CORNEODESMOSIN BASED TEST AND MODEL FOR INFLAMMATORY DISEASE

The present invention relates to a polynucleotide encoding the corneodesmosin protein having one or more nucleotide insertions, deletions, or substitutions at one or more novel positions. The invention also relates to the corneodesmosin protein having one or more amino acid insertions, deletions, and substitutions. These nucleotide and amino acid polymorphisms are useful in diagnosing or determining susceptibility to corneodesmosin-mediated disease, such as inflammatory diseases, including psoriasis, and in treating such disease. Host cells and transgenic non-human animals comprising polynucleotides or proteins of the invention are provided. Methods of screening for agents for use in treating corneodesmosin-mediated disease are also provided.

Latest YORK PHARMA (R & D) LIMITED Patents:

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This is a divisional of U.S. application Ser. No. 10/204,884, filed Jan. 29, 2003, which is the National Stage of International Application No. PCT/GB01/00795, filed Feb. 23, 2001, which claims priority from GB 0004312.5, filed Feb. 23, 2000.

FIELD OF THE INVENTION

The present invention relates to nucleotide substitutions, deletions, or insertions in the corneodesmosin gene, and the exploitation of these polymorphisms in the detection and/or treatment of corneodesmosin-mediated disease, such as inflammatory diseases, including psoriasis. The present invention also relates to polynucleotides encoding the corneodesmosin protein having one or more nucleotide polymorphisms, and to proteins encoded by said polynucleotides. Also provided are transgenic non-human animals comprising the polynucleotides of the present invention, and methods and kits for treating, diagnosing, or determining susceptibility to corneodesmosin-mediated disease, in particular by way of gene therapy.

BACKGROUND OF THE INVENTION

In recent years, it has been recognized that there is considerable genetic diversity in human populations, with common polymorphisms occurring on average at least every kilobase in the genome. Polymorphisms which affect gene expression or activity of the encoded gene product may account for susceptibility to, or expression of, disease conditions, either directly or through interaction with other genetic and environmental factors.

Understanding the molecular basis for disease, by sequencing the human genome and characterizing polymorphisms, will enable the identification of those individuals at greatest risk of disease. This will allow the better matching of treatment and disease, and enable the production of new and improved targets for drugs. Screening and treatment of disease may also be better targeted to those in need, thus increasing the cost-effectiveness of health-care provision.

One area in need of such approaches is the diagnosis and treatment of inflammatory diseases. Inflammation, which can be broadly defined as the destructive sequelae to activation of elements of the body's immune system, is a feature of many diseases including infection, autoimmune disorders, and benign and malignant hyperplasia. The identification of genetic factors which influence susceptibility to such disorders will provide important new insights into inflammatory disease, and may yield important new diagnostic and/or prognostic tests and treatments.

Psoriasis is a chronic inflammatory cutaneous disorder which affects approximately 2% of the population in the UK and US. Psoriasis manifests itself as red scaly skin patches, principally on the scalp, elbows and knees, and is caused by epidermal hyperproliferation, and abnormal differentiation and infiltration of inflammatory cells. Psoriasis may also be associated with other inflammatory diseases such as arthritis, Crohn's disease, and HIV infection. Population, family, and twin studies all suggest an important genetic component in the pathogenesis of psoriasis, coupled with environmental triggers such as streptococcal infection and stress.

Psoriasis is one of a number of autoimmune diseases that display significant human leukocyte antigen (HLA) associations. The analysis of population-specific HLA haplotypes has provided evidence that susceptibility to psoriasis is linked to the class I and II major histocompatibility complexes (MHC) on human chromosome 6 (Jenisch et al. (1998) Am. J. Hum. Genet 63:191-199). These studies show that psoriasis consists of two distinct disease subtypes (Type I and Type II), which differ in age of onset and in the frequency of HLA types. Type I psoriasis has an age of onset of prior to 40 years and HLA types Cw6, B57, and DR7 are strongly increased. Patients with Type I psoriasis are much more likely to have a positive family history for the disease. In contrast, only about 10% of Cw6-positive individuals develop Type II psoriasis disease, with HLA-Cw2 being over-represented in this group.

Linkage analysis and association studies suggest the presence of a major genetic determinant of psoriasis within the MHC, the strongest candidate gene marker being HLA-C. The most significant association has been shown between HLA-Cw6 and disease Type IA, which has the earliest onset of disease at 0 to 20 years. However, specific involvement of the HLA-Cw6 genotype in disease pathogenesis has yet to be established.

Recently, attention has focused on non-HLA genes close to HLA-C, in particular the corneodesmosin gene (also known as the S gene), which is located approximately 160 kb telomeric of the HLA-C locus. The corneodesmosin gene consists of 2 exons spanning approximately 5.3 kb of genomic DNA sequence. Two corneodesmosin mRNAs of 2.2 kb and 2.6 kb, resulting from alternative splicing, have been described (Guerrin et al. (1998) J. Biol. Chem. 273:22640-22647). Association studies (Ahnini et al. (1999) Hum. Mol. Genet. 8:1135-1140) suggest a strong, significant association between a polymorphism at position 1243 of the corneodesmosin gene and psoriasis. A corneodesmosin gene haplotype was subsequently defined, which by TDT analysis was shown to have a strong, significant association with psoriasis (Allen et al. (1999) Lancet 353:1589-90).

In human epidermis and other cornified squamous epithelia, corneodesmosin is located in the desmosomes of the upper living layers, and in related structures of the cornified layers, the corneodesmosomes. During maturation of the cornified layers, the protein undergoes a series of cleavages, thought to be a prerequisite of desquamation (shedding of the cuticle or epidermis). Corneodesmosin is detected as a glycosylated and phosphorylated basic protein with an apparent molecular mass of 52-56 kDa. During stratum corneum maturation, corneodesmosin is progressively proteolysed until desquamation occurs. In superficial corneocytes, the 52-56 kDa form is no longer detected and immunoreactive fragments of 45 to 30 kDa predominate. Since location, biochemical characteristics and processing of corneodesmosin are similar in several mammals, it is likely that the protein is essential for the function of corneodesmosomes and corneocyte cohesion. It has been shown that expression of the 56 kDa epidermal keratin polypeptide is increased in psoriatic lesions compared with normal skin and transformation of desmosomes into corneodesmosomes is altered in psoriatic epidermis.

Psoriasis affects approximately 6.4 million people in the US and causes varying ranges of physical discomfort, pain and disability. At present, the causes of psoriasis are unknown. There is no specific test for psoriasis or susceptibility thereto, and diagnosis is based solely on clinical examination and skin histopathology.

It is likely that corneodesmosin is implicated in a range of skin diseases, including psoriasis. In this text, diseases in which corneodesmosin is implicated in the pathology will be referred to as “corneodesmosin-mediated disease.”

The present invention aims to overcome or ameliorate previous limitations in the art by providing means and methods for the detection and treatment of individuals having, or being susceptible to, corneodesmosin-mediated disease, in particular inflammatory conditions such as psoriasis.

In a first aspect, the present invention provides an isolated or recombinant polynucleotide comprising a nucleic acid sequence encoding the corneodesmosin gene of FIG. 1 (SEQ ID NO:1), wherein said nucleic acid sequence comprises a nucleotide substitution, deletion or insertion at one or more of positions 6984, 7068, 7077, 7107, 7164, 8884, 8906, 8931, 9538, 9607, 9608, 9647, 9667, 9745, 9761, 9926, 9952, 9968, 10082, 10161, 10162, 10363, 11567, 11641, 11649, 11808, 11839, 11885, 11977, 12018, 12136, 12149; 12198, 12283, 12318, 12345, 12373, 12901, 13001, 13020, 13108, 13117, 13178, 13224, 13316, 13365, 13562, 13605, 13670, 13859, 13889 and 13914 of FIG. 1 (corresponding to positions 284, 368, 377, 407, 464, 2184, 2206, 2231, 2838, 2907, 2908, 2947, 2967, 3045, 3061, 3226, 3252, 3268, 3382, 3461, 3462, 3663, 4867, 4941, 4949, 5108, 5139, 5185, 5277, 5318, 5436, 5449, 5498, 5583, 5618, 5645, 5673, 6201, 6301, 6320, 6408, 6417, 6478, 6524, 6616, 6665, 6862, 6905, 6970, 7159, 7189 and 7214 of SEQ ID NO:1). These novel polymorphisms in the corneodesmosin gene, at the positions indicated above, may be responsible for corneodesmosin-mediated disease. In particular, the polymorphisms of the present invention may be useful in identifying individuals susceptible or resistant to corneodesmosin-mediated disease, and in the diagnosis or treatment of such conditions. Preferred combinations of the polymorphisms of the invention are the haplotypes shown in Tables 10a and b. The most preferred haplotype is B of Table 10a.

The polynucleotide of this invention is preferably DNA, or may be RNA or other options.

By “isolated” is meant a polynucleotide sequence which has been purified to a level sufficient to allow allelic discrimination. For example, an isolated sequence will be substantially free of any other DNA or protein product. Such isolated sequences may be obtained by PCR amplification, cloning techniques, or synthesis on a synthesizer. By recombinant is meant polynucleotides which have been recombined by the hand of man.

The corneodesmosin gene sequence shown in FIG. 1 (SEQ ID NO:1) refers to the genomic clone of corneodesmosin, detailed in GenBank Accession No. AC006163 (a genomic clone of the MHC region on chromosome 6p21.3). The single nucleotide polymorphisms of the invention are shown in bold type and underlined on this figure, and have each been given a positional reference with respect to this figure. For reference and comparison with prior art publications, the positional references with respect to the coding sequence have also been given in Table 6, Column 2, where nucleotide position 1 corresponds to the first nucleotide of exon 1 and nucleotides upstream of this are given a negative prefix.

A polymorphism is typically defined as two or more alternative sequences, or alleles, of a gene in a population. A polymorphic site is the location in the gene at which divergence in sequence occurs. Examples of the ways in which polymorphisms are manifested include restriction fragment length polymorphisms, variable number of tandem repeats, hypervariable regions, minisatellites, di- or multi-nucleotide repeats, insertion elements and nucleotide deletions, additions or substitutions. The first identified allele is usually referred to as the reference allele, or the wild type. Additional alleles are usually designated alternative or variant alleles. Herein, the sequence exactly as shown in FIG. 1 is designated the reference sequence, and is not part of the invention. Nucleic acid sequences of the present invention which differ from the sequence of FIG. 1 (SEQ ID NO:1) at one or more of the positions indicated above may be referred to as variants of FIG. 1 (SEQ ID NO:1).

A single nucleotide polymorphism is a variation in sequence between alleles at a site occupied by a single nucleotide residue. Single nucleotide polymorphisms (SNP's) arise from the substitution, deletion or insertion of a nucleotide residue at a polymorphic site. Typically, this results in the site of the variant sequence being occupied by any base other than the reference base. For example, where the reference sequence contains a “T” base at a polymorphic site, a variant may contain a “C,” “G” or “A” at that site. Single nucleotide polymorphisms may result in corresponding changes to the amino acid sequence. For example, substitution of a nucleotide residue may change the codon, resulting in an amino acid change. Similarly, the deletion or insertion of three consecutive bases in the nucleic acid sequence may result in the insertion or deletion of an amino acid residue. For ease of reference, where a single nucleotide polymorphism of the present invention results in the insertion or deletion of a nucleotide or amino acid residue, the numbering system of FIGS. 1 (SEQ ID NO:1) and 2 (SEQ ID NO:2) have been maintained.

The single nucleotide polymorphisms of the present invention which occur within the protein coding sequence may contribute to the phenotype of an organism by affecting protein structure or function. The effect may be neutral, beneficial or detrimental, depending upon the circumstances. Whatever the effect, the identification of such polymorphisms enables for the first time determination of susceptibility to disease, and new methods of treatment. The single nucleotide, polymorphisms of the invention which occur in the non-coding 5′ or 3′ untranslated regions, may not affect protein sequence, but may exert phenotypic effects by influencing replication, transcription and/or translation. A polymorphism may affect more than one phenotypic trait or may be related to a specific phenotype. In the present invention, polymorphisms in the corneodesmosin gene are likely to affect the phenotype of an individual with respect to corneodesmosin-mediated disease, such as inflammatory disease, in particular psoriasis.

The single nucleotide polymorphisms of the corneodesmosin gene, including those of the present invention, are listed in Table 6 where:

    • Column 1 designates each single nucleotide polymorphism a reference number.
    • Column 2 provides the positional reference of the polymorphism with respect to FIG. 1.
    • Column 3 indicates position of the SNP with respect to the corneodesmosin coding sequence.
    • Column 4 shows the location of the polymorphisms in the gene.
    • Column 5 shows the sequence flanking the polymorphism, the polymorphism itself being shown in bold type. For example, the polymorphism at position 6984 is shown as C/T, meaning that the variant sequence comprises a T residue, rather than the native C residue.
    • Column 6 denotes the standard IUB code.
    • Column 7 denotes the SEQ ID NO of the corresponding flanking sequence.

