-related phenotype (e.g., predisposition to such a disorder and/or disorder progression). The presence of at least one allele from Tables 5, 6.1 and 6.2 that is associated with baldness disorder ("associated allele"), at least 5 or 10 associated alleles from Tables 5, 6.1 and 6.2, at least 50 associated alleles from Tables 5, 6.1 and 6.2, at least 100 associated alleles from Tables 5, 6.1 and 6.2, or at least 200 associated alleles from Table 5-7 determined in the sample is an indication of baldness or a related disorder, a disposition or predisposition to such kinds of disorders, or a prognosis for such disorder progression. Samples may be obtained from any parts of the body such as the scalp, blood, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, genitalia, vessels and endothelium. Some non-limiting examples of cells that can be used are hair (follicle, bulb, root, shaft and papilla) cells, scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
In other embodiments, alterations in a gene from Tables 2-4 can be identified by hybridizing sample and control nucleic acids, e.g., DNA or RNA, to high density arrays or bead arrays containing tens to thousands of oligonucleotide probes (Cronin et al., 1996; Kozal et al., 1996). For example, alterations in a gene from Tables 2-4 can be identified in two dimensional arrays containing light-generated DNA probes as described in Cronin et al., (1996). Briefly, a first hybridization array of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear arrays of sequential overlapping probes. This step allows the identification of point mutations, associated alleles, particular alleles of a polymorphic locus, or the like. This step is followed by a second hybridization array that allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants, mutations, alleles detected Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene
In yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence a gene from Tables 2-4 and detect an associated allele, a particular allele of a polymorphic locus, or the like by comparing the sequence of the sample gene from Tables 2-4 with the corresponding wild-type (control) sequence Examples of sequencing reactions include those based on techniques developed by Maxam and Gilbert (1977) or Sanger (1977) It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays (Bio/Techniques 19 448, 1995) including sequencing by mass spectrometry (see, e g PCT International Publication No WO 94/16101 , Cohen et al , 1996, and Griffin ef al 1993), real-time pyrophosphate sequencing method (Ronaghi et al , 1998, and Permutt et al , 2001 ) and sequencing by hybridization (see e g Drmanac ef al , 2002)
Other methods of detecting an associated allele, a particular allele of a polymorphic locus, or the likes in a gene from Tables 2-4 include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA, DNA/DNA or RNA/DNA heteroduplexes (Myers et al , 1985) In general, the art technique of "mismatch cleavage" starts by providing heteroduplexes formed by hybridizing (labeled) RNA or DNA containing the wild-type gene from Tables 2-4 sequence with potentially mutant RNA or DNA obtained from a tissue sample The double-stranded duplexes are treated with an agent that cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the cort/cl and sa^p'e strands cor instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with S1 nuclease to enzymatically digest the mismatched regions In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of an associated allele, a particular allele of a polymorphic locus, or the like (see, for example, Cotton et al , 1988, Saleeba ef a/ , 1992) In a preferred embodiment, the control DNA or RNA can be labeled for detection, as described herein In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point an associated allele, a particular allele of a polymorphic locus, or the likes in a gene from Tables 2-4 cDNAs obtained from samples of cells. For example, the imutY enzyme of E. coli cleaves A at G/A mismatches (Hsu ef a/., 1994). Other examples include, but are not limited to, the MutHLS enzyme complex of E. coli (Smith and Modrich., 1996) and CeI 1 from the celery (Kulinski ef a/., 2000) both cleave the DNA at various mismatches. According to an exemplary embodiment, a probe based on a gene sequence from Tables 2-4 is hybridized to a cDNA or other DNA product from a test cell or cells. The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected using electrophoresis protocols or the like. See, for example, U.S. Pat. No. 5,459,039. Alternatively, the screen can be performed in vivo following the insertion of the heteroduplexes in an appropriate vector. The whole procedure is known to those ordinary skilled in the art and is referred to as mismatch repair detection (see e.g. Fakhrai-Rad ef a/., 2004).
In other embodiments, alterations in electrophoretic mobility can be used to identify an associated allele, a particular allele of a polymorphic locus, or the likes in genes from Tables 2-4. For example, single strand conformation polymorphism (SSCP) can be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids (Orita ef a/., 1993; see also Cotton, 1993; and Hayashi ef a/., 1992). Single- stranded DNA fragments of sample and control nucleic acids from Tables 2-4 will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence; the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In a preferred embodiment, the method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Kee et al., 1991 ).
In yet another embodiment, the movement of mutant or wild-type fragments in a polyacrylamide gel containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers ef a/., 1985). When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum ef al., 1987). In another embodiment, the mutant fragment is detected using denaturing HPLC (see e.g. Hoogendoorn et al., 2000).
Examples of other techniques for detecting point mutations, an associated allele, a particular allele of a polymorphic locus, or the like include, but are not limited to, selective oligonucleotide hybridization, selective amplification, selective primer extension, selective ligation, single-base extension, selective termination of extension or invasive cleavage assay. For example, oligonucleotide primers may be prepared in which the known associated allele, particular allele of a polymorphic locus, or the like is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al., 1986; Saiki ef a/., 1989). Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different associated allele, a particular allele of a polymorphic locus, or the likes where the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA. Alternatively, the amplification, the allele-specific hybridization and the detection can be done in a single assay following the principle of the 5' nuclease assay (e.g. see Livak ef al., 1995). For example, the associated allele, a particular allele of a polymorphic locus, or the like locus is amplified by PCR in the presence of both allele- specific oligonucleotides, each specific for one or the other allele. Each probe has a different fluorescent dye at the 5' end and a quencher at the 3' end. During PCR, if one or the other or both allele-specific oligonucleotides are hybridized to the template, the Taq polymerase via its 5' exonuclease activity will release the corresponding dyes. The latter will thus reveal the genotype of the amplified product.
The hybridization may also be carried out with a temperature gradient following the principle of dynamic allele-specific hybridization or like (e.g. Jobs ef al., 2003; and Bourgeois and Labuda, 2004). For example, the hybridization is done using one of the two allele-specific oligonucleotides labeled with a fluorescent dye, an intercalating quencher under a gradually increasing temperature. At low temperature, the probe is hybridized to both the mismatched and full-matched template. The probe melts at a lower temperature when hybridized to the template with a mismatch. The release of the probe is captured by an emission of the fluorescent dye, away from the quencher. The probe melts at a higher temperature when hybridized to the template with no mismatch. The temperature-dependent fluorescence signals therefore indicate the absence or presence of the associated allele, particular allele of a polymorphic locus, or the like (e.g. Jobs et al. supra). Alternatively, the hybridization is done under a gradually decreasing temperature. In this case, both allele-specific oligonucleotides are hybridized to the template competitively. At high temperature none of the two probes is hybridized. Once the optimal temperature of the full-matched probe is reached, it hybridizes and leaves no target for the mismatched probe. In the latter case, if the allele-specific probes are differently labeled, then they are hybridized to a single PCR-amplified target. If the probes are labeled with the same dye, then the probe cocktail is hybridizes twice to identical templates with only one labeled probes, different in the two cocktails, in the presence of the unlabeled competitive probe.
Alternatively, allele specific amplification technology that depends on selective PCR amplification may be used in conjunction with the present invention. Oligonucleotides used as primers for specific amplification may carry the associated allele, particular allele of a polymorphic locus, or the like of interest in the center of the molecule, so that amplification depends on differential hybridization (Gibbs et al., 1989) or at the extreme 31 end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner, 1993). In addition it may be desirable to introduce a novel restriction site in the region of the associated allele, particular allele of a polymorphic locus, or the like to create cleavage-based detection (Gasparini et al., 1992). It is anticipated that in certain embodiments amplification may also be performed using Taq ligase for amplification (Barany, 1991 ). In such cases, ligation will occur only if there is a perfect match at the 3' end of the 5' sequence making it possible to detect the presence of a known associated allele, a particular allele of a polymorphic locus, or the like at a specific site by looking for the presence or absence of amplification. The products of such an oligonucleotide ligation assay can also be detected by means of gel electrophoresis. Furthermore, the oligonucleotides may contain universal tags used in PCR amplification and zip code tags that are different for each allele. The zip code tags are used to isolate a specific, labeled oligonucleotide that may contain a mobility modifier (e.g. Grossman ef a/., 1994).
In yet another alternative, allele-specific elongation followed by ligation will form a template for PCR amplification. In such cases, elongation will occur only if there is a perfect match at the 3' end of the allele-specific oligonucleotide using a DNA polymerase. This reaction is performed directly on the genomic DNA and the extension/ligation products are amplified by PCR To this end, the oligonucleotides contain universal tags allowing amplification at a high multiplex level and a zip code for SNP identification The PCR tags are designed in such a way that the two alleles of a SNP are amplified by different forward primers, each having a different dye The zip code tags are the same for both alleles of a given SNP and they are used for hybridization of the PCR-amplified products to oligonucleotides bound to a solid support, chip, bead array or like For an example of the procedure, see Fan et al (Cold Spring Harbor Symposia on Quantitative Biology, VoI LXVIII, pp 69-78, 2003)
Another alternative includes the single-base extension/ligation assay using a molecular inversion probe, consisting of a single, long oligonucleotide (see e g Hardenbol et al , 2003) In such an embodiment, the oligonucleotide hybridizes on both side of the SNP locus directly on the genomic DNA, leaving a one-base gap at the SNP locus The gap- filhng, one-base extension/ligation is performed in four tubes, each having a different dNTP Following this reaction, the oligonucleotide is circularized whereas unreactive, linear oligonucleotides are degraded using an exonulease such as exonuclease I of E coli The circular oligonucleotides are then linearized and the products are amplified and labeled using universal tags on the oligonucleotides The original oligonucleotide also contains a SNP-specific zip code allowing hybridization to oligonucleotides bound to a solid support, chip, bead array or like This reaction can be performed at a highly multiplexed level
In another alternative, the associated allele, particular allele of a polymorphic locus, or the like is scored by single-base extension (see e g U S Pat No 5,888,819) The template is first amplified by PCR The extension oligonucleotide is then hybridized next to +he SNP locus and the extension reaction is performed LG'ng a thermostable polymerase such as ThermoSequenase (GE Healthcare) in the presence of labeled ddNTPs This reaction can therefore be cycled several times The identity of the labeled ddNTP incorporated will reveal the genotype at the SNP locus The labeled products can be detected by means of gel electrophoresis, fluorescence polarization (e g Chen et al., 1999) or by hybridization to oligonucleotides bound to a solid support, chip, bead array or like In the latter case, the extension oligonucleotide will contain a SNP-specific zip code tag
In yet another alternative, the variant is scored by selective termination of extension The template is first amplified by PCR and the extension oligonucleotide hybridizes in vicinity to the SNP locus, close to but not necessarily adjacent to it The extension reaction is carried out using a thermostable polymerase such as Thermo Sequenase (GE Healthcare) in the presence of a mix of dNTPs and at least one ddNTP. The latter has to terminate the extension at one of the alleles of the interrogated SNP, but not both such that the two alleles will generate extension products of different sizes. The extension product can then be detected by means of gel electrophoresis, in which case the extension products need to be labeled, or by mass spectrometry (see e.g. Storm et al., 2003).