As discussed above, where a single nucleotide polymorphism of the present invention comprises a nucleotide substitution, the substitution may comprise the replacement of the reference base at a polymorphic site with any other base. Each single nucleotide polymorphism described in Table 6, Column 4, represents a preferred embodiment of the invention.

It will be appreciated by those skilled in the art that corneodesmosin gene sequences of the invention may comprise one or more nucleotide substitutions, deletions or insertions in addition to one or more of the single nucleotide polymorphisms of the invention.

In a second aspect, fragments of the above polynucleotides are provided, which comprise one or more nucleotide substitutions, insertions or deletions at one or more of the above mentioned positions of FIG. 1 (SEQ ID NO:1). Preferably, a fragment may comprise, or even consist of, the polynucleotide sequence of Table 6, Column 4. The novelty of a fragment according to the present embodiment. may be easily ascertained by comparing the nucleotide sequence of a fragment with sequences catalogued in databases such as GenBank, or by using computer programs such as DNASIS (Hitachi Engineering, Inc.) or Word Search or FAST A of the Genetic Computer Group (Madison, Wis.).

Preferably, the fragments do not encode a full length protein, as is generally the case with the aforementioned polynucleotides, but otherwise satisfy the requirements of the first aspect. Preferred fragments may be 10 to 150 nucleotides in length. More preferably, the fragments are between 5 to 10, 5 to 20, 10 to 20, 20 to 50, or 50 to 100 nucleotides in length. For example, the fragments may be 5, 8, 10, 12, 15, 18, 20, 22, 25, 28, 30, or 35 nucleotides in length. The fragments may be useful in a variety of diagnostic, prognostic or therapeutic methods, or may be useful as research tools for example in drug screening.

In a third aspect of the invention, there is provided non-coding, complementary sequences which hybridize to the corneodesmosin gene sequence. Such “anti-sense” sequences are useful as probes or primers for detecting an allele of a polymorphism of the invention, or in the regulation of the corneodesmosin gene. They may also be used as agents for use in the identification and/or treatment of individuals having or being susceptible to corneodesmosin-mediated disease.

The anti-sense sequences of the invention include those which hybridize to an allele of a polymorphism of the invention, and also those which hybridize a region flanking the polymorphic site to enable amplification of an allele of one or more polymorphisms. These sequences may be useful as probes or primers. To be useful as a probe, the antisense sequence should bind preferentially one allele of one or more polymorphisms of the present invention and will, preferably, comprise the exact complement of one allele of one or more polymorphisms of the invention. Thus, for example, where the variant comprises a “G” residue at position 7068 of FIG. 1 (corresponding to position 368 of SEQ ID NO:1), it is preferred that the anti-sense sequence will comprise a “C” residue. Such anti-sense sequences which are capable of specific hybridization to detect a single base mismatch may be designed according to methods known in the art and described in Maniatis et al. (1989) Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, NY, and Berger et al. (eds.) (1987) Methods in Enzymology, Vol. 152, Guide to Molecular Cloning Techniques, Academic Press Inc., San Diego, Calif.; Gibbs et al. (1989) Nuc. Acids Res. 17:2437; Kwok et al. (1990) Nuc. Acids Res. 18:999; and Miyada et al. (1987) Methods Enzymol. 154:94. Variation in the sequence of these anti-sense sequences is acceptable for the purposes of the present invention, provided that the ability of the anti-sense sequence to distinguish between alleles of a polymorphism is not compromised. Similarly, variation in the sequence of a primer sequence is acceptable, provided its ability to mediate amplification of a polymorphic site is not compromised. Preferably, a primer sequence will hybridize to the corneodesmosin gene under stringent conditions which are defined below.

In relation to the present invention, “stringent conditions” refers to the washing conditions used in a hybridization protocol. In general, the washing conditions should be a combination of temperature and salt concentration so that the denaturation temperature is approximately 5 to 20° C. below the calculated Tm of the nucleic acid under study. The Tm of a nucleic acid probe of 20 bases or less is calculated under standard conditions (1M NaCl) as [4° C.×(G+C)+2° C.×(A+T)], according to Wallace rules for short oligonucleotides. For longer DNA fragments, the nearest neighbor method, which combines solid thermodynamics and experimental data may be used, according to the principles set out in Breslauer et al. (1986) PNAS 83:3746-3750. The optimum salt and temperature conditions for hybridization may be readily determined in preliminary experiments in which DNA samples immobilized on filters are hybridized to the probe of interest and then washed under conditions of different stringencies. While the conditions for PCR may differ from the standard conditions, the Tm may be used as a guide for the expected relative stability of the primers. For short primers of approximately 14 nucleotides, low annealing temperatures of around 44° C. to 50° C. are used. The temperature may be higher, depending upon the base composition of the primer sequence used.

The anti-sense polynucleotides of this embodiment may be the full length of the corneodesmosin gene of FIG. 1 (SEQ ID NO:1), or more preferably may be 5 to 200 nucleotides in length. Preferred polynucleotides are 5 to 10, 10 to 20, 20 to 50, 50 to 100 or 100 to 200 nucleotides in length. Primers, in particular, are typically 10 to 15 nucleotides long, and may occasionally be 16 to 25.

In a preferred embodiment, the polynucleotides of the aforementioned aspects of the invention may be in the form of a vector, to enable the in vitro or in vivo expression of the polynucleotide sequence. The polynucleotides may be operably linked to one or more regulatory elements including a promoter; regions upstream or downstream of a promoter such as enhancers which regulate the activity of the promoter; an origin of replication; appropriate restriction sites to enable cloning of inserts adjacent to the polynucleotide sequence; markers, for example antibiotic resistance genes; ribosome binding sites; RNA splice sites and transcription termination regions; polymerization sites; or any other element which may facilitate the cloning and/or expression of the polynucleotide sequence. Where two or more polynucleotides of the invention are introduced into the same vector, each may be controlled by its own regulatory sequences, or all sequences may be controlled by the same regulatory sequences. In the same manner, each sequence may comprise a 3′ polyadenylation site. The vectors may be introduced into microbial, yeast or animal DNA, either chromosomal or mitochondrial, or may exist independently as plasmids. Examples of suitable vectors will be known to persons skilled in the art and include pBluescript II, LambdaZap, and pCMV-Script (Stratagene Cloning Systems, La Jolla (USA)) Appropriate regulatory elements, in particular, promoters will usually depend upon the host cell into which the expression vector is to be inserted. Where microbial host cells are used, promoters such as the lactose promoter system, tryptophan (Trp) promoter system, β-lactamase promoter system or phage lambda promoter system are suitable. Where yeast cells are used, preferred promoters include alcohol dehydrogenase I or glycolytic promoters. In mammalian host cells, preferred promoters are those derived from immunoglobulin genes, SV40, Adenovirus, Bovine Papilloma virus, etc. Suitable promoters for use in various host cells would be readily apparent to a person skilled in the art (see, for example, Ausubel et al. (eds.), Current Protocols in Molecular Biology, published by Wiley).

In a fourth aspect of the present invention there is provided a protein or protein fragment comprising an amino acid substitution, deletion or insertion at one or more of positions 18, 130 or 180 of the amino acid sequence of FIG. 2 (SEQ ID NO:2). Preferably, the protein or protein fragment is encoded by a polynucleotide according to the first aspect of the invention, and comprises a nucleotide insertion, deletion or substitution at one or more of positions 7164, 10082, 10161, 10162 and 10363 of FIG. 1 (corresponding to positions 464, 3382, 3461, 3462 and 3663 of SEQ ID NO:1, respectively). The corneodesmosin protein or protein fragments of the invention may comprise one or more polymorphisms in addition to one or more of the above-mentioned polymorphisms of FIG. 2.

The amino acid sequence exactly as shown in FIG. 2 (SEQ ID NO:2) may be referred to as the reference sequence, and is not part of the invention. The amino acid sequence of FIG. 2 (SEQ ID NO:2) having an amino acid substitution, deletion or insertion at one or more of the positions indicated above may be referred to as a variant of FIG. 2 (SEQ ID NO:2). The reference amino acid at one or more of the above polymorphic sites may be replaced by any other amino acid residue to produce a variant sequence. Amino acid sequences of FIG. 2 (SEQ ID NO:2) having one or more of the polymorphisms disclosed in Table 4 are each preferred embodiments of the invention.

Protein fragments may be functional or non-functional and may be useful in drag screening or gene therapy. Functional fragments may be defined as those which have characteristics of the corneodesmosin protein. The fragments may be at least 10, preferably at least 15, 20, 25 30, 35, 40 or 50 amino acids in length.

In a fifth aspect of the present invention, there are provided antibodies which react with an antigen of a protein or protein fragment of the fourth aspect. Antibodies can be made by the procedure set forth by standard procedures (Harlow and Lane (1988) Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY). Briefly, purified antigen can be injected into an animal in an amount and in intervals sufficient to elicit an immune response. Antibodies can either be purified directly, or spleen cells can be obtained from the animal. The cells are then fused with an immortal cell line and screened for antibody secretion. The antibodies can be used to screen DNA clone libraries for cells secreting the antigen. Those positive clones can then be sequenced as described in, for example, Kelly et al. (1992) Bio/Technology 10:163-167 and Bebbington et al. (1992) Bio/Technology 10:169-175. Preferably, the antigen being detected and/or used to generate a particular antibody will include proteins or protein fragments according to the fourth aspect.

In a sixth aspect of the present invention, there is provided host cell comprising a polynucleotide according to any of the aforementioned aspects, for expression of the polynucleotide. The host cell may comprise an expression vector, or naked DNA encoding said polynucleotides. A wide variety of suitable host cells are available, both eukaryotic and prokaryotic. Examples include bacteria such as E. coli, yeast, filamentous fungi, insect cells, mammalian cells, preferably immortalized, such as mouse, CHO, HeLa, myeloma or Jurkat cell lines, human and monkey cell lines and derivatives thereof. Such host cells are useful in drug screening systems to identify agents for use in diagnosis or treatment of individuals having, or being susceptible to corneodesmosin-mediated disease.

The method by which said polynucleotides are introduced into a host cell will usually depend upon the nature of both the vector/DNA and the target cell, and will include those known to a person skilled in the art. Suitable known methods include fusion, conjugation, transfection, transduction, electroporation or injection, as described in Sambrook et al.

In a seventh aspect of the present invention, there is provided a transgenic non-human animal comprising a polynucleotide according to an aforementioned aspect of the invention. Preferably, the transgenic, non-human animal comprises a polynucleotide according to the first or second aspects. The transgenic animal may be either homozygous or heterozygous for the variant sequence. The animal, and cells derived therefrom, are useful for screening biologically active agents that may modulate corneodesmosin function. Such screening methods are of particular use for determining the specificity and action of potential therapies for corneodesmosin-mediated disease, such as psoriasis. The animals are useful as a model to investigate the role of corneodesmosin in normal skin function. Transgenic non-human animals are also useful for the analysis of the single nucleotide polymorphisms and their phenotypic effect.

Expression of a polynucleotide of the invention in a transgenic non-human animal is usually achieved by operably linking the polynucleotide to a promoter and/or enhancer sequence, preferably to produce a vector of the fourth aspect, and introducing this into an embryonic stem cell of a host animal by microinjection techniques (Hogan et al. (1986), Manipulating the Mouse Embryo: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY, and Capecchi (1989) Science 244:1288-1292). Preferably, the construct to be introduced into the animal additionally comprises (a) a first homology region with substantial identity to a first corneodesmosin gene sequence; and (b) a second homology with substantial identity to a second corneodesmosin gene sequence. The first and second homology regions are of sufficient length for homologous recombination to occur with an endogenous corneodesmosin gene. Those embryonic stem cells comprising the desired polynucleotide sequence may be selected, usually by monitoring expression of a marker gene, and used to generate a non-human transgenic animal. Preferred host animals include mice and other rodents. Further development of such an embryonic stem cell may produce a transgenic animal having cells that are descendant from the embryonic stem cell and thus carry the variant sequence in their genome. Such animals can then be selected and bred to produce animals having the variant sequence in all somatic and germ cells. Such mice can then be bred to homozygosity.

In a preferred embodiment, the transgenic non-human animal may comprise an anti sense nucleic acid sequence of the third aspect. The expression of an anti-sense sequence in a transgenic non-human animal may be useful in determining the effects of such sequences in treating corneodesmosin-mediated disease, or in neutralizing deleterious effects of variant corneodesmosin genes in an animal. Preferably, the host animal will be one which suffers from corneodesmosin-mediated disease. The disease may be naturally occurring or artificially introduced.

In some preferred embodiments, for example where the mediated disease has been artificially induced, the transgenic non-human animal will be modulated to no longer expresses the endogenous corneodesmosin gene. Such animals may be referred to as “knock out.” In some cases, it may be appropriate to modulate the expression of the endogenous corneodesmosin gene, or express the polynucleotides of the present invention, in specific tissues. This approach removes viability problems if the expression of a gene is abolished or induced in all tissues. Preferably, the specific tissue would be skin. Where the heterologous gene is human, the animal may be useful in identifying agents which inhibit expression or activity of the variant corneodesmosin sequences of the invention, either in vivo or in vitro.

In an eighth aspect of the present invention there is provided a method of screening for agents for use in the prognosis, diagnosis or treatment of individuals having, or being susceptible to, corneodesmosin-mediated disease, said method comprising contacting a putative agent with a polynucleotide or protein according to an aforementioned aspect of the present invention, and monitoring the reaction there between. Preferably, the method further comprises contacting a putative agent with a reference polynucleotide or protein of FIG. 1 or 2 (SEQ ID NO:1 or SEQ ID NO:2, respectively), and comparing the reaction between (i) the agent and the polynucleotide or protein encoding the reference allele; and (ii) the agent and polynucleotide or protein of the invention. Potential agents are those which react differently with a variant of the invention and a reference allele. It is envisaged that the present method may be carried out by contacting a putative agent with a host cell or transgenic non-human animal comprising a polynucleotide or proteu1 according to the invention. Putative agents will include those known to persons skilled in the art, and include chemical or biological compounds, such as anti-sense polynucleotide sequences, complementary to the coding sequences of the first aspect, or polyclonal or monoclonal antibodies which bind to a product such as a protein or protein fragment of the second aspect. The agents identified in the present method may be useful in determining susceptibility to corneodesmosin-mediated disease, or in the diagnosis, prognosis or treatment of said disease.

In a ninth aspect of the present invention, there is provided a method of diagnosing, or determining susceptibility of a subject to corneodesmosin-mediated disease, said method comprising determining which allele of one or more of the polymorphisms of the invention is present in a subject. The above method maybe used in diagnosing or determining susceptibility of a subject to any disease in which corneodesmosin is implicated in the pathology, in particular inflammatory disease, such as psoriasis. The method of the ninth aspect may also be used to identify the presence of a combination of single nucleotide polymorphisms in a subject which define a haplotype linked to corneodesmosin-mediated disease. The haplotype may be any particular combination of the above single nucleotide polymorphisms, optionally including known polymorphisms. Preferred haplotypes are those shown in Table 10a, the most preferred haplotype being B of Table 10a.

Any method, including those known to persons skilled in the art, may be used to determine which allele of one or more polymorphisms of the invention is present. Preferably, the method comprises first removing a sample from a subject. More preferably, the method comprises isolating from a sample a polynucleotide or protein to determine therein which allele of one or more polymorphisms of the invention is present.

Any biological sample comprising cells containing nucleic acid or protein is suitable for this purpose. Examples of suitable samples include whole blood, semen, saliva, tears, buccal, skin or hair. For analysis of cDNA, mRNA or protein, the sample must come from a tissue in which the corneodesmosin gene is expressed, and thus it is preferable to use skin samples.

In a preferred embodiment, the method for diagnosing, or determining susceptibility of a subject to a corneodesmosin-mediated disease, comprises determining which allele of one or more polymorphisms of the invention is present, in a polynucleotide. Any method for determining alleles in a polynucleotide may be used, including those known to persons skilled in the art. Preferably, the method may comprise the use of anti-sense polynucleotides, as defined above. Such polynucleotides may include sequences which are able to distinguish between alleles of one or more polymorphisms of the invention, by preferential binding, and sequences which hybridize under stringent conditions to a region either side of a polymorphism of the invention to enable amplification of one or more of the polymorphisms.

Methods of this embodiment include those known to persons skilled in the art, for example direct probing, allele specific hybridization, PCR methodology including Allele Specific Amplification (ASA), and RFLP.

Determination of an allele of a polymorphism using direct probing involves the use of anti-sense sequences of the third aspect of the invention. These may be prepared synthetically or by nick translation. The anti-sense probes may be suitably labeled using, for example, a radiolabel, enzyme label, fluoro-label, biotin-avidin label for subsequent visualization in, for example, a southern blot procedure. A labeled probe may be reacted with a sample DNA or RNA, and the areas of the DNA or RNA which carry complimentary sequences will hybridize to the probe, and become labeled themselves. The labeled areas may then be visualized, for example by autoradiography.

Allele specific amplification (ASA) discriminates between alleles of a polymorphism on the basis of primers which carry 3′ nucleotides specific for a particular polymorphism. Typically, first and second forward primers are provided, wherein the first forward primer hybridizes to one allele of a polymorphism of the invention, and the second forward primer comprises a mismatch at the polymorphic site, thus preventing hybridization. These primers are used in combination with a backward primer, which hybridizes to a distal site to enable amplification of the region between a forward primer and the backward primer. As the first forward primer will only bind to a polymorphic site with which it exhibits perfect complementarity, amplification of the region between the forward and backward primers will indicate the presence of a particular allele. The second forward primer having a mismatch at the polymorphic site will not hybridize to the particular allele of a polymorphism, and the absence of a amplification product when this primer is used indicates the absence of the polymorphism. Preferably, the forward primer will be an anti-sense sequence according to the third aspect of the invention. Preferably, the first forward primer will comprise the complement of a single nucleotide polymorphism of the invention at the 3′ most position. The backward primer may hybridize to any suitable portion of the corneodesmosin gene to enable amplification of the intervening region (see, for example, WO 93/22456).

Thus, in a preferred embodiment, there is provided a method for diagnosing or determining susceptibility of a subject to corneodesmosin-mediated disease, said method comprising removing a sample from a subject and isolating the nucleic acid therefrom; contacting the sample with either a forward primer which preferentially hybridizes to one allele of one or more polymorphisms of the present invention or a forward primer which comprises a mis-match at the polymorphic site and does not hybridize thereto, and a backward primer which hybridizes to a distal site; subjecting the nucleic acid sample to amplification; and monitoring for presence of an amplification product which is indicative of the presence of a particular allele of one or more of the polymorphisms of the invention. Preferably, a first reaction is performed using one of the forward primers, and a control reaction is then performed using the other forward primer. It is envisaged that a number alleles of the single nucleotide polymorphisms of the invention may be detected in a single reaction by using multiple primer pairs. Amplification products may then be distinguished by size, using techniques known in the art such as gel electrophoresis, or southern blotting. This method allows the unambiguous identification of individuals homozygous for either allele as well as heterozygous individuals.

“RFLP” refers to restriction fragment length polymorphism and is defined as a method of discriminating between two alleles based upon differences in sequence which result in the presence or absence of a restriction enzyme recognition site. In a preferred embodiment of the present aspect there is provided a method for diagnosing or determining susceptibility to corneodesmosin-mediated disease, said method comprising removing a nucleic acid sample from a subject, and contacting with one or more appropriate restriction enzymes. The size of fragments produced is indicative of which allele of one or more single nucleotide polymorphism according to the invention is present. An allele of a polymorphism of the invention may naturally produce a restriction enzyme site, thus allowing for determination of its presence by analysis of the restriction fragments produced. In some cases, however, an allele of a polymorphism does not create a restriction enzyme site, and one must be artificially introduced. This may be done by using a suitable mis-match primer, according to methods known in the art.

The appropriate restriction enzyme will, of course, be dependent upon the polymorphism and restriction site, and will include those known to persons skilled in the art. Preferred restriction enzymes are listed in Table 3 (ii), Column 8, with the expected fragment sizes in Columns 9, 10, and 11. Analysis of the digested fragments may be performed using any method in the art, for example gel analysis, or southern blots.

Preferably, the method may first comprise the amplification of a region of the corneodesmosin gene containing one or more of the polymorphic sites of the invention, for example, using PCR techniques. The probes of the present invention may be useful for this purpose.

The above-described methods may require amplification of the DNA sample from the subject, and this can be done by techniques known in the art, such as PCR (see Erlich (ed.) (1992) PCR Technology: Principles and Applications for DNA Amplification Freeman Press, NY; Innis et al. (1990) PCR Protocols: A Guide to Methods and Applications, Academic Press, San Diego, Calif.; Mattila et al. (1991) Nuc. Acids Res. 19:4967; Eckert et al. (1991) PCR Methods and Applications 1:17; and U.S. Pat. No. 4,683,202). Other suitable amplification methods include ligase chain reaction (LCR) (Wu et al. (1989) Genomics 4:560; Landegran et al. (1988) Science 241:1077); transcription amplification (Kwoh et al. (1989) PNAS 86:1173); self-sustained sequence replication (Guatelli et al. (1990) PNAS 87:1874) and nucleic acid-based sequence amplification (NASBA). The latter two methods both involve isothermal reactions based on isothermal transcription which produce both single stranded RNA and double-stranded DNA as the amplification products, in a ratio of 30 or 100 to 1, respectively.

It may often be desirable to identify the presence of multiple single nucleotide polymorphisms in a sample from a subject. This may be the case in the present invention where the corneodesmosin gene contains 39 polymorphisms, each of which may be indicative of a different phenotype. For this purpose, nucleic acid arrays may be useful, as described in WO 95/11995. The array may contain a number of probes, each designed to identify one or more of the above single nucleotide polymorphisms of the corneodesmosin gene, as described in WO 95/11995.

In a further preferred embodiment of the ninth aspect, the method may comprise determining which allele of one or more polymorphisms is present in a protein of the invention Any method for determining the presence of a particular form, or allele, of a protein is present, may be used. One such method involves the use of antibodies in diagnosing or determining susceptibility to corneodesmosin-mediated disease. The method may comprise removing a sample from a subject, contacting the sample with an antibody to an antigen of a protein or protein fragments according to the second aspect of the present invention, and detecting binding of the antibody to the antigen, wherein binding is indicative of the presence of a particular allele or form of the protein and thus risk to corneodesmosin-mediated disease. Tissue samples as described above are suitable for this method.

The detection of binding of the antibody to the antigen in a sample may be assisted by methods known in the art, such as the, use of a secondary antibody which binds to the first antibody, or a ligand. Immunoassays including immunofluorescence assays (IFA) and enzyme linked immunosorbent assays (ELISA) and immunoblotting may be used to detect the presence of the antigen. For example, where ELISA is used, the method may comprise binding the antibody to a substrate, contacting the bound antibody with the sample containing the antigen, contacting the above with a second antibody bound to a detectable moiety (typically an enzyme such as horse radish peroxidase or alkaline phosphatase), contacting the above with a substrate for the enzyme, and finally observing the color change which is indicative of the presence of the antigen in the sample.

In a tenth aspect of the invention, there is provided a method of treating a subject who has been diagnosed as having, or being susceptible to, corneodesmosin-mediated disease such as psoriasis. The mode of treatment will depend upon the nature of the polymorphism(s) and the phenotypic effect, and preferably comprises negating the effect of the disease causing polymorphism(s). Where a subject has been diagnosed according to the methods of the invention, treatment to negate the effect of the disease causing polymorphism may include any suitable means. A suitable treatment includes the administration of a polynucleotide sequence which hybridizes, preferably under stringent conditions (as defined above) to the corneodesmosin gene. Such polynucleotide sequences may include the anti-sense sequences of the third aspect. Alternatively, the treatment may comprise a polynucleotide sequence encoding the corneodesmosin gene or a fragment thereof, and having either a reference or variant allele of a polymorphism of the invention. Preferably, the method comprises (i) determining which allele of one or more polymorphisms of the invention are present; and (ii) administering a polynucleotide sequence which hybridizes under stringent conditions to the corneodesmosin gene, or a polynucleotide sequence encoding the reference sequence of the corneodesmosin gene or a fragment thereof, or a polynucleotide sequence of the first aspect.

In an alternative embodiment of this aspect, there is provided the use of a polynucleotide sequence of the tenth aspect in the manufacture of a medicament for use in the diagnosis and treatment of corneodesmosin-mediated disease.

This method of diagnosis and treatment may comprise determining and introducing alleles in the form of a polynucleotide or protein. In the above embodiments, the allele of a polymorphism may be determined using any method, as discussed above. The treatment may be introduced in the form of a protein or polynucleotide. Any suitable means for introduction of a protein may be used. Introduction of a polynucleotide may use gene therapy methods including those known in the art. In general, a polynucleotide encoding the allele will be introduced into the target cells of a subject, usually in the form of a vector and preferably in the form of a pharmaceutically acceptable carrier. Any suitable delivery vehicle may be used, including viral vectors, such as retroviral vector systems which can package a recombinant genome. The retrovirus could then be used to infect and deliver the polynucleotide to the target cells. Other delivery techniques are also widely available, including the use of adenoviral vectors, adeno-associated vectors, lentiviral vectors, pseudotyped retroviral vectors, and pox or vaccinia virus vectors. Liposomes may also be used, including commercially available liposome preparations such as Lipofectin®, Lipofectamine®, (GIBCO-BRL, Inc., Gaithersburg, Md.), SuperFect® (Qiagen GmbH, Hilden, Germany), and Transfectam® (Promega Biotec Inc., Madison, Wis.).