In another alternative, the associated allele, particular allele of a polymorphic locus, or the like is detected using an invasive cleavage assay (see U.S. Pat. No. 6,090,543). There are five oligonucleotides per SNP to interrogate but these are used in a two step- reaction. During the primary reaction, three of the designed oligonucleotides are first hybridized directly to the genomic DNA. One of them is locus-specific and hybridizes up to the SNP locus (the pairing of the 3' base at the SNP locus is not necessary). There are two allele-specific oligonucleotides that hybridize in tandem to the locus-specific probe but also contain a 5' flap that is specific for each allele of the SNP. Depending upon hybridization of the allele-specific oligonucleotides at the base of the SNP locus, this creates a structure that is recognized by a cleavase enzyme (U.S. Pat. No. 6,090,606) and the allele-specific flap is released. During the secondary reaction, the flap fragments hybridize to a specific cassette to recreate the same structure as above except that the cleavage will release a small DNA fragment labeled with a fluorescent dye that can be detected using regular fluorescence detector. In the cassette, the emission of the dye is inhibited by a quencher.
Other types of markers can also be used for diagnostic purposes. For example, microsatellites can also be useful to detect the genetic predisposition of an individual to a given disorder. Microsatellites consist of short sequence motifs of one or a few nucleotides repeated in tandem. The most common motifs are polynucleotide runs, dinucleotide repeats (particularly the CA repeats) and trinucleotide repeats. However, other types of repeats can also be used. The microsatellites are very useful for genetic mapping because they are highly polymorphic in their length. Microsatellite markers can be typed by various means, including but not limited to DNA PCR fragment sizing, oligonucleotide ligation assay and mass spectrometry. For example, the locus of the microsatellite is amplified by PCR and the size of the PCR fragment will be directly correlated to the length of the microsatellite repeat. The size of the PCR fragment can be detected by regular means of gel electrophoresis. The fragment can be labeled internally during PCR or by using end-labeled oligonucleotides in the PCR reaction (e.g. Mansfield et al., 1996). Alternatively, the size of the PCR fragment is determined by mass spectrometry. In such a case, however, the flanking sequences need to be eliminated. This can be achieved by ribozyme cleavage of an RNA transcript of the microsatellite repeat (Krebs et al., 2001 ). For example, the microsatellite locus is amplified using oligonucleotides that include a T7 promoter on one end and a ribozyme motif on the other end. Transcription of the amplified fragments will yield an RNA substrate for the ribozyme, releasing small RNA fragments that contain the repeated region. The size of the latter is determined by mass spectrometry. Alternatively, the flanking sequences are specifically degraded. This is achieved by replacing the dTTP in the PCR reaction by dUTP. The dUTP nucleosides are then removed by uracyl DNA glycosylases and the resulting basic sites are cleaved by either abasic endonucleases such as human AP endonuclease or chemical agents such as piperidine. Bases can also be modified post- PCR by chemical agents such as dimethyl sulfate and then cleaved by other chemical agents such as piperidine (see e.g. Maxam and Gilbert, 1977; U.S. Pat. No. 5,869,242; and U.S. Patent pending serial No. 60/335,068).
In another alternative, an oligonucleotide ligation assay can be performed. The microsatellite locus is first amplified by PCR. Then, different oligonucleotides can be submitted to ligation at the center of the repeat with a set of oligonucleotides covering all the possible lengths of the marker at a given locus (Zirvi ef al., 1999). Another example of design of an oligonucleotide assay comprises the ligation of three oligonucleotides; a 5' oligonucleotide hybridizing to the 5' flanking sequence, a repeat oligonucleotide of the length of the shortest allele of the marker hybridizing to the repeated region and a set of 3' oligonucleotides covering all the existing alleles hybridizing to the 3' flanking sequence and a portion of the repeated region for all the alleles longer than the snortest one. For the shortest allele, the 3' oligonucleotide exclusively hybridizes to the 3' flanking sequence (U.S. Pat. No. 6,479,244).
The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid selected from the SEQ ID of Tables 2-5, 6.1 and 6.2, or antibody reagent described herein, which may be conveniently used, for example, in a clinical setting to diagnose patient exhibiting symptoms or a family history of a disorder or disorder involving abnormal activity of genes from Tables 2-4. Method to treat an animal suspected of having baldness
The present invention provides methods of treating a disorder associated with baldness by expressing in vivo the nucleic acids of at least one gene from Tables 2-4. These nucleic acids can be inserted into any of a number of well-known vectors for the transfection of target cells and organisms as described below. The nucleic acids are transfected into cells, ex vivo or in vivo, through the interaction of the vector and the target cell. The nucleic acids encoding a gene from Tables 2-4, under the control of a promoter, then expresses the encoded protein, thereby mitigating the effects of absent, partial inactivation, or abnormal expression of a gene from Tables 2-4.
Such gene therapy procedures have been used to correct acquired and inherited genetic defects, cancer, and viral infection in a number of contexts. The ability to express artificial genes in humans facilitates the prevention and/or cure of many important human disorders, including many disorders which are not amenable to treatment by other therapies (for a review of gene therapy procedures, see Anderson, 1992; Nabel & Feigner, 1993; Mitani & Caskey, 1993; Mulligan, 1993; Dillon, 1993; Miller, 1992; Van Brunt, 1998; Vigne, 1995; Kremer & Perricaudet 1995; Doerfler & Bohm 1995; and Yu et a/., 1994).
Delivery of the gene or genetic material into the cell is the first critical step in gene therapy treatment of disorder. A large number of delivery methods are well known to those of skill in the art. Preferably, the nucleic acids are administered for in vivo or ex vivo gene therapy uses. Non-viral vector delivery systems include DNA plasmids, naked nucleic acid, and nucleic acid complexed with a delivery vehicle such as a liposome. Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell. For a review of gene therapy procedures, see the references included in the above section.
The use of RNA or DNA viral based systems for the delivery of nucleic acids take advantage of highly evolved processes for targeting a virus to specific cells in the body and trafficking the viral payload to the nucleus. Viral vectors can be administered directly to patients (in vivo) or they can be used to treat cells in vitro and the modified cells are administered to patients (ex vivo). Conventional viral based systems for the delivery of nucleic acids could include retroviral, lentivirus, adenoviral, adeno-associated and herpes simplex virus vectors for gene transfer. Viral vectors are currently the most efficient and versatile method of gene transfer in target cells and tissues. Integration in the host genome is possible with the retrovirus, lentivirus, and adeno-associated virus gene transfer methods, often resulting in long term expression of the inserted transgene. Additionally, high transduction efficiencies have been observed in many different cell types and target tissues.
The tropism of a retrovirus can be altered by incorporating foreign envelope proteins, expanding the potential target population of target cells. Lentiviral vectors are retroviral vector that are able to transduce or infect non-dividing cells and typically produce high viral titers. Selection of a retroviral gene transfer system would therefore depend on the target tissue. Retroviral vectors are comprised of cis-acting long terminal repeats with packaging capacity for up to 6-10 kb of foreign sequence. The minimum cis-acting LTRs are sufficient for replication and packaging of the vectors, which are then used to integrate the therapeutic gene into the target cell to provide permanent transgene expression. Widely used retroviral vectors include those based upon murine leukemia virus (MuLV), gibbon ape leukemia virus (GaLV), Simian lmmuno deficiency virus (SIV), human immuno deficiency virus (HIV), and combinations thereof (see, e.g., Buchscher ef al., 1992; Johann ef al., 1992; Sommerfelt ef al., 1990; Wilson et al., 1989; Miller et a/.,1999;and PCT/US94/05700).
In applications where transient expression of the nucleic acid is preferred, adenoviral based systems are typically used. Adenoviral based vectors are capable of very high transduction efficiency in many cell types and do not require cell division. With such vectors, high titer and levels of expression have been obtained. This vector can be produced in large quantities in a relatively simple system. Adeno-associated virus ("AAV") vectors are also used to transduce cells with target nucleic acids, e.g., in the in vitro production of nucleic acidε ard pept'des, and for in vivo and εx vivo gene therapy procedures (see, e.g., West ef al., 1987; U.S. Pat. No. 4,797,368; WO 93/24641 ; Kotin, 1994; Muzyczka, 1994). Construction of recombinant AAV vectors are described in a number of publications, including U.S. Pat. No. 5,173,414; Tratschin ef al., 1985; Tratschin, et al., 1984; Hermonat & Muzyczka, 1984; and Samulski et al., 1989.
In particular, numerous viral vector approaches are currently available for gene transfer in clinical trials, with retroviral vectors by far the most frequently used system. All of these viral vectors utilize approaches that involve complementation of defective vectors by genes inserted into helper cell lines to generate the transducing agent. pLASN and MFG- S are examples are retroviral vectors that have been used in clinical trials (Dunbar ef al., 1995; Kohn ef al., 1995; Malech ef al., 1997). PA317/pLASN was the first therapeutic vector used in a gene therapy trial (Blaese ef al., 1995). Transduction efficiencies of 50% or greater have been observed for MFG-S packaged vectors (Ellem ef al., 1997; and Dranoff et al., 1997).
Recombinant adeno-associated virus vectors (rAAV) are a promising alternative gene delivery systems based on the defective and nonpathogenic parvovirus adeno- associated type 2 virus. All vectors are derived from a plasmid that retains only the AAV 145 bp inverted terminal repeats flanking the transgene expression cassette. Efficient gene transfer and stable transgene delivery due to integration into the genomes of the transduced cell are key features for this vector system. (Wagner ef a/., 1998, Kearns ef a/1996).
Replication-deficient recombinant adenoviral vectors (Ad) are predominantly used in transient expression gene therapy; because they can be produced at high titer and they readily infect a number of different cell types. Most adenovirus vectors are engineered such that a transgene replaces the Ad E1 a, E1 b, and E3 genes; subsequently the replication defector vector is propagated in human 293 cells that supply deleted gene function in trans. Ad vectors can transduce multiple types of tissues in vivo, including nondividing, differentiated cells such as those found in the liver, kidney and muscle system tissues. Conventional Ad vectors have a large carrying capacity. An example of the use of an Ad vector in a clinical trial involved polynucleotide therapy for antitumor immunization with intramuscular injection (Sterman et al., 1998). Additional examples of the use of adenovirus vectors for gene transfer in clinical trials include Rosenecker ef al., 1996; Sterman ef a/., 1998; Welsh ef a/., 1995; Alvarez ef a/., 1997; Topf ef a/., 1998.
Packaging cells are used to form virus particles that are capable of infecting a host cell. Such cells include 293 cells, which package adenovirus, and ψ2 cells or PA317 cells, which package retrovirus. Viral vectors used in gene therapy are usually generated by producer cell line that packages a nucleic acid vector into a viral particle. The vectors typically contain the minimal viral sequences required for packaging and subsequent integration into a host, other viral sequences being replaced by an expression cassette for the protein to be expressed. The missing viral functions are supplied in trans by the packaging cell line. For example, AAV vectors used in gene therapy typically only possess ITR sequences from the AAV genome which are required for packaging and integration into the host genome. Viral DNA is packaged in a cell line, which contains a helper plasmid encoding the other AAV genes, namely rep and cap, but lacking ITR sequences. The cell line is also infected with adenovirus as a helper. The helper virus promotes replication of the AAV vector and expression of AAV genes from the helper plasmid. The helper plasmid is not packaged in significant amounts due to a lack of ITR sequences. Contamination with adenovirus can be reduced by, e.g., heat treatment to which adenovirus is more sensitive than AAV.