The polynucleotide or vehicle may be administered parenterally (e.g., intravenously), transdermally, by intramuscular injection, topically, or the like. As corneodesmosin-mediated diseases are usually manifested in the skin, topical administration is preferred. The exact amount of polynucleotide or vehicle to be administered will vary from subject to subject and will depend upon age, weight, general condition, and severity or mechanism of the disorder.

In a further aspect, the present invention provides a kit for the detection in a subject of a single nucleotide polymorphism according to the present invention. Preferably, the kit will contain polynucleotides according to the aforementioned aspects, most preferably the anti-sense sequences of the third aspect for use as probes or primers; antibodies of the fifth aspect; or restriction enzymes for use in detecting the presence of a polynucleotide, protein, or protein fragment of the invention. Preferably, the kit will also comprise means for detection of a reaction, such as nucleotide label detection means, labelled secondary antibodies or size detection means. In yet a further preferred embodiment, the polynucleotides, or antibodies may be fixed to a substrate, for example an array, as described in WO 95/11995.

The preferred embodiments of each aspect apply to the other aspects of the invention, mutatis mutandis.

The present invention will now be described by way of a non-limiting example with reference to the following figures in which:

FIG. 1 shows the nucleotide sequence (SEQ ID NO:1) of the genomic clone of the corneodesmosin gene, of GenBank Accession No. AC006163;

FIG. 2 shows the amino acid sequence (SEQ ID NO:2) of the corneodesmosin protein and coding sequence therefor; and

FIG. 3 shows the exon and intron structure of the corneodesmosin gene.

EXAMPLES Determination of Gene Structure

The mRNA sequence of the corneodesmosin gene (GenBank Accession ID NM001264) was used to screen the following public DNA databases: (available through the National Centre for Biotechnology Information Web site—www.ncbi.nlm.nih.gov); NR (Non-Redundant DNA), HTGS (High Throughput Genomic Sequence), and GSS (Genome Survey Sequence). The analysis was performed using the BLASTN algorithm (Altschul et al. (1990) J. Mol. Biol. 215:403-410). Any genomic sequences containing the corneodesmosin gene were identified by their degree of sequence identity. The gene structure was determined by comparison of the mRNA sequence with the genomic clones, The deduced exon-intron organisation of the corneodesmosin gene is presented in FIG. 3.

Oligonucleotide Primer Design—for Corneodesmosin Gene Sequencing

Five pairs of oligonucleotide primers (S1F/S1R; S2.1F/S2.1R; S2.2F/S2.2R; S2.3F/S2.3R; S2.4F/S2.4R, S2.5F/S2.5R—Table 1) were designed to amplify exons 1 and 2 of the corneodesmosin gene including 350 bp 5′ untranslated region (UTR) and 909 bp 3′ UTR sequences. Oligonucleotide primer sequences were derived from human chromosome 6p21 genomic DNA sequence (GenBank Accession AC006163).

TABLE 1 Oligonucleotide Primer DNA Sequences Primer ID Primer Sequence SEQ ID NO S 1F dCTGGGTCCCGTGGCAAGA 5 S 1R dGTCCTCTCCCGGAGTCTC 6 S 2.1F dGGTGAGGGAGGAAGCCAAG 7 S 2.1R dGAGCTGACGCTTTGGCCAC 8 S 2.2F dGCCAACCAATGACAACTCTTACC 9 S 2.2R dGCCTCCACAGAGCTGGAC 10 S 2.3F dGGCAAATACTTCTCCAGCAACC 11 S 2.3R dGGCCTTCTCCCATATGGGA 12 S 2.4F dCCAAGGAGAGTTACTCGACAG 13 S 2.4R dGGCATATTGGGTGGGTTGAC 14 S 2.5F dCATCTGGAAACAGTGGCCAC 15 S 2.5R dGTCTTCCTCCTCTGTGGGAG 16

Corneodesmosin Gene Amplification

Genomic DNA from a panel of 24 unrelated individuals was amplified using primer pairs S1F/S1R; S2.1F/S2.1R; S2.2F/S2.2R; S2.3F/S2.3R; S2.4F/S2.4R, S2.5F/S2.5R. 100 ng genomic DNA was amplified by PCR in a total reaction volume of 25 μl containing 50 mM KCl, 20 mM Tris-HCl (pH 8.4), 2 mM MgCl2 200 μM each dATP, dCTP, dGTP, dTTP, 1 μM each oligonucleotide primer and 0.5 units AmpliTaq Gold DNA polymerase (Applied Biosystems). Reactions were thermocycled with an initial denaturation step of 95° C./10 min followed by 35 cycles of 94° C./30 sec; Tm annealing/30 sec; 72° C./30 sec. A final elongation step of 72° C./10 min completed the amplification. Annealing temperatures (Tm) for specific primer pairs are presented in Table 2.

TABLE 2 Primer Annealing Temperatures and Amplimer Sizes Fragment size Tm Amplimer Primer Pairs (bp) (° C.) 1 S1F and S1R 495 63 2.1 S2.1F and 610 62 S2.1R 2.2 S2.2F and 619 62 S2.2R 2.3 S2.3F and 621 63 S2.3R 2.4 S2.4F and 532 59 S2.4R 2.5 S2.5F and 474 61 S2.5R

Heteroduplex Analysis Using DHPLC

Oligos were designed to amplify products of between 400-800 bp in length from the genomic DNA of 12-25 individuals. Denaturing high-performance liquid chromatography (DHPLC) analysis was performed using the WAVE™ DNA fragment analysis system (Transgenomic) (Kuklin et al. (1997-98) Genet. Test. 1(3):201-206). The temperature required for successful resolution of heteroduplex molecules within each PCR product was determined empirically by injecting PCR product at a series of increasing mobile phase temperatures and constructing a fragment specific melting curve. A universal gradient for double-stranded DNA was used to determine the appropriate acetonitrile concentration for the heteroduplex identification. For mutation detection, 1-2 ul aliquots of the PCR reactions from each of the eleven individuals were injected onto the WAVE™ column. Mutation detection gradients were for four minutes. Results were graphically visualized using the D-7000 HSM software (Transgenomic).

Direct Sequencing of PCR Products

50-100 ng of PCR products were sequenced in both orientations using the DYEnamic ET terminator cycle sequencing premix kit from Amersham. Reactions were fractionated on ABI 377 automated sequencers using standard procedures. Chromatographic traces were analyzed using the SEQUENCHER program (Gene Codes, USA), to identify SNP positions.

Detection of Variant Alleles—Assay Design for Genotyping

The fragment sequence containing the polymorphism was analyzed for the creation or deletion of a naturally occurring restriction enzyme recognition site in response to variation in the nucleotide sequence. If the polymorphism did not result in any changes in restriction enzyme recognition sites, then the sequence was interrogated with the Primer Design Mismatch Program™. This is an adaptation of the program described by Davidow (1992) Comput. Appl. Biosci. 8:193-194.

Detection of Polymorphisms in 24 Population Controls

The application of the approach outlined above resulted in the identification of 39 SNPs. These are described in Table 3, in which:

    • Column 1 designates each single nucleotide polymorphism a reference number.
    • Column 2 provides the positional reference of the polymorphism with respect to FIG. 1, together with details of the polymorphism itself. For example, the reference “C6948T” indicates a substitution of the nucleotide “C” for nucleotide “T” at position 6984 of FIG. 1.
    • Column 3 of (i) provides the corresponding positional references with respect to the coding sequence of the corneodesmosin gene.
    • Column 4 of (i) indicates the region of the gene which the polymorphism occurs.
    • Column 5 of (i) shows the sequence flanking the polymorphism, the polymorphism itself being shown in bold type. The single nucleotide polymorphisms are defined using standard IUB code.
    • Column 6 of (i) indicates the SEQ ID NO of the corresponding flanking sequence.
    • Columns 3 and 5 of (ii) show primer sequences which may be used to amplify a region of the corneodesmosin gene to enable detection of the single nucleotide polymorphism by using restriction enzyme analysis. The amplified product size is shown in Column 7 of (ii).
    • Columns 4 and 6 of (ii) indicate the SEQ ID NO of the corresponding primer sequence.
    • Columns 8 to 10 of (ii) list the restriction enzymes used to digest the amplified product, and the sizes of fragments generated by the reference, variant and heterozygous sequences respectively.

RFLP or ASA assays were developed for all of these SNPs, and the corresponding primers along with amplification product and digestion fragment sizes are also given in Table 3. Of these 39 SNPs, 9 give rise to amino acid changes. These are shown in Table 4.

Additional Corneodesmosin Polymorphisms

In a subsequent experiment, DNAs from 96 individuals comprising 24 type IA psoriatics, 24 type IB psoriatics, 24 type II psoriatics, and an additional 24 healthy controls were sequenced as described above using primers designed to cover the remainder of the corneodesmosin gene (see Table 5a)

The sequencing reactions were carried out with 50-100 ng of PCR products sequenced in both orientations using the DYEnamic ET terminator cycle sequencing premix kit from Amersham according to the following protocol.

The PCR products were Exo/Sap treated and desalted using p10 columns, prior to setting up the sequencing reactions in a thermowell plate including:

200-400 ng PCR Product

1 μl primer@10 pmol ml−1

8 μl ET Termination mix

H2O to 20 μl

The plates were sealed with an MJ Research Microseal film and then vortexed to mix samples, followed by a spin to ensure reaction is at the bottom of the wells.

PCR was carried out according to the following protocol:

    • No Predenaturation
    • 95° C. for 30 sec
    • 50° C. for 15 sec
    • 60° C. for 1 min
    • for 40 cycles and then hold at 10° C. until ready to purify.

After removing the plate from the thermocycler, the products were purified by ethanol precipitation. To each well we added 2 μl 7.5M ammonium acetate followed by 80 μl 100% ethanol, and incubated at room temperature for 10 minutes before spinning at 4000 rpm for 1 hour at room temperature. The supernatant was discarded and the pellet washed with 70% ethanol before centrifugation for a further 30 minutes. The supernatant was discarded and remaining ethanol removed gently by pipetting using p10 tips before allowing the pellets to air dry.

The samples were then resuspended in 10 μl MegaBACE Loading Buffer (Molecular Dynamics) and transferred to a Robbins plate prior to loading onto the MegaBACE. Reactions were fractionated on a Molecular Dynamics MegaBACE capillary sequencer using standard procedures. Chromatographic traces were analysed using the SEQUENCHER programme (Gene Codes, USA) to identify SNP positions.

A total of 28 novel SNPs were identified (additional to those given in the example above). For reference, these are SNPs 6-18 and 53-67 in Table 5b. A combined list of corneodesmosin SNPs is given in Table 6.

Corneodesmosin Gene Association with Psoriasis

A total of 21 SNPs (see Table 7) were genotyped in 147 families identified through a proband with psoriasis (a total of 499 individuals, of whom 233 were affected). The genotyping was carried out using a variety of methods (single base extension using the Snapshot kit from Amersham Pharmacia Biotech, Pyrosequencing (Ahmadian et al. (2000) Anal. Biochem. 280:103-110) or direct sequencing as given in Table 7. All these methods used established methodologies that are provided by the equipment manufacturers and/or are well known to those skilled in the art.

Linkage Disequilibrium

The extent of linkage disequilibrium (LD) between markers was calculated using genotype data from 199 unrelated, unaffected individuals and is expressed as correlation coefficients in Table 8. This analysis shows that there is extensive linkage disequilibrium between many of the corneodesmosin polymorphisms.

Single Point Association

Single point associations between each SNP and psoriasis affected status were calculated using the TRANSMIT program (D. Clayton, MRC Biostatistics Unit, Cambridge)—see Table 9. Highly significant associations were observed between SNPs 19, 21, 23, 24, 26, 28, 30, 332 34, 37, 38, and psoriasis. The single SNP showing the most significant association with psoriasis that has been previously reported is SNP 33 (Tazi-Ahnini et al. (1999) Hum. Mol. Genet. 8:1135-1140; Allen et al. (1999) Lancet 353:1589-1590).

This study has identified nine SNPs, (19, 21, 24, 26, 28, 30, 34, 37, and 38) which show global chi-squared values greater than that seen for SNP 33, and are therefore more powerfully predictive of affected status.

Haplotype Analysis

A total of 19 SNPs were used for haplotype analysis (SNPs at positions 29 and 32 were excluded due to low information content). Three common haplotypes were identified (Table 10). Of the three common haplotypes, haplotype B is significantly associated with psoriasis. The alleles are coded alphabetically (Table 10b) such that the nucleotide first in the alphabet is coded as 1, and the other nucleotide is coded as 2. Thus, A is always 1, T is always 2, and G or C are coded depending on the other nucleotide. For example, in SNP No. 1, which is a C to T substitution, the presence of the C allele is coded as 1 and the presence of the T allele is coded as 2 (see Table 10b). In Table 10a, this means that haplotypes A and B have C residues, and haplotype C has a T residue at this position. For an A to C substitution, the A allele will be coded as 1, and the C allele as 2. In a C to G substitution, the C allele will be coded as 1 and the G allele as 2.