In many gene therapy applications, it is desirable that the gene therapy vector be delivered with a high degree of specificity to a particular tissue type. A viral vector is typically modified to have specificity for a given cell type by expressing a ligand as a fusion protein with a viral coat protein on the viruses outer surface. The ligand is chosen to have affinity for a receptor known to be present on the cell type of interest. For example, Han et al., 1995, reported that Moloney murine leukemia virus can be modified to express human heregulin fused to gp70, and the recombinant virus infects certain human breast cancer cells expressing human epidermal growth factor receptor. This principle can be extended to other pairs of virus expressing a ligand fusion protein and target cell expressing a receptor. For example, filamentous phage can be engineered to display antibody fragments (e.g., Fab or Fv) having specific binding affinity for virtually any chosen cellular receptor. Although the above description applies primarily to viral vectors, the same principles can be applied to nonviral vectors. Such vectors can be engineered to contain specific uptake sequences thought to favor uptake by specific target cells.
Gene therapy vectors can be delivered in vivo by administration to an individual patient, typically by systemic administration (e.g., intravenous, intraperitoneal, intramuscular, subdermai, or intracranial infusion) or topical application. Alternatively, vectors can be delivered to cells ex vivo, such as cells explanted from an individual patient (e.g., lymphocytes, bone marrow aspirates, and tissue biopsy) or universal donor hematopoietic stem cells, followed by reimplantation of the cells into a patient, usually after selection for cells which have incorporated the vector.
Ex vivo cell transfection for diagnostics, research, or for gene therapy (e.g., via re- infusion of the transfected cells into the host organism) is well known to those of skill in the art. In a preferred embodiment, cells are isolated from the subject organism, transfected with a nucleic acid (gene or cDNA), and re-infused back into the subject organism (e.g., patient). Various cell types suitable for ex vivo transfection are well known to those of skill in the art (see, e.g., Freshney et al., 1994; and the references cited therein for a discussion of how to isolate and culture cells from patients). In one embodiment, stem cells are used in ex vivo procedures for cell transfection and gene therapy The advantage to using stem cells is that they can be differentiated into other cell types in vitro, or can be introduced into a mammal (such as the donor of the cells) where they will engraft in the bone marrow Methods for differentiating CD34+ cells in vitro into clinically important immune cell types using cytokines such a GM-CSF, IFN-γ and TNF-α are known (see lnaba ef a/ , 1992)
Stem cells are isolated for transduction and differentiation using known methods For example, stem cells are isolated from bone marrow cells by panning the bone marrow cells with antibodies which bind unwanted cells, such as CD4+ and CD8+ (T cells), CD45+ (panB cells), GR-1 (granulocytes), and lad (differentiated antigen presenting cells)
Vectors (e g , retroviruses, adenoviruses, liposomes, etc ) containing therapeutic nucleic acids can be also administered directly to the organism for transduction of cells in vivo Alternatively, naked DNA can be administered
Administration is by any of the routes normally used for introducing a molecule into ultimate contact with blood or tissue cells, as described above The nucleic acids from Tables 2-4 are administered in any suitable manner, preferably with the pharmaceutically acceptable carriers described above Suitable methods of administering such nucleic acids are available and well known to those of skill in the art, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route (see Samulski et al , 1989) The present invention is not limited to any method of administering such nucleic acids but preferentially uses the methods described herein
The present invention further provides other methods of treating baldness such as administering to an individual having baldness an effective amount of an agent that regulates the expression, activity or physical state of at least one gene from Tables 2-4 An "effective amount" of an agent is an amount that modulates a level of expression or activity of a gene from Tables 2-4, in a cell in the individual at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80% or more, compared to a level of the respective gene from Tables 2-4 in a cell in the individual in the absence of the compound The preventive or therapeutic agents of the present invention may be administered, either orally or parenterally, systemically or locally For example, intravenous injection such as drip infusion, intramuscular injection, intraperitoneal injection, subcutaneous injection, suppositories, intestinal lavage, oral enteric coated tablets, and the like can be selected, and the method of administration may be chosen, as appropriate, depending on the age and the conditions of the patient. The effective dosage is chosen from the range of 0.01 mg to 100 mg per kg of body weight per administration. Alternatively, the dosage in the range of 1 to 1000 mg, preferably 5 to 50 mg per patient may be chosen. The therapeutic efficacy of the treatment may be monitored by observing various parts of the body, such as the scalp, by any monitoring method known in the art. Others ways of monitoring efficacy can be, but are not limited to monitoring hair growth, size of hair follicles, length and thickness of hair, activity of hair follicles, hair loss and status of hair at the crown of the head.
The present invention further provides a method of treating an individual clinically diagnosed with baldness. The methods generally comprises analyzing a biological sample that includes a cell, in some cases, a skin cell, from an individual clinically diagnosed with baldness for the presence of modified levels of expression of at least 1 gene, at least 10 genes, at least 50 genes from Tables 2-4. A treatment plan that is most effective for individuals clinically diagnosed as having a condition associated with baldness is then selected on the basis of the detected expression of such genes in a cell. Treatment may include administering a composition that includes an agent that modulates the expression or activity of a protein from Tables 2-4 in the cell. Information obtained as described in the methods above can also be used to predict the response of the individual to a particular agent. Thus, the invention further provides a method for predicting a patient's likelihood to respond to a drug treatment for a condition associated with baldness, comprising determining whether modified levels of a gene from Tables 2- 4 is present in a cell, wherein tne presence of protein is predictive of the patient's likelihood to respond to a drug treatment for the condition. Examples of the prevention or improvement of symptoms accompanied by baldness that can monitor for effectiveness include prevention or improvement of hair growth on the scalp, the size of hair follicles, length and thickness of hair, activity of hair follicles, hair loss and status of hair at the crown of the head.
The invention also provides a method of predicting a response to therapy in a subject having baldness by determining the presence or absence in the subject of one or more markers associated with baldness described in Tables 2-5, 6.1 and 6.2, diagnosing the subject in which the one or more markers are present as having baldness, and predicting a response to a therapy based on the diagnosis e.g., response to therapy may include an efficacious response and/or one or more adverse events. The invention also provides a method of optimizing therapy in a subject having baldness by determining the presence or absence in the subject of one or more markers associated with a clinical subtype of baldness, diagnosing the subject in which the one or more markers are present as having a particular clinical subtype of baldness, and treating the subject having a particular clinical subtype of baldness based on the diagnosis. As an example, baldness can be treated with surgical procedures, such as hair transplants and scalp reduction. Recently, however, there have been some advances in medical treatment of baldness. The most publicized of these is minoxidil (Rogaine). Another treatment is the topical use of progesterone, a molecule similar to testosterone. The side effects of this treatment are not known and again this treatment has not proven to be efficient on a vast majority of balding individuals.
Thus, while there are a number of treatments for baldness currently available, they all are accompanied by various side effects, high costs, and long complicated treatment protocols, which are often not available and effective in a large number of balding individuals. Accordingly, there remains a need in the art for more effective and otherwise improved methods for treating and preventing baldness. Thus, there is a continuing need in the medical arts for genetic markers of baldness and guidance for the use of such markers. The present invention fulfills this need and provides further related advantages.
EXAMPLES
Example 1 : Identification of cases and controls
All individuals were sampled from the Quebec founder population (QFP). Membership in the founder population was defined as having four grandparents with French Canadian family names who were born in the Province of Quebec, Canada or in adjacent areas of the Provinces of New Brunswick and Ontario or in New England or New York State. The Quebec founder population has two distinct advantages over general populations for LD mapping. Because it is relatively young, about 12 to 15 generations from mid 17th century to present, and because it has a limited but sufficient number of founders, approximately 2600 effective founders (Charbonneau et al. 1987), the Quebec population is characterized both by extended LD and by decreased genetic heterogeneity. The increased extent of LD allows the detection of disorder genes using a reasonable marker density, while still allowing the increased meiotic resolution of population-based mapping. The number of founders is small enough to result in increased LD and reduced allelic heterogeneity, yet large enough to insure that all of the major disorder genes involved in general populations are present in Quebec. Reduced allelic heterogeneity will act to increase relative risk imparted by the remaining alleles and so increase the power of case/control studies to detect gene an associated allele, a particular allele of a polymorphic locus, or the likes involved in complex disorders within the Quebec population. The specific combination of age in generations, optimal number of founders and large present population size makes the QFP optimal for LD-based gene mapping. The existence of close family relationships among the selected individuals is verified. When subjects are linked by a common relative, according to the information available, only one is kept for the study.
Patient inclusion criteria for the study included diagnosis for baldness with the onset of the condition occurring before age 25.
Patients that were diagnosed with baldness due to infection, stress, medication, burns or dermatological lesions (such as scaring) were excluded from the study. All human sampling was subject to ethical review procedures.
All enrolled QFP subjects (patients and controls) provided a 30 ml blood sample (3 barcoded tubes of 10 ml). Samples were processed immediately upon arrival at Genizon's laboratory. All samples were scanned and logged into a LabVantage Laboratory Information Management System (LIMS), which served as a hub between the clinical data management system and the genetic analysis system. Following centrifugation, the buffy coat containing the white blood cells was isolated from each tube. Genomic DNA was extracted from the buffy coat from one of the tubes, and stored at 4°C until required for genotyping. DNA extraction was performed with a commercial kit using a guanidine hydrochloride based method (FlexiGene, Qiagen) according to the manufacturer's instructions. The extraction method yielded high molecular weight DNA, and the quality of every DNA sample was verified by agarose gel electrophoresis. Genomic DNA appeared on the gel as a large band of very high molecular weight. The remaining two buffy coats were stored at -800C as backups.
The samples were collected as 468 male patients (cases) and 470 male controls. The DNA extracted from cases and control samples is pooled together in various pools. Pools are designed to segregate probands (cases) from controls.
Proband pools, consisting of DNA extracted from recruited cases, were separated according to level of baldness as determined on the Norwood/Hamilton scale (Norwood 1975), and further separated by age group. Six proband pools consisted of Norwood/Hamilton type Vl, and 2 proband pools consisted of Norwood/Hamilton type VII. The control poolsconsisted of 5 pools of 57 samples with Norwood/Hamilton type I, and 3 pools of 60, 60, or 65 samples with Norwood/Hamilton type II.
Example 2: Genome Wide Association
Genotyping was performed using Perlegen's ultra-high-throughput platform. Loci of interest were amplified and hybridized to wafers containing arrays of oligonucleotides. Allele discrimination was performed through allele-specific hybridization. In total, 248,535 SNPs, spread over 3 microarrays, were genotyped. This set of markers contained the QLDM (Quebec LD Map), a map created specifically for the Quebec founder population, which possesses a base density of one marker per 40 kb and up to one marker per 10 kb in low-LD regions, the lower the LD is in a given area, the higher the marker density will be. The QLDM markers and other markers were selected from various databases including the -1.6 million SNP database of Perlegen Life Sciences (Patil, 2001 ), the hapmap consortium database and dbSNP at NCBI. The SNPs were chosen to maximize uniformity of genetic coverage and as much as possible, with a minor allele frequency of 10% or higher. The genotyping information was entered into a Unified Genotype Database (a proprietary database under development) from which it was accessed using custom-built programs for export to the genetic analysis pipeline. Analyses of these genotypes were performed with the statistical tools described in Example 3. The GWS permitted the identification of 65 candidate regions that are further analyzed by the Fine Mapping approache described below. Four candidate regions out of the 65 were presently confirmed use the Fine Mapping approach described below. The rest of the candidate regions Fine Mapping is ongoing.
Example 3: Genetic Analysis
The raw data generated by the GWS approach (Example 2 herein) was analyzed by various means to identify candidate regions (see also Fine Mapping described in Example 5).
Raw data analysis by Perlegen
The data analysis process compares the relative fluorescence intensities of features corresponding to the reference allele of a given SNP with those corresponding to the alternate allele, to calculate a p-hat value. The latter is proportional to the fluorescence signal from perfect match features for the reference allele divided by the sum of fluorescence signals from perfect match features for the reference plus the alternate alleles. P-hat assumes values close to 1 (typically 0.9) for pure reference samples and close to 0 (typically 0.1 ) for pure alternate samples, and can be used as a measured estimate of the reference allele frequency of a SNP in a DNA pool. The difference between case and controi poois, delta p-hat, is calcuiaied using the weigπted average of case and control p-hats. Delta p-hat is a reliable estimate of the allele frequency difference between the cases and controls.
Data analysis by Genizon
Analysis of the data by Genizon was based upon Perlegen's p-hat values.
Data analysis of p-hat.
The data consisted of 8 pools of cases and 8 pools of controls, with a p-hat value for each pool (provided by Perlegen's analysis methods described above). The weighted mean p-hat value for all case pools was calculated as 2-ι < < Z-ι < where Ni is the number of alleles (2 X the number of people) in each case pool and the weighted
p-hat value for all control pools were calculated as Z-. y J 2-, i where Li is the number of alleles in each control pool. The estimate of delta-phat, the difference in allele frequency between cases and controls was X - Y and the significance of delta-phat was X - Y
determined using a Student's t statistic where ^ '' "-^ ' ' ^ ' ' ' where Sp is the pooled variance under the assumption that both case and control variances are
equal. The variance of T X was calculated as sx = ( /A 11 )^∑ N, (Phat, ~ *J and
similarly the variance of - v« was calculated as SY = ( /Al °-& i L{phat, -Yj . The
Student's t test was performed with 8+8-2 = 14 degrees of freedom. Single Marker P values were calculated for all markers within the genome wide scan map.
Combined P values
In addition to single marker P values, combined P values across multi-marker sliding windows were calculated after the method of Fisher (Statistical Methods for Research
Y1 = Yin/3 Workers, 14th edition Hafner Press N.Y. 1970 pp 99-100) where Λ *-> ' with 2k degrees of freedom where Pi is the P value for association of each of k markers within the sliding window. The combined P values identify regions of multiple single marker associations. However, Fisner's method assumes independence of association for each marker which id known not to be the case because of strong LD between adjacent markers. Therefore the magnitude of the combined P value was affected both by LD and association and these cannot be taken at face value. The combined P values were best used as a means to identify regions of multi-marker association which can then be assessed on the basis of the magnitude of single marker association.
Example 4: Fine Mapping
Four of the top regions identified as being associated with baldness by the GWS, were further analyzed by Fine mapping (genotyping all cases and controls samples individually) using a denser set of markers, in order to validate and/or refine the signal. Fine mapping is carried out using the lllumina BeadStation 500GX SNP genotyping platform. Alleles are genotyped using an allele-specific elongation assay that involves ligation to a locus-specific oligonucleotide. The assay is performed directly on genomic DNA at a highly multiplex level and the products are amplified using universal oligonucleotides. For each candidate region, a set of SNP markers is selected with an average inter-marker distance varying with the mean extent of LD throughout the region as determined by delta-M (I M), where M is the number of markers present in the 300 kb window centered at each reference marker is defined as the square root of the average r2 or Δ2ij measures of LD between all ({M(M-1 )}/2) pairwise comparisons of all (M) markers within the 300 kb window (Dawson et a/., 2002). This produces an average multi-marker measure of LD analogous to Hill's Δ statistic for two marker LD. Regions with a signal harbouring a high -Log10 P value and with mean delta-M of 0.3 or below are mapped with a target density of one marker per 10 kb. Regions showing a signal with a high -Log10 P value and with mean delta-M between 0.3 and 0.35 as well as selected regions with a signal with a lower -Log10 P value and a delta-M value below 0.35 are mapped with a target density of one marker per 10-20 kb. The principle is that low-LD regions will be mapped at a higher SNP density. Selected regions with a delta-M value above 0.35 are mapped with a density of one marker per 20-30 kb, including the markers used in the GWS. The selected regions are delimited by the location where the LDSTATS -Log10 P values reach the background level. The cohort consisted of 652 cases and 652 controls (these included the cases and controls used in the GWS and other cases and controls recruited following the GWS genotyping using the same criteria described in Example 1 ).
Example 5: Gene identification and characterization
Any gene or EST mapping to the interval based on public map data or proprietary map data was considered as a candidate baldness gene. The approach used to identify all genes located in the critical regions is described below.
Public gene mining
Once regions were identified using the analyses described above, a series of public data mining efforts were undertaken, with the aim of identifying all genes located within the critical intervals as well as their respective structural elements (i.e., promoters and other regulatory elements, UTRs, exons and splice sites). The initial analysis relied on annotation information stored in public databases (e.g. NCBI, UCSC Genome Bioinformatics, Entrez Human Genome Browser, OMIM - see below for database URL information).
Figure imgf000071_0001
For some genes the available public annotation was extensive, whereas for others very little was known about a gene's function. Customized analysis was therefore performed to characterize genes that corresponded to this latter class. Importantly, the presence of rare splice variants and artifactual ESTs was carefully evaluated. Subsequent cluster analysis of novel ESTs provided an indication of additional gene content in some cases. The resulting clusters were graphically displayed against the genomic sequence, providing indications of separate clusters that may contribute to the same gene, thereby facilitating development of confirmatory experiments in the laboratory. While much of this information was available in the public domain, the customized analysis performed revealed additional information not immediately apparent from the public genome browsers.
A unique consensus sequence was constructed for each splice variant and a trained reviewer assessed each alignment. This assessment included examination of all putative splice junctions for consensus splice donor/acceptor sequences, putative start codons, consensus Kozak sequences and upstream in-frame stops, and the location of polyadenylation signals. In addition, conserved noncoding sequences (CNSs) that could potentially be involved in regulatory functions were included as important information for each gene. The genomic reference and exon sequences were then archived for future reference. A master assembly that included all splice variants, exons and the genomic structure was used in subsequent analyses (i.e., analysis of polymorphisms).
An important component of these efforts was the ability to visualize and store the results of the data mining efforts. A customized version of the highly versatile genome browser GBrowse (http://www.gmod.org/) was implemented in order to permit the visualization of several types of information against the corresponding genomic sequence h addition the results of the statistical analyses were plotted against the genomic interval, thereby greatly facilitating focused analysis of gene content.
Computational Analysis of Genes and GeneMaps
In order to assist in the prioritization of candidate genes for which minimal annotation existed, a series of computational analyses were performed that included basic BLAST searches and alignments to identify related genes. In some cases this provided an indication of potential function. In addition, protein domains and motifs were identified that further assisted in the understanding of potential function, as well as predicted cellular localization. A comprehensive review of the public literature was also performed in order to facilitate identification of information regarding the potential role of candidate genes in the pathophysiology of baldness. In addition to the standard review of the literature, public resources (Medline and other online databases) were also mined for information regarding the involvement of candidate genes in specific signaling pathways. A variety of pathway and yeast two hybrid databases were mined for information regarding protein-protein interactions. These included BIND, MINT, DIP, Interdom, and Reactome, among others. By identifying homologues of genes in the baldness candidate regions and exploring whether interacting proteins had been identified already, knowledge regarding the GeneMaps for baldness was advanced. The pathway information gained from the use of these resources was also integrated with the literature review efforts, as described above.
Expression Studies
In order to determine the expression patterns for genes, relevant information was first extracted from public databases. The UniGene database, for example, contains information regarding the tissue source for ESTs and cDNAs contributing to individual clusters. This information was extracted and summarized to provide an indication in which tissues the gene was expressed. Particular emphasis was placed on annotating the tissue source for bona fide ESTs, since many ESTs mapped to Unigene clusters are artifactual. In addition, SAGE and microarray data, also curated at NCBI (Gene Expression Omnibus), provided information on expression profiles for individual genes. Particular emphasis was placed on identifying genes that were expressed in tissues known to be involved in the pathophysiology of baldness.
Polymorphism analysis
Polymorphisms identified in candidate genes, including those from the public domain as well as those identified by sequencing candidate genes and regions, are evaluated for potential function. Initially, polymorphisms are examined for potential impact upon encoded proteins. If the protein is a member of a gene family with reported 3- dimensional structural information, this information is used to predict the location of the polymorphism with respect to protein structure. This information provided insight into the potential role of polymorphisms in altering protein or ligand interactions, as well as suitability as a drug target. In a second phase of analysis we evaluate the potential role of polymorphisms in other biological phenomena, including regulation of transcription, splicing and mRNA stability, etc. There are many examples of the functional involvement of naturally occurring polymorphisms in these processes. As part of this analysis, polymorphisms located in promoter or other regulatory elements, canonical splice sites, exonic and intronic splice enhancers and repressors, conserved noncoding sequences and UTRs are localized.
In addition, some known functions and characteristics of the genes identified in the WGAS on pooled samples and confirmed in a fine-mapping study for male pattern baldness (MPB) were evaluated using the Ingenuity Pathway Analysis application (IPA, Ingenuity systems) in order to identify direct biological interactions between these genes, and also to identify molecular regulators acting on those genes (indirect interactions) that can be involved in MPB. The purpose of this effort was to decipher the molecules involved in contributing to MPB susceptibility. These gene interaction networks are very valuable tools in the sense that they facilitate extension of the map of gene products that could represent potential drug targets for MPB (see text detailed in the DETAILED DESCRIPTION OF THE INVENTION section herein).
Prophetic Example 6: SNP and Polymorphism Discovery (SNPD)
Candidate genes and regions are selected for sequencing in order to identify all polymorphisms. In cases where the critical interval, identified by fine mapping, was relatively small (-50 kb), the entire region, including all introns, is sequenced to identify polymorphisms, in situations where the region is large (>50 kb), candidate genes are prioritized for sequencing, and/or only functional gene elements (promoters, exons and splice sites) are sequenced.