Construction of Corneodesmosin Gene Targeting Vector

As the genetic data pointed strongly to an involvement of the corneodesmosin gene in the pathophysiology of psoriasis, we decided to engineer mouse strains in which the mouse orthologue of the corneodesmosin gene is knocked out by homologous recombination using a vector construct designed to remove exon 2 of the corneodesmosin gene.

Murine corneodesmosin genomic clones were isolated from a mouse large insert PAC library, using mouse corneodesmosin cDNA sequence as a probe by standard techniques. The isolated murine corneodesmosin genomic clones were then restriction mapped in the region of the corneodesmosin gene using small oligonucleotide probes and standard techniques. The murine genomic locus was partially sequenced to enable the design of homologous arms to clone into the targeting vector. The murine corneodesmosin gene is a two-exon gene. A 4 kb 5′ homologous arm and a 1 kb 3′ homologous arm where amplified by PCR and the fragment cloned into the targeting vector. The position of these arms was chosen to functionally disrupt the corneodesmosin gene by deleting the majority of the coding sequence. A targeting vector was prepared where the deleted corneodesmosin sequence was replaced with non-homologous sequences composed of an endogenous gene expression reporter (an in frame fusion with lacZ) upstream of a selection cassette composed of a self promoted neomycin phosphotransferase (neo) gene in the same orientation as the corneodesmosin gene.

Transfection and Analysis of Embryonal Stem Cells

Embryonal stem cells (Evans and Kaufman (1981) Nature 292:154-156) were cultured on a neomycin-resistant embryonal fibroblast feeder layer grown in Dulbecco's Modified Eagles medium supplemented with 20% Fetal Calf Serum, 10% new-born calf serum, 2 mM glutamine, non-essential amino acids, 100 μM 2-mercaptoethanol and 500 u/ml leukemia inhibitory factor. Medium was changed daily and ES cells were subcultured every three days. 5×106 ES cells were transfected with 5 μg of linearized plasmid by electroporation (25 μF capacitance and 400 Volts). 24 hours following electroporation the transfected cells were cultured for nine days in medium containing 200 μg/ml neomycin. Clones were picked into 96 well plates, replicated and expanded before being screened by PCR to identify clones in which homologous recombination had occurred between the endogenous corneodesmosin gene and the targeting construct. From 96 picked clones, 45 targets were identified. These clones where expanded to allow replicas to be frozen and sufficient high quality DNA to be prepared for Southern blot confirmation of the targeting event using external 5′ and 3′ probes, all using standard procedures (Russ et al. (2000) Nature 404:95-99).

Generation of Corneodesmosin-Deficient Mice

C57BL/6 female and male mice were mated and blastocysts were isolated at 3.5 days of gestation. 10-12 cells from Clone 7 (described in Example 2) were injected per blastocyst and 7-8 blastocysts were implanted in the uterus of a pseudopregnant F1 female. Five chimeric pups were born of which one male was 100% agouti (indicating cells descendent from the targeted clone). This male chimera was mated with female and MF1 and 129 mice, and germline transmission was determined by the agouti coat color and by PCR genotyping, respectively.

Corneodesmosin Knock-Out Mouse as a Model of Corneodesmosin-Mediated Disease

Mice heterozygous for the corneodesmosin knockout are superficially normal. Staining for expression of the lacZ reporter gene fused to the corneodesmosin promoter in the knockout construct shows clear expression in desquamating skin. We then genotyped surviving offspring from intercrosses of heterozygous knockout mice on an outbred genetic background in an attempt to isolate mice homozygous for the knockout.

From 44 surviving progeny we identified:

    • 17 wild type
    • 27 heterozygotes
    • 0 homozygous mutant.

Statistical analysis of these data indicate that the ratio of wild type:heterozygous animals conforms to a 1:2 ratio consistent with a homozygous lethal phenotype (Chi square=0.557).

In keeping with this analysis, two pups found dead 24-48 hours after birth were homozygous mutant. Together, these data indicate the corneodesmosin deficiency in mice is lethal with pups dying soon after birth, most likely through dehydration as a result of failure to establish a permeability barrier in the skin.

We conclude from this that altering the activity of corneodesmosin (e.g., by modulating expression or altering its proteolytic processing) will be useful in developing models of disease in which epithelial integrity is increased (e.g., psoriasis) or decreased (e.g., dermatitis), and for testing novel agents for the alleviation of corneodesmosin-mediated disease.

TABLE 3i S Gene SNPs With Location and Assay Details Corneo- desmosin SEQ SNP nt ID SNP nt position position Location Flanking Sequence NO 1 C6984T −115 5′ UTR CTCCCGGCCA CACCAACTTC CCCCYGGGCA CCCACCCCCT CCACCTCTCC 17 2 A7068G −31 5′ UTR AATGTCCAGCTCTGGCATAA AGGACCCRGG TGTCCTCGAG CTGCCATCAG 18 3 C7077T −22 5′ UTR TCTGGCATAA AGGACCCAGG TGTCCTYGAG CTGCCATCAG TCAGGAGGCC 19 4 C7107T 9 5′ UTR CTGCCATCAG TCAGGAGGCCGTGCAGYCCG AGATGGGCTC GTCTCGGGCA 20 5 A7164T 66 Coding Sequence GGCGTGTGGGTGGGCACGGG ATGWTGGCAC TGCTGCTGGC TGGTCTCCTC 21 6 C10039T 137 Coding Sequence CTAAGAGCAT TGGCACCTTC TCAGACCCYT GTAAGGACCCCACGCGTATC 22 7 C10082T 180 Coding Sequence ACCTCCCCTAACGACCCCTGCYTCACTGGGAAGGGTG 23 8 C10134T 206 Coding Sequence CAGTAGCTAC AGTGGCTCCA GCAYTTCTGG CAGCTCCATTTCCAGTGCCA 24 9 G10344A 442 Coding Sequence GAGCAGCAGC TCTCACTCGG GAARCAGCGGCTCTCACTCG GGAAGCAGCA 25 10 10363(AAG)ins 461 Coding Sequence GAAGCAGCGGCTCTCACTCG GG(AAG)CAGCA GCTCTCATTCGAGCAGCAGC 26 11 A10516G 614 Coding Sequence CTGGACAAAGCTCTTCCTCT TCCCARACCT CTGGGGTATC CAGCAGTGGC 27 12 C10521T 619 Coding Sequence CTGGACAAAGCTCTTCCTCT TCCCAAACCT YTGGGGTATC CAGCAGTGGC 28 13 T10624C 722 Coding Sequence GGAGGGCCCA TCGTCTCGCA CTCYGGCCCC TACATCCCCA GCTCCCACTC 29 14 G10669A 767 Coding Sequence GCTCCCACTCTGTGTCAGGG GGTCAGAGRC CTGTGGTGGT GGTGGTGGAC 30 15 T10873C 971 Coding Sequence CCTACAGTAA GGGTAAAATC TAYCCTGTGG GCTACTTCAC CAAAGAGAAC 31 16 G11020A 1118 Coding Sequence AGCCAGTCGGCAGCTTCCTC GGCCATTGCR TTCCAGCCAG TGGGGACTGG 32 17 A11117G 1215 Coding Sequence CTCCCTCCAGTTCTCGAGTC CCCAGCRGTT CTAGCATTTC CAGCAGCTCC 33 18 T11138G 1236 Coding Sequence CCCAGCAGTT CTAGCATTTC CAGCAGCKCC GGTTCACCCTACCATCCCTG 34 19 G11142T 1240 Coding Sequence CTAGCATTTC CAGCAGCTCC GKTTCACCCTACCATCCCTGCGGCAGTGCT 35 20 C11145T 1243 Coding Sequence CTAGCATTTC CAGCAGCTCC GGTTYACCCTACCATCCCTGCGGCAGTGCT 36 21 G11233C 1331 Coding Sequence GCAGCAGCTC CAGTTCCCAA TCSAGTGGCA AAATCATCCTTCAGCCTTGT 37 22 T11260C 1358 Coding Sequence TCGAGTGGCA AAATCATCCTTCAGCCTTGY GGCAGCAAGT CCAGCTCTTC 38 23 G11495A 1593 Coding Sequence TTCCTACCCC AAGGAGAGTT ACTCRACAGTCCATAAGTCA ACTGTTGTGT 39 24 11505(AAG)ins 1603 3′ UTR GAGAGTTACTCGACAGTCCATAAG(AAG)TCAACTGTTGTGTGTGTGCATGC 40 25 G11576T 1674 3′ UTR TACACTATATCCCATATGGGAGAAGKCCAGTGCCCAGGCATAGGGTTAGC 41 26 T11641C 1739 3′ UTR CCCAAAAGAGTGGTTCTGCTTTCTCYACTACCCTAAGGTTGCAGACTCTC 42 27 T11649C 1747 3′ UTR AGTGGTTCTGCTTTCTCTACTACCCYAAGGTTGCAGACTCTCTCTTATCA 43 28 T11808G 1906 3′ UTR CCCCTTACAATTCCCTCTACTGTGTKGAAATGGTCCATTGAGTAACACCC 44 29 C11839G 1937 3′ UTR GGTCCATTGAGTAACACCCCCATCASCTTCTCAACTGGGAAACCCCTGAA 45 30 C11885T 1983 3′ UTR TGAAATGCTCTCAGAGCACCTCTGAYGCCTGAAGAAGTTATACCTTCCTC 46 31 C11977T 2075 3′ UTR AAACAGTGGC CACTTTTCAC TGACCTYTCT TCGACATCTA GTCAACCCAC 47 32 T12018C 2116 3′ UTR CAACCCACCCAATATGCCACTGGGCYTTCGCTCCCAATTCCACCCCACCC 48 33 T12136C 2234 3′ UTR TTATCTCAGCCCCTTCCTGTGGCCAYTTCCCTCAGTGCCCAGATGATTCC 49 34 C12149T 2247 3′ UTR TTCCTGTGGCCATTTCCCTCAGTGCYCAGATGATTCCCTGGGTGAGGGAG 50 35 G12198A 2296 3′ UTR GACACTGGGGCACCCTCAGAGGTTGRAGCAGGCTCCCTGCTGTCCCTGGA 51 36 G12283A 2381 3′ UTR GGTGCAGACTTTTTGCCTTCTTGGARTCCTGGGTCTCCTCTGAGAGTCTG 52 37 T12318C 2416 3′ UTR TCCTCTGAGAGTCTGGGTGGTGCTCYTCCTACGCCTCTAGAGGTCTCTGT 53 38 C12345T 2443 3′ UTR CCTACGCCTCTAGAGGTCTCTGTGTYCCTCATTTTCCTTCAAAAGCGGGC 54 39 G12373A 2471 3′ UTR TCATTTTCCTTCAAAAGCGGGCTGTRTTTCTCTTCTACCTTCCAGCTCCT 55