The samples to be sequenced are selected according to which haplotypes contribute to the association signal observed in the region. The purpose is to select a set of samples that covered all the major haplotypes in the given region. Each major haplotype must be present in a few copies. The first step therefore consisted of determining the major haplotypes in the region to be sequenced.
Once a region is defined with the two boundary markers, all the markers used in fine mapping that are located within the region are used to determine the major haplotypes. Long haplotypes covering the whole region are thus inferred using the middle marker as an anchor The results included two series of haplotype themes that define the major haplotypes, comparing the cases and the controls This exercise is repeated using an anchor in the peripheral regions to ensure that major haplotype subsets that are not anchored at the original middle marker are not missed
Once the major haplotypes are determined as described above, appropriate genomic DNA samples are selected such that each major haplotype and haplotype subset are represented in at least two to four copies
The protocol includes the following steps, once a region is delimited
1 Primer design
The design of the primers is performed using a proprietary primer design tool A primer quality control is included in the primer design process Primers that successfully passed the control quality process were synthesized by Integrated DNA Technologies (IDT) The sense and anti-sense ohgos are separated such that the sense oligos are placed on one plate in the same position as their anti-sense counterparts are on another plate Two additional plates are created from each storage plate, one for use in PCR and the other for sequencing For PCR, the sense and anti-sense oligos of the same pair are combined in the same well to achieve a final concentration of 1 5 μM for each oligonucleotide
2 PCR OPTIMIZATION
PCR conditions are optimized by testing a variety of conditions that included varying salt concentrations and temperatures, as well as including various additives PCR products are checked for robust amplification and minimal background by agarose gel electrophoresis
3 PCR on selected samples
PCR products to be used for sequencing are amplified using the conditions chosen during optimization The PCR products are purified free of salts, dNTPs and unincorporated primers by use of a MultiScreen PCR384 filter plate manufactured by Milhpore Following PCR, the amplicons are quantified by use of a lambda/Hind III standard curve This is done to ensure that the quantity of PCR product required for sequencing had been generated. The raw data was measured against the standard curve data in Excel by use of a macro.
4. Sequencing
Sequencing of PCR products is performed by DNA Landmarks using ABI 3730 capillary sequencing instruments.
5. Sequence analysis
The ABI Prism SeqScape software (Applied Biosystems) is used for SNP identification. The chromatogram trace files were imported into a SeqScape sequencing project and the base calling is automatically performed. Sequences are then aligned and compared to each other using the SeqScape program. The base calling is checked manually, base by base; editing was performed if needed.
Prophetic Example 7: Ultra Fine Mapping (UFM)
Once polymorphisms are identified by sequencing efforts as described in Example 6, additional genotyping of all newly found polymorphisms is performed on the samples used in the fine mapping studies. Various types of genotyping assays may need to be utilized based on the type of polymorphism identified (i.e., SNP, indel, microsatellite). The assay type can be, but is not restricted to, Sentrix Assay Matrix on lllumina BeadStations, microsatellite on MegaBACE, SNP on ABI or Orchid. The frequencies of genotypes and haplotypes in cases and controls are analyzed in a similar manner as the GWS and fine mapping data. By examining all SNPs in a region, polymorphisms are identified that increase an individual's susceptibility to baldness. The goal of ultra-fine mapping is to identify the polymorphism that is most associated with disorder phenotype as part of the search for the actual DNA polymorphism that confers susceptibility to disorder. This statistical identification may need to be corroborated by functional studies.
Example 8: Confirmation of Candidate regions and genes in a general population
The confirmation of any putative associations described in Example 7 is performed in an independent general population patient sample. These DNA samples consist of at least 400 male controls and 400 male patients with baldness.
All publications, patents and patent applications mentioned in the specification and reference list are herein incorporated by reference in their entirety for all purposes. Various modifications and variations of the described method and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those skilled in molecular biology, genetics, or related fields are intended to be within the scope of the following claims.
The practice of the present invention will employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology, transgenic biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. See, for example, Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1989); DNA Cloning, Volumes I and H (D. N. Glover ed., 4 ); Oligonucleotide Synthesis (M. J. Gait ed., 1984); Mullis et al. U.S. Patent No. 4.683,195; Nucleic Acid Hybridization (B. D. Hames & S. J. Higgins eds. 1984); Transcription And Translation (B. D. Haines & S. J. Higgins eds. 1984); Culture OfAnimal Cells (R. 1. Freshney, Alan R. Liss, Inc., 1987); Immobilized Cells And Enzymes (IRL Press, 1986); B. Perbal, A Practical Guide To Molecular Cloning (1984); the treatise, Methods In Enzymology (Academic Press, Inc., N.Y.); Gene Transfer Vectors For Mammalian Cells (J. H. Miller and M. P. Calos eds., 1987, Cold Spring Harbor Laboratory); Methods In Enzymology, VoIs. 154 and 155 (Wu et al. eds.), Immunochemical Methods In Cell And Molecular Biology (Mayer and Walker, eds., Academic Press, London, 1987); Handbook Of Experimental Immunology, Volumes I-IV (D. M. Weir and C. C. Blackwell, eds., 1986); Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y., 1986).
REFERENCES
1 Austen, B. M. and Westwood, O. M. R. (1991 ). Protein Targeting and Secretion. New York, Oxford University Press. 85. p.
2 Bishop, M. J., Ed. (1998). Guide to Human Genome Computing. Toronto, Academic Press. 306p.
3 Botchkarev, V. A. and Fessing, M. Y. (2005). "Edar Signaling in the Control of Hair Follicle Development." J Investig Dermatol Svmp Proc. 10(3): 247-51.
4 Cowell, I. G. and Austin, C. A., Eds. (1997). Cdna Library Protocols. Methods in Molecular Biology. Totowa, N. J., Humana Press. 321 p.
5 Ellis, J. A. and Harrap, S. B. (2001). "The Genetics of Androgenetic Alopecia." Clin Dermatol 19(2): 149-54.
6 Freshney, R. I., Ed. (1986). Animal Cell Culture: A Practical Approach. Oxford, IRL Press. 248p.
7 Freshney, R. I. (1987). Culture of Animal Cells: A Manual of Basic Technigue. New York, A.R. Liss. 397p.
8 Fuchs, E., Merrill, B. J., Jamora, C. and DasGupta, R. (2001 ). "At the Roots of a Never-Ending Cycle." Dev Cell 1 (1 Y 13-25.
9 Glover, D. M., Ed. (1985). DNA Cloning: A Practical Approach. Practical Approach Series. Oxford, Washington, D. C, IRL Press.
10 Gribskov, M. and Devereux, J., Eds. (1994). Seguence Analysis Primer. Uwbc Biotechnical Resource Series. New York, Stockton Press. 296p.
11 Griffin, A. M. and Griffin, H. G., Eds. (1994). Computer Analysis of Seguence Data. Part 1. Methods in Molecular Biology. Totowa, N. J., Humana Press. 392p.
12 Hames, B. D. and Higgins, S. J., Eds. (1984). Transcription and Translation: A Practical Approach. Practical Approach Series. Oxford, IRL Press. 328p.
13 Hames, B. D. and Higgins, S. J., Eds. (1985). Nucleic Acid Hybridization. A Pratical Approach. Pratical Approach Series. Oxford, IRL Press. 245p.
14 Harlow, E. and Lane, D., Eds. (1988). Antibodies: A Laboratory Manual. New York, Cold Spring Harbor Laboratory. 726p.
15 Hayes, V. M., Severi, G., Eggleton, S. A., Padilla, E. J., Southey, M. C1 Sutherland, R. L., Hopper, J. L. and Giles, G. G. (2005). "The E211 G>a Androgen Receptor Polymorphism Is Associated with a Decreased Risk of Metastatic Prostate Cancer and Androgenetic Alopecia." Cancer Epidemiol Biomarkers Prev 14(4): 993-6.
16 Heijne, G. V. (1987). Sequence Analysis in Molecular Biology: Treasure Trove or Trivial Pursuit. San Diego, Academic Press. 188p.
17 Hillmer, A. M., Hanneken, S., Ritzmann, S., Becker, T., Freudenberg, J., Brockschmidt, F. F., Flaquer, A., Freudenberg-Hua, Y., Jamra, R. A., Metzen, C, Heyn, U., Schweiger, N., Betz, R. C, Blaumeiser, B., Hampe, J., Schreiber, S., Schulze, T. G., Hennies, H. C, Schumacher, J., Propping, P., Ruzicka, T., Cichon, S., Wienker, T. F., Kruse, R. and Nothen, M. M. (2005). "Genetic Variation in the Human Androgen Receptor Gene Is the Major Determinant of Common Early-Onset Androgenetic Alopecia." Am J Hum Genet 77(1 ): 140-8.
18 Hogan, B., Costantini, F. and Lacy, E., Eds. (1986). Manipulating the Mouse Embryo: A Laboratory Manual. New York, Cold Spring Harbor Laboratory. 332p.
19 Huber, B. E. and Carr, B. I., Eds. (1994). Molecular and Immunologic Approaches. Cancer Therapy in the Twenty-First Century. Mt. Kisco, New York, Futura Publishing Co. 307p.
20 Jones, J. (1992). Amino Acid and Peptide Synthesis. New York, Oxford University Press. 86p.
21 Kaufman, P. B., William, W. and Donghern, K., Eds. (1995). Handbook of Molecular and Cellular Methods in Biology and Medicine. Boca Raton, CRC Press. 484p.
22 Lesk. A. M.. Ed. (1988). Computational Molecular Biology: Sources and Methods for Sequence Analysis. New York, Oxford University Press. 254p.
23 Levy-Nissenbaum, E., Bar-Natan, M., Frydman, M. and Pras, E. (2005). "Confirmation of the Association between Male Pattern Baldness and the Androgen Receptor Gene." Eur J Dermatol 15(5): 339-40.
24 Maier, H., Ostraat, R., Gao, H., Fields, S., Shinton, S. A., Medina, K. L., Ikawa, T., Murre, C, Singh, H., Hardy, R. R. and Hagman, J. (2004). "Early B Cell Factor Cooperates with Runxi and Mediates Epigenetic Changes Associated with Mb-1 Transcription." Nat Immunol 5(10): 1069-77.
25 Male, D., Cooke, A., Owen, M., Trowsdale, J. and Champion, B., Eds. (1996). Advanced Immunology. London, Baltimore, Mosby. 273p.
26 McPherson, M. J., Ed. (1991 ). Directed Mutagenesis: A Practical Approach. Practical Approach Series. New York, IRL Press. 257p.
27 McPherson, M. J., Quirke, P. and Taylor, G. R., Eds. (1991 ). Per, a Practical Approach. Pratical Approach Series. New York, IRL Press. 253p.
28 Miller, J. H. and Calos, M. P., Eds. (1987). Gene Transfer Vectors for Mammalian Cells. Current Communications in Molecular Biology. New York, Cold Spring Harbor Laboratory. 169p.
29 O'Riordan, M. and Grosschedl, R. (1999). "Coordinate Regulation of B Cell Differentiation by the Transcription Factors Ebf and E2a." Immunity 11(1 ): 21-31.
30 Pawlowitzki, I. H., Edwards, J. H. and Thompson, E. A., Eds. (1997). Genetic Mapping of Disease Genes. San Diego, Academic Press. 288p.
31 Perbal, B. V. (1984). A Practical Guide to Molecular Cloning. New York, Wiley. 554p. 32 Peruski, L. F. and Peruski, A. H. (1997). The Internet and the New Biology. Tools for Genomic and Molecular Research. Washington, DC, ASM Press. 314p.
33 Raveh, E., Cohen, S, Levanon, D., Negreanu, V., Groner, Y. and Gat, U. (2006). "Dynamic Expression of Runxi in Skin Affects Hair Structure." Mech Dev.
34 Sambrook, J., Fritsch, E. F. and Maniatis, T. (1989). Molecular Cloning: A Laboratory Manual. New York, Cold Spring Harbor Laboratory.
35 Sell, S. (1996). Immunology, lmmunopathology and Immunity. Stamford, CT, Appleton and Lange. 1014p.
36 Smith, D. W., Ed. (1993). Biocomputing. Informatics and Genome Projects. New York, Academic Press. 336p.
37 Staal, F. J. and Clevers, H. (2000). "Tcf/Lef Transcription Factors During T-CeII Development: Unique and Overlapping Functions." Hematol J 1(1): 3-6.
38 Stenn, K. S. and Paus, R. (2001 ). "Controls of Hair Follicle Cycling." Physiol Rev 81 (1 ): 449-494.
39 Stites, D. P. and Terr, A. I., Eds. (1991 ). Basic and Clinical Immunology. Norwalk, CT, Appleton and Lange. 87Op.
40 Walker, J. M. (1998). "Protein Protocols on Cd-Rom."
41 Weir, D. M., Herzenberg, L. A., Blackwell, C. and Herzenberg, L. A., Eds. (1986). Handbook of Experimental Immunology. Oxford, England, Blackwell Scientific Publications.
42 Woodward, J., Ed. (1985). Immobilised Cells and Enzymes: A Practical Approach. Practical Approach Series. Oxford, England, IRL Press. 177p.
43 Wu, R., Ed. (1987). Methods in Enzvmology. Recombinant DNA Part F. Amsterdam, Elsevier Science. 628p.
44 Wu, R. and Grossman, L., Eds. (1987). Methods in Enzymology. Recombinant DNA Part E. Amsterdam, Elsevier Science. 576p.
45 Yan, M., Wang, L. C, Hymowitz, S. G., Schilbach, S., Lee, J., Goddard, A., de Vos, A. M., Gao, W. Q. and Dixit, V. M. (2000). "Two-Amino Acid Molecular Switch in an Epithelial Morphogen That Regulates Binding to Two Distinct Receptors." Science. 290(5491 ): 523-7.
Patents
WO 30/42661 A2 WO 1997US0005216 PCT/US94/05700 WO 0181628A1 WO 94/16101 U.S. Pat. No. 60/335,068 U.S. Pat. No. 5,869,242 U.S. Pat. No. 6,090,606 U.S. Pat. No. 5,888,819 U.S. Pat. No. 5,459,039 U.S. Pat. No. 5,498,531 U.S. Pat. No. 5,807,718 U.S. Pat. No. 4,683,195 U.S. Pat. No. 4,683,202 U.S. Pat. No. 4,797,368 U.S. Pat. No. 6,090,543 U.S. Pat No. 6,107,065 U.S. Patent No. 4,952,501. U.S. 20040009479A1 U.S. Patent No. 5,315,000 U.S. Patent No. 5,585,089 U.S. Patent No. 6,090,606 U.S. Patent No. 5,869,242 U.S. Patent No. 6,479,244 U.S. Patent No. 4,797,368 U.S. Patent No. 5,173,414