TABLE 3ii PCR SNP SEQ SEQ product nt ID ID size Allele Allele Hetero- SNP position Primer sequence NO Primer sequence NO (bp) Enzyme 1 2 zygote 1 C6984T dCTGGGTCCCGTGGCAAGA 5 dGTCCTCTCCCGGAGTCTC 6 496 Ava I 313, 220, 313, 220, 32, 16, 93, 32, 135, 93, 135 16, 135 32 16 2 A7068G dCTGGGTCCCGTGGCAAGA 5 dCTGACTGATGGCAGCTCGAG 58 333 Pvu II 333 309, 24 333, 309, GACAGC 24 3 C7077T dCTGGGTCCCGTGGCAAGA 5 dGTCCTCTCCCGGAGTCTC 6 496 Tag I 496 315, 496, 315, 181 181 4 C7107T dCCCACCCCCTCCACCTCT 59 dCCGTCCCCTTCGCTGGGTCC 60 283 Ava I 150, 150, 150, 85, TC 85, 48 85, 32, 48, 32, 16 16 5 A7164T dATTACCACGCTCCTCCCG 61 dGCAGGAGGAGACCAGCCAGC 62 249 Hinc 249 220, 29 249, 220, AGCAGTGTCA II 29 6 C10039T dCAGTTCTTCCTCCTTTCTCCA 63 dAGGGGAGGTGATACGCGTGG 64 215 BstN I 215 184, 31 215, 184, T GGTCCTTCCA 31 7 C10082T dGACCTTGGCTAAGAGCATTG 65 dCCTGGCTTAAAAGATCCTGC 66 240 Mnl I 240 151, 240, 151, 70, 19 70, 19 8 C10134T dGGTGAGGGAGGAAGCCAAG 7 dAGAACTGCTGGAGCCACTGT 68 193 Pst I 193 163, 30 193, 163, AGCTACTGCA 30 9 G10344A dCAGCTGGGGAGCAGCAGCTCT 69 dGAGCTGACGCTTTGGCCAC 8 243 Bsl I 243 219, 22 243, 219, CCCTCGGGA 22 10 10363 dAGCGGCTCTCACTCGGGAAG 71 dTGACGCTTTGGCCACTGCTG 72 204 204 204 204 (AAG)ins dAGCGGCTCTCACTCGGGCAG 73 11 A10516G dCAGCCTGGACAAAGCTCTTCC 74 dCTGGAAGGCCACCATTGCTA 75 269 Dde I 269 243, 26 269, 243, TCTTCTCA 26 12 C10521T dGCCAACCAATGACAACTCTTA 9 dGCCTCCACAGAGCTGGAC 10 620 BsrF I 162 132, 30 162, 132, CC 30 13 T10624C dCTGCAGTGGAGGGCCCATCGT 78 dCTGGAAGGCCACCATTGCTA 79 162 BsmA 451, 401, 451, 401, CTCGCACAC I 169 169, 50 169, 50 14 G10669A dGCCAACCAATGACAACTCTTA 76 dGCCTCCACAGAGCTGGAC 77 620 Mbo I 190, 221 221, 190, CC 31 31 15 T10873C dAGGCATGACCTACAGTAAGGG 80 dGCCTCCACAGAGCTGGAC 77 221 Mnl I 62, 62, 12, 270, 258, TAAAATCGA 270, 258, 186, 62, 45, 45, 57, 45, 186, 186, 57 12 57 16 G11020A dCAGCCAGTCGGCAGCTTCCTC 81 dTGAAGGAGCCGGTGCCTG 82 225 BstU I 225 195, 30 225, 195, GGCCATCGC 30 17 A11117G dTGCTCTCCCTCCAGTTCTCGA 83 dGTGTCAAGGAGGAGACAGAC 84 231 Pst I 231 199, 32 231, 199, GTCCCCTGC A 32 18 T11138G dGGCAAATACTTCTCCAGCAAC 11 dGGCCTTCTCCCATATGGGA 12 622 Hha I 622 440, 622, 440, C 182 182 19 G11142T dGGCAAATACTTCTCCAGCAAC 11 dGGCCTTCTCCCATATGGGA 12 622 Msp I 241, 181, 241, 214, C 214, 60, 181, 167, 167 214, 60 167 20 C11145T dGTTCTAGCATTTCCAGCAGCT 87 dGTGTCAAGGAGGAGACAGAC 88 200 Hinf I 200 176, 24 200, 176, CCGATT A 24 21 G11233C dGGCAAATACTTCTCCAGCAAC 11 dGGCCTTCTCCCATATGGGA 12 622 Taq I 146, 146, 389, 262, C 389, 127, 146, 127, 87 262, 87 87 22 T11260C dGGCAAATACTTCTCCAGCAAC 11 dGTGACCAGAAGAGCTGGACT 89 331 Hha I 331 304, 27 331, 304, C TGCTGGC 27 23 G11495A dGGCAAATACTTCTCCAGCAAC 11 dGGCCTTCTCCCATATGGGA 12 622 Taq I 146, 146, 349, 262, C 127, 127, 146, 127, 349 262, 87 87 24 11505 dGGAGAGTTACTCGACAGTCCA 90 dCAGTAGGAGAGAATCAAGAG 91 259 259 259 259 (AAG)ins TAAGAAG AGGAGC dGGAGAGTTACTCGACAGTCCA 92 dCAGTAGGAGAGAATCAAGAG 91 TAAGTCA AGGAGC 25 G11576T dAAGGAGAGTTACTCGACAGTC 93 dAGGAGAGAATCAAGAGAGGA 94 254 Hae 254 96, 158 254, 158, C GC III 96 26 T11641C dAAGGAGAGTTACTCGACAGTC 93 dTAAGAGAGAGTCTGCAACCT 95 190 Aci I 190 160, 30 190, 160, C TAGGGTAGC 30 27 T11649C dAAGGAGAGTTACTCGACAGTC 93 dAGGAGAGAATCAAGAGAGGA 94 254 Bsu 254 168, 86 254, 168, C GC 36 I 86 28 T11808G dAGGTTGCAGACTCTCTCTTAT 96 dATGGGGGTGTTACTCAATGG 97 186 Hinc 186 158, 28 186, 158, CACCC ACCATGTC II 28 29 C11839G dAGGTTGCAGACTCTCTCTTAT 96 dAAGTGGCCACTGTTTCCAGA 98 315 Alu I 315 252, 63 315, 252, CACCC TGATGG 63 30 C11885T dAGGTTGCAGACTCTCTCTTAT 96 dAAGTGGCCACTGTTTCCAGA 98 315 BsaH 315 234, 81 315, 234, CACCC TGATGG I 81 31 C11977T dCCATCATCTGGAAACAGTGG 99 dCGTGGTGAGCTCTGTAATGG 100 124 Ear I 124 80, 44 124, 80, 44 32 T12018C dACCATCATCTGGAAACAGTGG 101 dTGAGCTCTGTAATGGAGGGT 102 120 Ban II 120 80, 38, 120, 80, C GG 2 38, 2 33 T12136C dCCTTATCTCAGCCCCTTCCTG 103 dATCTGTCCAGGATCCAGGGA 104 126 Ear I 126 94, 32 126, 94, TGGCCT CAGC 32 34 C12149T dAACACACCCATTGCCTCTCAA 105 dCCACAGTTTACTGAGCCATC 106 167 Bsi 167 102, 65 167, 102, G TG HKA I 65 35 G12198A dCCTTATCTCAGCCCCTTCCTG 107 dAGGATCCAGGGACAGCAGGG 108 118 Sau 118 89, 29 118, 89, TGGC AGCCTGGT 96 I 29 36 G12283A dGGACAGATGGCTCAGTAAACT 109 dAGGGACACAGAGACCTCTAG 110 122 Tfi I 122 67, 55 122, 67, G 55 37 T12318C dGGACAGATGGCTCAGTAAACT 109 dAGGGACACAGAGACCTCTAG 110 122 Ear I 122 95, 27 122, 95, G 27 38 C12345T dCTCTTCCTACGCCTCTAGAGG 111 dGCAATGAGAGAGGAGGGAAA 112 179 Eco 179 151, 28 179, 151, TCTCTGGGT TGGCG 0109 I 28 39 G12373A dGTCCCTCATTTTCCTTCAAAA 113 dGGGAAGAGAATGGATTTCCT 114 174 Tsp 174 141, 33 174, 141, GCGGGCAG GGAGC R I 33

TABLE 4 Amino Acid Polymorphisms SNP POSITION LOCATION VARIANT I VARIANT 2 Effect on amino acid side chain 5 A7164T EXON 1 MET LEU Conservative 6 C10039T EXON 2 PRO PRO Neutral 7 C10082T EXON 2 LEU SER Hydrophobic - Hydrophilic 8 C10108T EXON 2 GLY GLY Neutral 9 G10344A EXON 2 SER ASN Conservative 10 10363 (AAG)ins EXON 2 SER insertion SER deletion SER insertion/deletion 11 A10516G EXON 2 GLN GLN Neutral 12 C10521T EXON 2 SER PHE Hydrophilic - Hydrophobic 13 T10624C EXON 2 SER SER Neutral 14 G10669A EXON 2 ARG ARG Neutral 15 T10873C EXON 2 TYR TYR Neutral 16 G11020A EXON 2 ALA ALA Neutral 17 A11117G EXON 2 SER GLY Hydrophilic - Hydrophobic 18 T11138G EXON 2 SER ALA Hydrophilic - Hydrophobic 19 G11142T EXON 2 GLY VAL Conservative 20 C11145T EXON 2 SER LEU Hydrophilic - Hydrophobic 21 G11233C EXON 2 SER SER Neutral 22 T11260C EXON 2 CYS CYS Neutral 23 G11495A EXON 2 ASP ASN Hydrophilic charged - Hydrophilic neutral

TABLE 5 Primer Sequence SEQ ID Primer Sequence SEQ ID Primer Name Forward NO Reverse NO SEEK INI_8 CAGTGAGCTGAGACCGTG 115 CTGGTACCAGTGTGTCAG 116 SEEK INI_8 CAGTGAGCTGAGACCGTG 115 CTGGTACCAGTGTGTCAG 116 SEEK INI_8 CAGTGAGCTGAGACCGTG 115 CTGGTACCAGTGTGTCAG 116 SEEK INI_6 GACTCCTCAGAGCCTCAG 117 GTAGCTACTGAAGCCGCTG 118 SEEK INI_6 GACTCCTCAGAGCCTCAG 117 GTAGCTACTGAAGCCGCTG 118 SEEK INI_6 GACTCCTCAGAGCCTCAG 117 GTAGCTACTGAAGCCGCTG 118 SEEK INI_6 GACTCCTCAGAGCCTCAG 117 GTAGCTACTGAAGCCGCTG 118 SEEK INI_6 GACTCCTCAGAGCCTCAG 117 GTAGCTACTGAAGCCGCTG 118 SEEK INI_6 GACTCCTCAGAGCCTCAG 117 GTAGCTACTGAAGCCGCTG 118 SEEK INI_6 GACTCCTCAGAGCCTCAG 117 GTAGCTACTGAAGCCGCTG 118 SEEK INI_6 GACTCCTCAGAGCCTCAG 117 GTAGCTACTGAAGCCGCTG 118 SEEK INI_6 GACTCCTCAGAGCCTCAG 117 GTAGCTACTGAAGCCGCTG 118 SEEK INI_6 GACTCCTCAGAGCCTCAG 117 GTAGCTACTGAAGCCGCTG 118 SEEK INI_6 GACTCCTCAGAGCCTCAG 117 GTAGCTACTGAAGCCGCTG 118 SEEK PROM3 CCTAGATCAAGAGGCCCAG 119 ACAGCAGGAGACTCGAGG 120 SEEK PROM3 CCTAGATCAAGAGGCCCAG 119 ACAGCAGGAGACTCGAGG 120 SEEK PROM3 CCTAGATCAAGAGGCCCAG 119 ACAGCAGGAGACTCGAGG 120 SEEK PROM3 CCTAGATCAAGAGGCCCAG 119 ACAGCAGGAGACTCGAGG 120 SEEK PROM3 CCTAGATCAAGAGGCCCAG 119 ACAGCAGGAGACTCGAGG 120 SEEK PROM3 CCTAGATCAAGAGGCCCAG 119 ACAGCAGGAGACTCGAGG 120 SEEK PROM3 CCTAGATCAAGAGGCCCAG 119 ACAGCAGGAGACTCGAGG 120 SEEK PROM3 CCTAGATCAAGAGGCCCAG 119 ACAGCAGGAGACTCGAGG 120 SEEK PROM3 CCTAGATCAAGAGGCCCAG 119 ACAGCAGGAGACTCGAGG 120 SEEK PROM2 CCTCAGATGCTTCATGAATGG 121 GTGAAGTCAGCCGAATAGC 122 SEEK PROM2 CCTCAGATGCTTCATGAATGG 121 GTGAAGTCAGCCGAATAGC 122 SEEK PROM2 CCTCAGATGCTTCATGAATGG 121 GTGAAGTCAGCCGAATAGC 122 SEEK PROM2 CCTCAGATGCTTCATGAATGG 121 GTGAAGTCAGCCGAATAGC 122 SEEK PROM2 CCTCAGATGCTTCATGAATGG 121 GTGAAGTCAGCCGAATAGC 122 SEEK PROM2 CCTCAGATGCTTCATGAATGG 121 GTGAAGTCAGCCGAATAGC 122

TABLE 6 Corneo- AC006163 desmosin Location SEQ nt nt in IUB ID SNP position position gene Flanking Sequence Code NO 1  6,984 bp −115 5′UTR TACCACGCTCCTCCCGGCCACACCAACTTCCCCC/TGGGGCACCCACCCCCTCCACCTCTC Y 123 CTCCTCTCCC 2  7,068 bp −31 5′UTR TGCCCAGGGAATGTCCAGCTCTGGCATAAAGGACCCA/GGGTGTCCTCGAGCTGCCATCAG R 124 TCAGGAGGCCG 3  7,077 bp −22 5′UTR AGCTCTGGCATAAAGGACCCAGGTGTCCTC/TGAGCTGCCATCAGTCAGGAGGCCGTGCAG Y 125 CCCGAGATGGGC 4  7,107 bp 9 5′UTR GAGCTGCCATCAGTCAGGAGGCCGTGCAGC/TCCGAGATGGGCTCGTCTCGGGCACCCTGG Y 126 ATGGGGCGT 5  7,164 bp 66 Exon 1 GCACCCTGGATGGGGCGTGTGGGTGGGCACGGGATGA/TTGGCACTGCTGCTGGCTGGTCT W 127 CCTCCTGCCAGG 6  8,884 bp Intron I Intron I CTGGAGGGGCTAGGGAAGGCAGAAGGAACGCAGGT/AGAAAGAGTCATGGAGGAACCATGG W 128 GGTAAGTT 7  8,906 bp Intron I Intron I CAGAAGGAACGCAGGTGAAAGAGTCATGGAGGAACCAT/CGGGGTAAGTTGGGCCTGGGGT Y 129 TTTGAGCAA 8  8,931 bp Intron I Intron I GGAGGAACCATGGGGTAAGTTGGGCCTGGGGTTTTG/CAGCAAAGGAAAGGAAAGATAAGG S 130 AAAGATGTGGCTC 9  9,538 bp Intron I Intron I CTGTCTCTTCAGGGTCCTTTCTTTTAGACCTAT/CTTGTTCCTGCCCCTTCTCCATTCCCT Y 131 CTTCTTTT 10  9,607 bp Intron I Intron I AAAAAAATTTTAATTAAAAAACAAAATACAGAT/CGGGGTCTATGTTGCCCAGGCTGGTCT Y 132 TGAACTCTGGGGCGC 11  9,608 bp Intron I Intron I AAAAAAATTTTAATTAAAAAACAAAATACAGATG/AGGGTCTATGTTGCCCAGGCTGGTCT R 133 TGAACTCTGGGGCGC 12  9,647 bp Intron I Intron I GGGTCTATGTTGCCCAGGCTGGTCTTGAACTCTGGGGCG/ACATGCAATCCTCCCACCTCA R 134 GCCTCCCAAAGTGCTGG 13  9,667 bp Intron I Intron I TCTTGAACTCTGGGGCGCATGCAATCCTCCCACCTCA/GGCCTCCCAAAGTGCTGGGATTA R 135 CCGGCGTGAGCCACT 14  9,745 bp Intron I Intron I AGCCCCCTCTTATATTCAATGTATTCCTTTGAGGT/CCACTCACTTTGGCACGTAATTTTC Y 136 TATTTTTCTGGTTG 15  9,761 bp Intron I Intron I TCAATGTATTCCTTTGAGGTCACTCACTTTGGCACG/CTAATTTTCTATTTTTCTGGTTGG S 137 TGTTTGCCCACCCTT 16  9,926 bp Intron I Intron I CCCTGCGCTCTGCTTGGGAGAAACCCGAGAGGCCGATT/GACTGAGATAAGGCAGAAAGGT K 138 GAGGGAGGAAGCCA 17  9,952 bp Intron I Intron I AGAGGCCGATTACTGAGATAAGGCAGAAAGGTGAGGG/AAGGAAGCCAAGCCTCTTTGGCC R 139 CTTACTAACCACTG 18  9,968 bp Intron I Intron I ACTGAGATAAGGCAGAAAGGTGAGGGAGGAAGCCAAGCCTCT/CTTGGCCCTTACTAACCA Y 140 CTGCTTTCCTCCACAGGGACCTTG 19 10,039 bp 137 Exon 2 CAGGGACCTTGGCTAAGAGCATTGGCACCTTCTCAGACCCC/TTGTAAGGACCCCACGCGT Y 141 ATCACCTCCCCTAACGACCCCT 20 10,082 bp 180 Exon 2 GGACCCCACGCGTATCACCTCCCCTAACGACCCCTGCC/TTCACTGGGAAGGGTGACTCCA Y 142 GCGGCT 21 10,108 bp 206 Exon 2 ACGACCCCTGCCTCACTGGGAAGGGTGACTCCAGCGGC/TTTCAGTAGCTACAGTGGCTCC Y 143 AGCAGTTCTGGCAGCTCCAT 22 10,344 bp 442 Exon 2 CCGGTTCCTCCCAGCTGGGGAGCAGCAGCTCTCACTCGGGAAG/ACAGCGGCTCTCACTCG R 144 GGAAGCAGCAGCTCTCATTCG 23 10,363 bp 461 Exon 2 GGAGCAGCAGCTCTCACTCGGGAAGCAGCGGCTCTCACTCGGG(AAG)CAGCAGCTCTCAT Ins/del 145 (ins) TCGAGCAGCAGCAGCAGCTT 24 10,516 bp 614 Exon 2 AATACTAAACCCTTCCCAGCCTGGACAAAGCTCTTCCTCTTCCCAA/GACCTYTGGGGTAT R 146 CCAGCAGTGGCCAAAGCGTCAGCTCC 25 10,521 bp 619 Exon 2 AATACTAAACCCTTCCCAGCCTGGACAAAGCTCTTCCTCTTCCCAAACCTC/TTGGGGTAT Y 147 CCAGCAGTGGCCAAAGCGTCAGCTCC 26 103624 bp 722 Exon 2 CGACTCTCCCTGCAGTGGAGGGCCCATCGTCTCGCACTCT/CGGCCCCTACATCCCCAGCT Y 148 CCCACTCTGTGTC 27 10,669 bp 767 Exon 2 CCTACATCCCCAGCTCCCACTCTGTGTCAGGGGGTCAGAGG/ACCTGTGGTGGTGGTGGTG R 149 GACCAGCACGGTTCTGGTGC 28 10,873 bp 971 Exon 2 ACAGTTATCTGGTTCCAGGCATGACCTACAGTAAGGGTAAAATCTAT/CCCTGTGGGCTAC Y 150 TTCACCAAAGAGAACCCTGTGA 29 11,020 bp 1118 Exon 2 ACCCCATCATCCCCAGCCAGTCGGCAGCTTCCTCGGCCATTGCG/ATTCCAGCCAGTGGGG R 151 ACTGGTGGGGTCCAGC 30 11,117 bp 1215 Exon 2 CCAAGGGACCCTGCTCTCCCTCCAGTTCTCGAGTCCCCAGCA/GGTTCTAGCATTTCCAGC R 152 AGCTCCGGTTCACCCTA 31 11,138 bp 1236 Exon 2 CTCGAGTCCCCAGCAGTTCTAGCATTTCCAGCAGCT/GCCGGTTCACCCTACCATCCCTGC K 153 GGCAGTGCTT 32 11,142 bp 1240 Exon 2 CTAGCATTTCCAGCAGGTCCG G/T TTCACCCTACCATCCCTGCGGCAGTGCT K 154 33 11,145 bp 1243 Exon 2 CCAGCAGTTCTAGCATTTCCAGCAGCTCCGGTTC/TACCCTACCATCCCTGCGGCAGTGCT Y 155 TCCCAGAG 34 11,233 bp 1331 Exon 2 GGCACCGGGTCCTTCAGCAGCAGCTCCAGTTCCCAATCG/CAGTGGCAAAATCATCCTTCA S 156 GCCTTGTGGCAGCAA 35 11,260 bp 1358 Exon 2 AGTTCCCAATCGAGTGGCAAAATCATCCTTCAGCCTTGT/CGGCAGCAAGTCCAGCTCTTC Y 157 TGGTCACCCTTGC 36 11,495 bp 1593 Exon 2 TGAAGTTTTCCTACCCCAAGGAGAGTTACTCG/AACAGTCCAT(AAG)AAGTCAACTGTTG R 158 TGTGTGTGCAT 37 11,505 bp 1603 3′ UTR TACCCCAAGGAGAGTTACTCGACAGTCCAT(AAG)AAGTCAACTGTTGTGTGTGTGCATGC Ins/del 159 (ins) CTTGGGCACAAA 38 11,575 bp 1674 3′ UTR GGCACAAACAAGCACATACACTATATCCCATATGGGAGAAGG/TCAGTGCCCAGGCATAGG K 160 GTTAGCTCAGTTTCCCTCCTTCCCA 39 11,641 bp 1739 3′ UTR AGCTCAGTTTCCCTCCTTCCCAAAAGAGTGGTTCTGCTTTCTCT/CACTACCCTAAGGTTG Y 161 CAGACTCTCTCTTATCAC 40 11,649 bp 1747 3′ UTR AAAAGAGTGGTTCTGCTTTCTCYACTACCCT/CAAGGTTGCAGACTCTCTCTTATCACCCC Y 162 TTCCTCCTTCCTC 41 11,808 bp 1906 3′ UTR AGATCACCACCCCTTACAATTCCCTCTACTGTGTT/GGAAATGGTCCATTGAGTAACACCC K 163 CCATCACCTTCTCAACT 42 11,839 bp 1937 3′ UTR GAAATGGTCCATTGAGTAACACCCCCATCAC/GCTTCTCAACTGGGAAACCCCTGAAATGC S 164 TCTCAGAGCACC 43 11,885 bp 1963 3′ UTR TGAAATGCTCTCAGAGCACCTCTGA T/C GCCTGAAGAAGTTATACCTTCCTC Y 165 44 11,977 bp 2075 3′ UTR AACCATCATCTGGAAACAGTGGCCACTTTTCACTGACCTC/TTCTTCGACATCTAGTCAAC Y 166 CCACCCAATATGC 45 12,018 bp 2116 3′ UTR ATCTAGTCAACCCACCCAATATGCCACTGGGCTT/CTCGCTCCCAATTCCACCCCACCCTC Y 167 CATTACAGAGCTCACCA 46 12,136 bp 2234 3′ UTR GCCTCTCAAGGCCCTTATCTCAGCCCCTTCCTGTGGCCAT/CTTCCCTCAGTGCCCAGATG Y 168 ATTCCCTGGGTGAGGGCAGACAC 47 12,149 bp 2247 3′ UTR CAGCCCCTTCCTGTGGCCATTTCCCTCAGTGCC/TCAGATGATTCCCTGGGTGAGGGAGAC Y 169 ACTGGGGCACCCTC 48 12,198 bp 2296 3′ UTR TTCCCTGGGTGAGGGAGACACTGGGGCACCCTCAGAGGTTGG/AAGCAGGCTCCCTGCTGT R 170 CCCTGGATCCTGGACAGA 49 12,283 bp 2381 3′ UTR GGTGCAGACTTTTTGCCTTCTTGGA G/A TCCTGGGTCTCCTCTGAGAGTCTG R 171 50 12,318 bp 2416 3′ UTR TCTTGGAGTCCTGGGTCTCCTCTGAGAGTCTGGGTGGTGCTCT/CTCCTACGCCTCTAGAG Y 172 GTCTCTGTGTCCCTCA 51 12,345 bp 2443 3′ UTR TGGGTGGTGCTCTTCCTACGCCTCTAGAGGTCTCTGTGTC/TCCTCATTTTCCTTCAAAAG Y 173 CGGGCTGTGTTTCT 52 12,373 bp 2471 3′ UTR TCATTTTCCTTCAAAAGCGGGCTGT G/A TTTCTCTTCTACCTTCCAGCTCCT R 174 53 12,901 bp 2999 3′ UTR TAGATCAAGAGGCCCAGCCTGTGGCAGAACAGAGCTGCCA/GGTGGTCTCTCCATCTTCAC R 175 ACTCCCTGCTCTGCTGGGGT 54 13,001 bp 3099 3′ UTR AACATGGCTCTCAGGTGAGGGCTGAGAAGGCAGAGTGCCCCA/CGTGGGAAAGAGGAGTCG M 176 CTTCCACTGGAGAAGAGAGA 55 13,020 bp 3118 3′ UTR GCTGAGAAGGCAGAGTGCCCCAGTGGGAAAGAGGAGTCGCT/CTCCACTGGAGAAGAGAGA Y 177 GAAAGTGGAGTGTGTGGTG 56 13,108 bp 3206 3′ UTR GACTTAAGTCCTGAGACAGGCAGGGAGAGGCTGAGGCGGAC/GGAAGTTCCCGCATCCCAA S 178 GGAGGGCAGAGTGGATT 57 13,117 bp 3215 3′ UTR TGAGACAGGCAGGGAGAGGCTGAGGCGGACGAAGTTCCC/TGCATCCCAAGGAGGGCAGAG Y 179 TGGATTGTGCTTGTCC 58 13,178 bp 3276 3′ UTR GGATTGTGCTTGTCCCTGTAGGAGCCCCACCCCCCACCCC/TAGGCCACCTCTCAGAGCCT Y 180 CTGCTTGGCTGCAAAGG 59 13,224 bp 3322 3′ UTR CTCAGAGCCTCTGCTTGGCTGCAAAGGAATTCACCCC/TTACTGTAGCACTTAACCCATTC Y 181 CCTCCTATCAGGGTGG 60 13,316 bp 3414 3′ UTR TGAATTTAGAACTGTTGAAACTCCAAGTCTGGAATCAGCAA/GAAATGTATTACATTGACC R 182 AGAAAGGGATTGAATCACCCT 61 13,365 bp 3463 3′ UTR ACATTGACCAGAAAGGGATTGAATCACCCTTGGTCCAGCA/GTCTGGCCCCTGATCTGCAG R 183 CCAATGGCAGGAATCGAGGTC 62 13,562 bp 3660 3′ UTR AGGCCTCTGGGCTCCATCCACTGCCAGTTCTGGAGA/TGGAGCTCTTCACTCCTCCAGTGG W 184 TTAAGCCAGCA 63 13,605 bp 3703 3′ UTR CTCTTCACTCCTCCAGTGGTTAAGCCAGCAGGGGCAGGT/CGGGGAGGACACAGCAGTAGA Y 185 ATCAGCCAACAGCTCAT 64 13,670 bp 3768 3′ UTR CATGTTTAGACCTTGGGCAGCCAGGGAAGCC/TTACTCCTGGGGCCTCCCGGAAGCCATGG Y 186 AGAGAAC 65 13,859 bp 3857 3′ UTR GATCAAGTCCTGGCCATTTGACAGCAGCATTTAAAGGCT/CCTCCTCTACTGTTACTTGGA Y 187 AATAGCCACTTTCTCCCAAGGT 66 13,889 bp 3897 3′ UTR CTCCTCTACTGTTACTTGGAAATAGCCACT/CTTCTCCCAAGGTTTCTTATACTCT Y 188 67 13,914 bp 3922 3′ UTR GAAATAGCCACTTTCTCCCAAGGTTTCTTATACTCTG/ATGGCACATCTGACCACCAGTAG R 189 CAGGCAGAATGATGT