Claims

WE CLAIM:
1. A method of constructing a baldness GeneMap in a human population, comprising screening for the expression level of or presence or absence of at least one allele of at least one gene from Tables 2-4 in at least one sample.
2. A method of claim 1 , wherein said population is a general population.
3. A method of claim 1 , wherein said population is a founder population.
4. A method of claim 3, wherein said founder population is the population of Quebec.
5. A method of constructing a GeneMap in a human population, comprising screening for the presence or absence of an allele of at least one single nucleotide polymorphisms (SNPs) from Tables 5, 6.1 and 6.2 in at least one sample.
6. A method of claim 5, wherein said population is a general population.
7. A method of claim 5, wherein said population is a founder population.
8. A method of claim 7, wherein said founder population is the population of Quebec.
9. A method of genetic mapping for detecting the association of at least one marker for baldness comprising: a) obtaining biological samples from at least one patient; b) screening for the presence or absence of an allele of at least one SNP or a group of SNPs from Tables 5, 6.1 and 6.2 within each biological sample; and c) evaluating whether said SNP or a group of SNPs shows a statistically significant skewed genotype distribution between a group of patients compared to a group of control.
10. A method of claim 9, wherein said biological samples are hair, fluid, serum, tissue or buccal swabs, saliva, mucus, urine, stools, spermatozoids, vaginal secretions, lymph, amniotic fluid, pleural liquid or tears.
11. A method of claim 9, wherein said patients and controls are from a human population.
12. A method of claim 11 , wherein said population is the general population.
13. A method of claim 11 , wherein said population is a founder population.
14. A method of claim 13, wherein said founder population is the population of Quebec.
15. A method of claim 9, wherein said patients and controls are recruited independently according to specific phenotypic criteria.
16. A method of claim 9, wherein said patients and controls are recruited in the form of trios, comprising two parents and one child or one parent and two children, or in the form of unrelated individuals.
17. A method of claim 9, wherein said screening is performed be a method selected from the group consisting of an allele-specific hybridization assay, an oligonucleotide ligation assay, an allele-specific elongation/ligation assay, an allele-specific amplification assay, a single-base extension assay, a molecular inversion probe assay, an invasive cleavage assay and a selective termination assay. Other assay: RFLP, sequencing assay, SSCP, mismatch-cleaving assay, denaturing gradient gel electrophoresis.
18. A method of claim 9, wherein said screening is carried out on each individual of a cohort at each of at least one SNP or a group of SNPs from Tables 5, 6.1 and 6.2.
19. A method of claim 9, wherein said screening is carried out on pools of patients and pools of controls.
20. A method of claim 9, wherein the genotype distribution is compared one SNP at a time.
21. A method of claim 9, wherein a genotype distribution is compared with a group of markers from Tables 5, 6.1 and 6.2 forming a haplotype.
22. A method of claim 9, wherein a genotype distribution is compared using the allelic frequencies between the patient pools and control pools.
23. A set of genetic markers comprising at least two SNPs of Tables 5, 6.1 and 6.2.
24. A set of nucleic acid probes that specifically detect said SNPs of claim 23.
25. A solid support or collection of solid supports comprising the nucleic acid probes of claim 24.
26. A solid support of claim 25, wherein the support is selected from the group consisting of at least one microarray and a set of beads.
27. A method for predicting the efficacy of a drug for treating baldness in a human patient, comprising: a) obtaining a sample of cells from the patient; b) obtaining a gene expression profile from the sample in the absence and presence of in vitro modulation of the cells with specific mediators; the gene expression profile comprising one or more genes from Tables 2-4; and c) comparing the gene expression profile of the sample with a reference gene expression profile, wherein similarity between the sample expression profile and the reference expression profile predicts the efficacy of the drug for treating baldness in the patient.
28. The method of claim 27, further comprising exposing the sample to the drug for treating baldness prior to obtaining the gene expression profile of the sample.
29. The method of claim 27, wherein the sample of cells is derived from a tissue selected from the group consisting of: the scalp, hair shaft, hair bulb, hair root, hair, hair follicle, muscle, sebaceous gland, nerve, blood, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, genitalia, vessels and endothelium.
30. The method of claim 29, wherein the cells are selected from the group consisting of: melanocytes, hair follicle cells, muscle cells, nerve cells, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
31. The method of claim 27, wherein the sample is obtained via biopsy.
32. The method of claim 27, wherein the gene expression profile comprises expression values for at least two of the genes listed in Tables 2-4.
33. The method of claim 32, wherein the gene expression profile of the sample is obtained by detecting the protein products of said genes.
34. The method of claim 27, wherein the gene expression profile of the sample is obtained using a hybridization assay to oligonucleotides contained in a microarray.
35. The method of claim 34, wherein the oligonucleotides comprises nucleic acid molecules at least 95% identical to SEQ ID from Tables 2-5, 6.1 and 6.2.
36. The method of claim 27, wherein the reference expression profile is that of cells derived from patients that do not have baldness.
37. The method of claim 27, wherein the drug is selected from the group consisting of symptom relievers and drugs for a baldness condition.
38. A method for inducing a baldness-like state in a resident tissue or cell, comprising contacting the tissue or cell with at least one gene from Tables 2-4 that induces a baldness-like state.
39. The method of claim 38, wherein the resident tissue cell is selected from the group consisting of hair (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
40. A method for screening drug candidates for treating baldness, comprising: a) contacting a resident cell induced by the method of claim 38 with a drug candidate for treating baldness; and b) assaying for a baldness-like state, such that an absence of the baldness-like state is indicative of the drug candidate being effective in treating baldness.
41. A method for inducing a resident tissue cell to mimic baldness, comprising modulating the expression of at least one gene from Tables 2-4 in the cells.
42. The method of claim 41 , wherein the resident tissue cell is selected from the group consisting of hair (follicle, bulb, root, shaft and papilla), scalp, muscle cells, nervous cells, blood and vessels, dermis, epidermis and other skin cells, cutaneous surfaces, intertrigious areas, sebaceous glands, genitalia, vessels and endothelium, melanocytes, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
43. A method for screening drug candidates for treating baldness, comprising: a) contacting the resident tissue cell induced according to the method of claim 41 with a drug candidate for treating baldness; and b) assaying for a baldness-like state, such that an absence of the baldness-like state is indicative of the drug candidate being effective in treating baldness.
44. A method for treating an animal having baldness, or related phenotype, comprising administering a drug identified by the method of claim 43.
45. A drug screening assay comprising: a) administering a test compound to an animal having baldness, or related phenotype, or a cell composition isolated therefrom; and b) comparing the level of gene expression of at least one gene from Tables 2-4 in the presence of the test compound with one or both of the level of said gene expression in the absence of the test compound or in normal cells; wherein test compounds which cause the level of expression of one or more genes from Tables 2-4 to approach normal are candidates for drugs to treat baldness.
46. A method for treating an animal having baldness, or related phenotype, comprising administering a compound identified by the assay of claim 45.
47. A pharmaceutical preparation for treating an animal having baldness, or related phenotype, comprising a compound identified by the assay of claim 45 and a pharmaceutically acceptable excipient.
48. A method for identifying a gene that regulates drug response in baldness, comprising: a) obtaining a gene expression profile for at least one gene from Tables 2-4 in a resident tissue cell induced for a baldness-like state in the presence of the candidate drug; and b) comparing the expression profile of said gene to a reference expression profile for said gene in a cell induced for the baldness-like state in the absence of the candidate drug, wherein genes whose expression relative to the reference expression profile is altered by the drug may identifies the gene as a gene that regulates drug response in baldness.
49. An expression profile indicative of the presence of baldness in a patient, comprising the level of expression of at least one gene of Tables 2-4.
50. A microarray comprising probes that hybridize to one or more genes of Tables 2- 4.
51. A method of diagnosing susceptibility to baldness in an individual, comprising screening for an at-risk haplotype of at least one gene or gene region from Tables 2-4, or at least one SNP from Tables 5, 6.1 and 6.2, that is more frequently present in an individual susceptible to baldness compared to a control individual, wherein the at-risk haplotype increases risk of baldness.
52. The method of claim 51 wherein the risk increase is at least about 20%.
53. A method of diagnosing susceptibility to baldness in an individual, comprising screening for an at-risk haplotype of at least one gene from Tables 2-4, or comprising at least one SNP from Tables 5, 6.1 and 6.2, that is more frequently present in an individual susceptible to baldness, compared to the frequency of its presence in a control individual, wherein the presence of the at-risk haplotype is indicative of a susceptibility to baldness.
54. The method of claim 53 wherein the at-risk haplotype is characterized by the presence of at least one single nucleotide polymorphism from Tables 5, 6.1 and 6.2.
55. The method of claim 53 wherein screening for the presence of an at-risk haplotype in at least one gene from Tables 2-4, or comprising at least one SNP from Tables 5, 6.1 and 6.2, comprises enzymatic amplification of nucleic acid from said individual or amplification using universal oligos on elongation/ligation products.
56. The method of claim 55 wherein the nucleic acid is DNA.
57. The method of claim 55 wherein the DNA is human DNA.
58. The method of claim 53 wherein screening for the presence of an at-risk haplotype in at least one gene from Tables 2-4 or comprising at least one SNP from Tables 5, 6.1 and 6.2 comprises: a) obtaining material containing nucleic acid from the individual; (b) amplifying said nucleic acid as for claim 55; and c) determining the presence or absence of an at-risk haplotype in said amplified nucleic acid.
59. The method of claim 58 wherein determining the presence of an at-risk haplotype is performed by electrophoretic analysis.
60. The method of claim 58 wherein determining the presence of an at-risk haplotype is performed by restriction length polymorphism analysis.
61. The method of claim 58 wherein determining the presence of an at-risk haplotype is performed by sequence analysis.
62. The method of claim 58 wherein determining the presence of an at-risk haplotype is performed by hybridization analysis.
63. A kit for diagnosing susceptibility to baldness in an individual comprising: primers for nucleic acid amplification of a region of at least one gene from Tables 2-4 or a region comprising at least one SNP from Tables 5, 6.1 and 6.2.
64. The kit of claim 63 wherein the primers comprise a segment of nucleic acids of length suitable for nucleic acid amplification of a target sequence, selected from the group consisting of: single nucleotide polymorphism from Tables 5, 6.1 and 6.2, primers flanking a SNP from Tables 5, 6.1 and 6.2, and combinations thereof.
65. A method of diagnosing a susceptibility to baldness, comprising detecting an alteration in the expression or composition of a polypeptide encoded by at least one gene from Tables 2-4 in a test sample, in comparison with the expression or composition of a polypeptide encoded by said gene in a control sample, wherein the presence of an alteration in expression or composition of the polypeptide in the test sample is indicative of a susceptibility to baldness.
66. The method of claim 65, wherein the alteration in the expression or composition of a polypeptide encoded by said gene comprises expression of a splicing variant polypeptide in a test sample that differs from a splicing variant polypeptide expressed in a control sample.
67. A method of treating baldness in a patient in need thereof, comprising expressing in vivo at least one gene from Tables 2-4 (wild type/non-disorder associated allele) in an amount sufficient to treat the disorder.
68. A method of claim 67, comprising: a) administering to a patient a vector comprising a gene selected from Tables 2-4 that encodes the protein; and b) allowing said protein to be expressed from said gene in said patient in an amount sufficient to treat the disorder.
69. A method of claim 68, wherein said vector is selected from the group consisting of an adenoviral vector, and a lentiviral vector.
70. A method of claim 68, wherein said vector is administered by a route selected from the group consisting of: topical administration, intraocular administration, parenteral administration, intranasal administration, intratracheal administration, intrabronchial administration and subcutaneous administration.
71. A method of claim 68, wherein said vector is a replication-defective viral vector.
72. A method of claim 68, wherein said gene encodes a human protein.
73. A method of treating baldness in a patient in need thereof, comprising administering an agent that regulates the expression, activity or physical state of at least one gene or its encoded RNA from Tables 2-4 in the patient.
74. A method of claim 73, wherein the encoded protein from said gene comprises an alteration.
75. A method of claim 73, wherein said gene comprises an associated allele, a particular allele of a polymorphic locus, or the like that modulates the expression of the encoded protein.
76. A method of claim 73, wherein said agent is selected from the group consisting of chemical compounds, oligonucleotides, peptides and antibodies.
77. A method of claim 73, wherein said agent is an antisense molecule or interfering RNA.
78. A method of claim 73, wherein said agent is an expression modulator.
79. A method of claim 78, wherein said modulator is an activator.
80. A method of claim 78, wherein said modulator is a repressor.
81. A method of claim 73, wherein said gene comprises an associated allele, a particular allele of a polymorphic locus, or the like that modifies at least one property or function of the encoded protein.
82. A method of claim 81 , wherein the agent modulates at least one property or function of said gene or a polymorphism wherein at least one allele of said polymorphism modifies at least one property or function of the encoded protein.
83. A method for preventing the occurrence of baldness in an individual in need thereof, comprising regulating the level of at least one gene from Tables 2-4 compared to a control.
84. The method of claim 83 wherein said level is regulated by regulating expression of at least one gene from Tables 2-4 using a binding agent, a receptor to said gene, a peptidomimetic, a fusion protein, a prodrug, an antibody or a ribozyme.
85. The method of claim 83 wherein said level is controlled by genetically altering the expression level of at least one gene from Tables 2-4, whereby the regulated level of said gene mimics the level in a control individual.
86. A method for monitoring the effectiveness of treatment on the regulation of expression of one or more genes from Tables 2-4 at the RNA or protein level, or its enzymatic activity by measuring RNA, protein or enzymatic activity in a sample of peripheral blood or cells derived thereof.
87. A method of diagnosing baldness, the predisposition to baldness, or the progression of baldness, comprising the steps of a) obtaining a biological sample of mammalian body fluid or tissue to be diagnosed; b) determining the amount and/or concentration of at least one polypeptide from Tables 2-4 and/or nucleic acids encoding the polypeptide present in said biological sample; and c) comparing the amount and/or concentration of said polypeptide determine in said biological sample with the amount and/or concentration of said polypeptide as determined in a control sample and/or comparing the amount and/or concentration of nucleic acids encoding said polypeptide determined in said biological sample with the amount and/or concentration of nucleic acids encoding said polypeptides measured in a control sample, wherein the difference in the amount of said polypeptides and/or nucleic acids encoding the polypeptides is indicative of baldness or the stage of baldness.
88. A method according to claim 87, wherein a nucleic acid probe is used for determining the amount and/or concentration of at least one nucleic acid from Tables 2-4 encoding the polypeptide.
89. A method according to claim 88, wherein said nucleic acid probe is derived from the nucleic acid sequence depicted in SEQ ID NO: 1 to 4388.
90. A method according to claim 88, wherein said nucleic acid probe comprises nucleic acids hybridizing to the nucleic acid sequence depicted in SEQ ID NO: 1 to 4388, and/or fragments thereof.
91. A method according to claim 88, wherein a PCR technique is employed.
92. A method according to claim 87, wherein a specific antibody is used for determining the amount and/or concentration of at least one polypeptide from Tables 2- 4.
93. A method according to claim 92, wherein said antibody is selected from the group comprising polyclonal antiserum, polyclonal antibody, monoclonal antibody, antibody fragments, single chain antibodies and diabodies.
94. A method of claim 87, wherein at least five polypeptides or nucleic acids are determined.
95. Use of a method according to claim 87, wherein the diagnosis serves as a basis for prevention and/or monitoring of baldness.
96. A method of treatment of baldness in a mammal in need thereof, comprising the steps of a) performing steps a) to c) according to claim 87; and b) treating the mammal in need of said treatment; wherein said medical treatment is based on the stage of the disorder.
97. A method for determining the phenotype of a cell comprising detecting the differential expression, relative to a normal cell, of at least one gene from Tables 2-4.
98. The method of claim 97, wherein said difference in the level of expression of said gene, is of at least a factor of about two.
99. The method of claim 98, including the further step of cloning said genes which are up- or down-regulated.
100. The method of claim 98, including the further step of generating nucleic acid probes for detecting the level of expression of said genes which are up- or down- regulated.
101. A kit for assessing a patient's risk of having or developing baldness, comprising: a) detection means for detecting the differential expression, relative to a normal cell, of at least one gene shown in Tables 2-4 or the gene product thereof; and b) instructions for correlating the differential expression of said gene or gene product with a patient's risk of having or developing baldness.
102. The kit of claim 101, wherein the detection means includes nucleic acid probes for detecting the level of mRNA of said genes.
103. A kit for assessing a patients risk of having or developing baldness, comprising: (a) at least one means for amplifying or detecting a sequence of at least one gene in Tables 2-4, or at least one sequence comprising a SNP in Tables 5, 6.1 and 6.2, wherein the detection means includes nucleic acid probes or primers for detecting the presence or absence of an associated allele, a particular allele of a polymorphic locus, or the like or changes to at least one sequence of Tables 2-4 or Tables 5, 6.1 and 6.2, and (b) instructions for correlating the presence or absence of at least one sequence of Tables 2-4 or Table 5-7 with a patient's risk of having or developing baldness.
104. The kit of claim 101 , wherein the detection means includes an immunoassay for detecting the level of at least one gene product from Tables 2-4.
105. A method of assessing a patient's risk of having or developing baldness, comprising: a) determining the level of expression of at least one gene from Tables 2-4 or gene products thereof, and comparing the level of expression to a normal cell; and b) assessing a patient's risk of having or developing baldness, if any, by determining the correlation between the differential expression of said genes or gene products with known changes in expression of said genes measured in at least one parent suffering from baldness.
106. A nucleic acid array comprising a solid support comprising nucleic acid probes which selectively hybridize to at least 5 different genes from Tables 2-4 or at least 5 different SNPs of Tables 5, 6.1 and 6.2.
107. The array of claim 106, wherein the solid support is selected from the group consisting of paper, membranes, filters, chips, pins, and glass.
108. A method of diagnosing baldness in a patient, comprising detecting a nucleic acid molecule encoding at least one protein from Tables 2-4 in a fluid or tissue sample from the patient.
109. A method of claim 108, wherein the detecting comprises detecting at least one associated allele, particular allele of a polymorphic locus, or the like in the nucleic acid molecule encoding said protein.
110. A method of claim 109, wherein said method comprises hybridizing a probe to said patient's sample of DNA or RNA under stringent conditions which allow hybridization of said probe to nucleic acid comprising said associated allele, a particular allele of a polymorphic locus, or the like, wherein the presence of a hybridization signal indicates the presence of said associated allele, particular allele of a polymorphic locus, or the like, in at least one gene from Tables 2-4.
111. A method of claim 110, wherein the patient's DNA or RNA has been amplified and said amplified DNA or RNA is hybridized.
112. A method of claim 111 , wherein said method comprises using a single-stranded conformation polymorphism technique to assay for said associated allele, particular allele of a polymorphic locus, or the like.
113. A method of claim 111 , wherein said method comprises sequencing at least one gene from Tables 2-4 in a sample of DNA from a patient.
114. A method of claim 27, wherein said patient's sample of DNA has been amplified or cloned.
115. A method of claim 111 , wherein said method comprises sequencing at least one gene from Tables 2-4 in a sample of RNA or DNA from a patient.
116. A method of claim 111 , wherein said method comprises determining the sequence of at least one gene from Tables 2-4 by preparing cDNA from RNA taken from said patient and sequencing said cDNA to determine the presence or absence of an associated allele, a particular allele of a polymorphic locus, or the like.
117. A method of claim 110, wherein said method comprises performing an RNAse assay.
118. A method of claim 110, wherein said probe is attached to a microarray or a bead.
119. A method of claim 110, wherein said probes are oligonucleotides.
120. A method of claim 108, wherein said sample is selected from the group consisting of blood, normal tissue and tumor tissue.
121. A method of claim 109, wherein the associated allele, particular allele of a polymorphic locus, or the like is selected from the group consisting of at least one of the SNPs from Tables 5, 6.1 and 6.2, alone or in combination.
122. A method for assaying the presence of a nucleic acid associated with resistance or susceptibility to baldness in a sample, comprising: contacting said sample with a nucleic acid recited in claim 1 or claim 5 under stringent hybridization conditions; and detecting a presence of a hybridization complex.
123. A method for diagnosing baldness or the predisposition to baldness, or the progression of baldness, comprising obtaining a sample from a patient; contacting the sample with a nucleic acid of Tables 5, 6.1 and 6.2; and detecting the presence or absence of a hybridization complex, wherein the presence or absence of a hybridization complex is a diagnosis of baldness.
124. A method for assaying the presence or amount of a polypeptide encoded by a gene of Tables 2-4 for use in diagnostics, prognostics, prevention, treatment, or study of baldness, comprising: contacting a sample with an antibody of claim that specifically binds to a gene of Tables 2-4 under conditions appropriate for binding; and assessing the sample for the presence or amount of binding of the antibody to the polypeptide.
125. A method for diagnosing or prognosticating baldness comprising comparing the level of expression or activity of a polypeptide encoded by a gene of Tables 2-4 in a test sample from a patient with the level of expression or activity of the same polypeptide in a control sample wherein a difference in the level of expression or activity between the test sample and control sample is indicative of baldness.
126. A method for identifying an agent that can alter the level of activity or expression of a polypeptide encoded by a gene of Tables 2-4 for use in diagnostics, prognostics, prevention, treatment, or study of baldness, comprising: contacting a cell, cell lysate, or the polypeptide, with an agent to be tested; assessing a level of activity or expression of the polypeptide; and comparing the level of activity or expression of the polypeptide with a control sample in an absence of the agent, wherein if the level of activity or expression of the polypeptide in the presence of the agent differs by an amount that is statistically significant from the level in the absence of the agent then the agent alters the activity or expression of the polypeptide.
127. A kit for detecting whether a hair follicle is becoming dormant, said kit comprising: (a) a probe which comprises a polynucleotide sequence according to Tables 2-4, associated with baldness; and (b) a label for detecting the presence of said probe.
128. A cosmetic composition for inhibiting baldness in a patient, said cosmetic composition comprising a compound that modulates hair loss.
129. The cosmetic composition of claim 128, wherein said composition is in a form selected from the group consisting of shampoos, conditioners, lotions, sprays, ointments, oils, and gels.
130. A method for predicting the efficacy of a drug for treating baldness in a human patient, comprising: a) obtaining a sample of cells from the patient; b) obtaining a set of genotypes from the sample, wherein the set of genotypes comprises genotypes of one or more polymorphic loci from Tables 5, 6.1 and 6.2; and c) comparing the set of genotypes of the sample with a set of genotypes associated with efficacy of the drug, wherein similarity between the set of genotypes of the sample and the set of genotypes associated with efficacy of the drug predicts the efficacy of the drug for treating baldness in the patient.
131. The method of claim 130, wherein the sample of cells is derived from a tissue selected from the group consisting of: the scalp, hair shaft, hair bulb, hair root, hair, hair follicle, muscle, sebaceous gland, nerve, blood, dermis, epidermis and other skin cells, cutaneous surfaces, interthgious areas, genitalia, vessels and endothelium.
132. The method of claim 131 , wherein the cells are selected from the group consisting of: melanocytes, hair follicle cells, muscle cells, nerve cells, keratinocytes, monocytes, neutrophils, langerhans cells, CD4+ and CD8+ T cells and lymphocytes.
133. The method of claim 130, wherein the sample is obtained via biopsy.
134. The method of claim 130, wherein the set of genotypes from the sample comprises genotypes of at least two of the polymorphic loci listed in Tables 5, 6.1 and 6.2.
135. The method of claim 130 wherein the set of genotypes from the sample is obtained by hybridization to allele-specific oligonucleotides complementary to the polymorphic loci from Tables 5, 6.1 and 6.2, wherein said allele-specific oligonucleotides are contained on a microarray.
136. The method of claim 135, wherein the oligonucleotides comprise nucleic acid molecules at least 95% identical to SEQ ID from Tables 2-5, 6.1 and 6.2.
137. The method of claim 130 wherein the set of genotypes from the sample is obtained by sequencing said polymorphic loci in said sample.
138. The method of claim 130, wherein the drug is selected from the group consisting of symptom relievers and drugs for a baldness condition.
139. A method of treating baldness in a patient in need thereof, comprising administering an agent that regulates the expression, activity or physical state of at least one polypeptide encoded by a gene from Tables 2-4 in the patient.
140. A method of claim 139, wherein the encoded protein from said gene comprises an alteration, wherein said alteration is encoded by a polymorphic locus in said gene.
141. A method of claim 139, wherein said gene comprises an associated allele, a particular allele of a polymorphic locus, or the like that modulates the expression of the encoded protein.
142. A method of claim 139, wherein said agent is selected from the group consisting of chemical compounds, oligonucleotides, peptides and antibodies.
143. A method of claim 139, wherein said agent is an antisense molecule or interfering RNA.
144. A method of claim 139, wherein said agent is an expression modulator.
145. A method of claim 144, wherein said modulator is an activator.
146. A method of claim 144, wherein said modulator is a repressor.
147. A method of claim 139, wherein said gene comprises an associated allele, a particular allele of a polymorphic locus, or the like that modifies at least one property or function of the encoded protein.
148. A kit for assessing a patient's risk of having or developing baldness, comprising: a) detection means for detecting the genotype of at least one polymorphic locus shown in Tables 5, 6.1 and 6.2; and b) instructions for correlating the genotype of said at least one polymorphic locus with a patient's risk of having or developing baldness.
149. The kit of claim 148, wherein the detection means includes nucleic acid probes for detecting the genotype of said at least one polymorphic locus.
150. A method of assessing a patient's risk of having or developing baldness, comprising: a) selecting at least one polymorphic locus from Tables 5, 6.1 and 6.2; b) determining a genotype for said at least one polymorphic locus from Tables 5, 6.1 and 6.2 in a patient; c) comparing said genotype of b) to a genotype for said at least one polymorphic locus from Tables 5, 6.1 and 6.2 that is associated with baldness; and d) assessing the patient's risk of having or developing baldness, wherein said patient has a higher risk of having or developing baldness if the genotype for said at least one polymorphic locus from Tables 5, 6.1 and 6.2 in said patient is the same as said genotype for said at least one polymorphic locus from Tables 5, 6.1 and 6.2 that is associated with baldness.
PCT/US2007/083147 2006-10-31 2007-10-31 Genemap of the human genes associated with male pattern baldness WO2008055196A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US85541506P 2006-10-31 2006-10-31
US60/855,415 2006-10-31

Publications (3)

Publication Number Publication Date
WO2008055196A2 WO2008055196A2 (en) 2008-05-08
WO2008055196A9 true WO2008055196A9 (en) 2008-06-26
WO2008055196A3 WO2008055196A3 (en) 2008-12-24

Family

ID=39345069

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/083147 WO2008055196A2 (en) 2006-10-31 2007-10-31 Genemap of the human genes associated with male pattern baldness

Country Status (1)

Country Link
WO (1) WO2008055196A2 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2572201B1 (en) 2010-05-17 2015-08-26 The Procter and Gamble Company Methods of detecting and demonstrating hair damage via evaluation of protein fragments
KR102464776B1 (en) * 2020-09-11 2022-11-09 서울대학교산학협력단 Genetic polymorphic markers associated with female pattern hair loss and uses thereof
EP4269609A1 (en) * 2022-04-28 2023-11-01 Eberhard Karls Universität Tübingen Medizinische Fakultät Method for making a genetic determination based on a hair root sample

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6582908B2 (en) * 1990-12-06 2003-06-24 Affymetrix, Inc. Oligonucleotides
WO2005121374A2 (en) * 2004-06-07 2005-12-22 Oklahoma Medical Research Foundation Molecular analysis of hair follicles for disease

Also Published As

Publication number Publication date
WO2008055196A3 (en) 2008-12-24
WO2008055196A2 (en) 2008-05-08

Similar Documents

Publication Publication Date Title
US20090305900A1 (en) Genemap of the human genes associated with longevity
US20100291551A1 (en) Genemap of the human associated with crohn&#39;s disease
US20100120628A1 (en) Genemap of the human genes associated with adhd
US20100144538A1 (en) Genemap of the human genes associated with schizophrenia
WO2008024114A1 (en) Genemap of the human genes associated with schizophrenia
US20100120627A1 (en) Genemap of the human genes associated with psoriasis
US20030211486A1 (en) Compositions and methods for detecting polymorphisms associated with pigmentation
WO2009026116A2 (en) Genemap of the human genes associated with longevity
US8097415B2 (en) Methods for identifying an individual at increased risk of developing coronary artery disease
WO2008085601A2 (en) Genemap of the human genes associated with asthma disease
WO2009039244A2 (en) Genemap of the human genes associated with crohn&#39;s disease
EP1203827B1 (en) Polymorphisms in the human KDR gene
EP1130123A2 (en) Diagnostic method
EP2118321A2 (en) Genemap of the human genes associated with endometriosis
WO2008055196A9 (en) Genemap of the human genes associated with male pattern baldness
WO2009139648A2 (en) Methods and compositions for assessment of pulmonary function and disorders
CA2486789A1 (en) Compositions and methods for inferring a response to a statin
EP1112381A1 (en) Polymorphisms in the human alpha4 integrin subunit gene, suitable for diagnosis and treatment of integrin ligand mediated diseases
WO2000006768A1 (en) Genetic polymorphisms in the human neurokinin 1 receptor gene and their uses in diagnosis and treatment of diseases
US20020160362A1 (en) Diagnostic method
WO2006136791A1 (en) Polymorphisms and haplotypes in p2x7 gene and their use in determining susceptibility for atherosclerosis-mediated diseases

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: 07844758

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07844758

Country of ref document: EP

Kind code of ref document: A2