TABLE 7 AC006163 Frequency SNP nt position nt position* SNP chemistry allele 1 allele 2 1  6,984 bp 44884 CDSN6984 PSQ 69.6 30.4 2  7,068 bp 44968 CDSN7068 PSQ 60.8 39.2 19 10,039 bp 47939 PS SEEK IN 1 6 C565T Sequenced 55 45 21 10,108 bp 48008 CDSN C10098T Sequenced not available not available 22 10,344 bp 48244 CDSN G10343A Sequenced not available not available 23 10,363 bp (ins) 48262 CDSN 10363 AAG ins Sequenced not available not available 24 10,516 bp 48416 CDSNx2.2A10516G PSQ 47.8 52.2 25 10,521 bp 48421 CDSNx2.2C10521T PSQ 20.5 79.5 26 10,624 bp 48524 CDSNx2T10614C SNaPshot 48.9 51.1 27 10,669 bp 48569 CDSNx2.2G10669A SNaPshot 85.7 14.3 28 10,873 bp 48773 CDSN T10873C SNaPshot 32.3 67.7 29 11,020 bp 48920 SEEKIN1_3 G27A PSQ 43.8 56.2 30 11,117 bp 49017 SEEKIN1_3 A124G PSQ 98.8 1.2 31 11,138 bp 49038 SEEKIN1_3 T145G PSQ 82.8 17.2 32 11,142 bp 49042 SEEKIN1_3 G149T PSQ 100 0 33 11,145 bp 49045 SEEKIN1_3 C152T PSQ 64.3 35.7 34 11,233 bp 49133 SEEKIN1_3 G241C PSQ 47.8 52.2 35 11,260 bp 49160 SEEKIN1_3 T268C PSQ 78.9 21.1 36 11,495 bp 49395 SEEK1in3 G503A SNaPshot 68.7 31.3 37 11,605 bp (ins) 49404-49407 SEEK1in3.511INS SNaPshot 43.4 56.6 38 11,575 bp 49479 CDSN G11576T SNaPshot 32.5 67.5

TABLE 8 SNP1 SNP2 SNP19 SNP21 SNP22 SNP23 SNP24 SNP25 SNP26 SNP27 SNP28 SNP1 1 0.76 0.89 0.75 −0.88 −0.19 0.55 0.23 −0.56 0.13 −0.47 SNP2 1 0.81 0.74 −0.8 −0.3 0.45 0.18 −0.42 0.17 −0.27 −0.6 SNP19 1 0.79 −0.91 −0.26 0.56 0.19 −0.54 0.15 −0.41 −0.6 SNP21 1 −0.84 −0.3 0.43 0.16 −0.39 0.07 −0.35 −0.48 SNP22 1 0.26 −0.56 −0.18 0.52 −0.13 0.41 0.55 SNP23 1 0.55 0.23 −0.55 0.17 −0.4 −0.47 SNP24 1 0.41 −0.99 0.36 −0.71 −1 SNP25 1 −0.43 −0.27 −0.59 −0.33 SNP26 1 −0.33 0.71 1 SNP27 1 0.34 −0.33 SNP28 1 0.65 SNP29 1 SNP30 SNP31 SNP32 SNP33 SNP34 SNP35 SNP36 SNP37 SNP38 SNP29 SNP30 SNP31 SNP32 SNP33 SNP34 SNP35 SNP36 SNP37 SNP37 SNP1 −0.57 −0.1 0.04 n/a 0.46 −0.55 0 −0.92 0.55 0.55 SNP2 0.03 0.16 n/a 0.41 −0.48 0.05 −0.8 0.45 0.29 0.29 SNP19 −0.09 0.12 n/a 0.46 −0.55 0.08 −0.93 0.55 0.42 0.42 SNP21 −0.01 0.05 n/a 0.33 −0.39 0.11 −0.8 0.4 0.34 0.34 SNP22 0.08 −0.11 n/a −0.45 0.54 −0.06 0.94 −0.53 −0.41 −0.41 SNP23 −0.14 0.14 n/a 0.52 −0.5 −0.26 0.27 0.54 0.4 0.4 SNP24 −0.16 0.3 n/a 0.86 −0.96 −0.28 −0.56 1 0.71 0.71 SNP25 0.02 −0.29 n/a 0.36 −0.34 −0.25 −0.2 0.43 0.58 0.58 SNP26 0.15 −0.28 n/a −0.86 0.94 0.22 0.55 −0.98 −0.71 −0.71 SNP27 0.08 0.93 n/a 0.27 −0.31 0 −0.16 0.33 −0.36 −0.36 SNP28 0.18 0.35 n/a −0.64 0.67 0.25 0.44 −0.71 −0.97 −0.97 SNP29 0.02 −0.14 n/a −0.67 1 −0.34 0.61 −1 −0.72 −0.72 SNP30 1 0.1 n/a −0.15 0.13 −0.02 0.11 −0.13 −0.18 −0.18 SNP31 1 n/a 0.3 −0.25 −0.01 −0.13 0.27 −0.37 −0.37 SNP32 n/a n/a n/a n/a n/a n/a n/a SNP33 1 −0.82 −0.25 −0.48 0.86 0.64 0.64 SNP34 1 0.18 0.58 −0.95 −0.68 −0.68 SNP35 1 −0.1 −0.23 −0.23 −0.23 SNP36 1 −0.55 −0.44 −0.44 SNP37 1 0.72 0.72 SNP38 1 1

TABLE 9 Number of Transmissions SNP Frequency chi- p value Allele 1 Allele 2 Number Position SNP Type (allele 1) Transmissions squared (bootstrap) observed expected observed expected 1 44884 Promoter 0.79 117 3.99 0.043 235 226 53 62 2 44968 Promoter 0.69 98 1.14 0.213 157 152 67 72 19 47939 Silent 0.79 133 11.43 0.002 260 244 50 66 21 48008 leu-ser 0.74 125 10.7 0 238 221 60 77 22 48244 ser-asn 0.2 132 3.44 0.061 55 64 259 250 23 48262 ins/del (ser) 0.82 112 9.28 0 231 219 33 45 24 48416 Silent 0.59 125 18.03 0 203 180 99 122 25 48421 ser-phe 0.18 120 1.43 0.18 44 49 232 227 26 48524 silent 0.43 140 22.93 0 113 143 229 199 27 48569 silent 0.13 139 5.97 0.025 35 45 305 295 28 48773 silent 0.56 142 36.51 0 152 188 194 158 29 48920 silent 0.47 26 0.99 0.283 23 26 33 31 30 49017 ser-gly 0.96 131 11.16 0 291 299 21 13 31 49038 ser-ala 0.13 135 4.55 0.051 34 42 290 282 32 49042 gly-val 1 33 49045 ser-leu 0.59 132 9.69 0 211 193 111 129 34 49133 ser-leu 0.43 133 11 0.002 115 135 203 183 35 49160 silent 0.33 102 0.74 0.381 77 81 169 165 36 49395 silent 0.22 140 6.47 0.02 61 74 281 268 37 49404 ins/del 0.58 139 18.32 0 223 197 113 139 38 49479 3′ UTR 0.44 144 34.99 0 194 158 156 192

TABLE 10a SNP Haplotype Number A B C 1 1 1 2 2 1 1 2 19 1 1 2 21 1 1 2 22 2 2 1 23 1 1 1 24 1 1 2 25 1 2 2 26 2 2 1 27 2 2 2 28 2 2 1 30 1 1 1 31 2 2 2 33 1 1 2 34 2 2 1 35 2 2 1 36 2 2 1 37 1 1 2 38 1 1 2

TABLE 10b Code Key 1 2 A/T A T A/G A G A/C A C C/G G C G/T G T C/T C T

Claims

1. A method comprising the detection of one or more variant alleles in the corneodesmosin gene at one or more nucleotide positions selected from the group consisting of 4805 and 4875 of SEQ ID NO:1.

2. The method of claim 1 wherein the one or more variant alleles are selected from the group consisting of 4805AAG (insert), and G4875T.

3. A method for diagnosing or determining susceptibility of a subject to a corneodesmosin-mediated disease comprising the detection of one or more variant alleles in the corneodesmosin gene at one or more nucleotide positions selected from the group consisting of 3339, 3408, 4805, and 4875 of SEQ ID NO:1.

4. The method of claim 3 wherein the one or more variant alleles are selected from C3339T, C3408T, 4805AAG (insert), and G4875T.

5. The method of claim 1 or 3 wherein the corneodesmosin-mediated disease is an inflammatory disease

6. The method of claim 1 or 3 wherein the corneodesmosin-mediated disease is psoriasis.

7. The method of claim 1 or 3 wherein detection of the one or more variant alleles is carried out using a polynucleotide isolated from a biological sample selected from the group consisting of whole blood, semen, saliva, tears, skin, hair, and a buccal sample.

8. The method of claim 1 or 3 wherein detection of the one or more variant alleles is carried out using an agent capable of detecting one or more of the variant alleles.

9. The method of claim 8 wherein the agent is an anti-sense polynucleotide that is complementary to one or more of the variant alleles or to a region flanking one or more of the variant alleles.

10. The method of claim 1 or 3 wherein detection of one or more of the variant alleles is carried out using a method selected from the group consisting of direct probing, allele specific hybridization, polymerase chain reaction (PCR), allele specific amplification (ASA), and restriction fragment length polymorphism (RFLP).

11. The method of claim 1 or 3 wherein detection of one or more of the variant alleles is carried out by detecting the presence of a variant protein.

12. The method of claim 11 wherein detection of the variant protein is carried out using an antibody to an antigen of the variant protein.

13. An agent capable of detecting one or more variant alleles in the corneodesmosin gene at one or more nucleotide positions selected from the group consisting of 3339, 3408, 4805, and 4875 of SEQ ID NO:1.

14. The agent of claim 13 wherein the one or more variant alleles are selected from the group consisting of C3339T, C3408T, 4805AAG (insert), and G4875T.

15. The agent of claim 13 for use in a method for the diagnosis of or determining the susceptibility of a subject to a corneodesmosin-mediated disease.

16. The agent of claim 13 wherein the agent is:

(a) an anti-sense polynucleotide that is complementary to one or more of the variant alleles or to a region flanking one or more of the variant alleles; or
(b) an antibody to an antigen of the protein expressed from one or more of the variant corneodesmosin genes.

17. A method of screening for an agent capable of detecting one or more variant alleles in the corneodesmosin gene at one or more nucleotide positions selected from the group consisting of 3339, 3408, 4805, and 4875 of SEQ ID NO:1, wherein the method is selected from the group consisting of:

(i) a method comprising the steps of contacting a putative agent with a polynucleotide comprising one or more variant alleles in the corneodesmosin gene at one or more nucleotide positions selected from the group consisting of 3339, 3408, 4805, and 4875 of SEQ ID NO:1, and monitoring the reaction there between; and
(ii) a method comprising the steps of contacting a putative agent with a protein expressed from one or more variant alleles of the corneodesmosin gene having an alteration at one or more nucleotide positions selected from the group consisting of 3339, 3408, 4805, and 4875 of SEQ ID NO:1, and monitoring the reaction there between.

18. The method of claim 17 wherein the one or more variant alleles are selected from C3339T, C3408T, 4805AAG (insert), and G4875T.

19. The method of claim 17 further comprising:

(a) contacting the putative agent with a polynucleotide having SEQ ID NO:1, and comparing the reaction between: (i) the putative agent and the polynucleotide comprising the variant allele; and (ii) the putative agent and the polynucleotide having SEQ ID NO:1; or
(b) contacting the putative agent with a protein having SEQ ID NO:2 and comparing the reaction between: (i) the putative agent and the protein expressed from the polynucleotide comprising the variant allele; and (ii) the putative agent and the protein having SEQ ID NO:2.

20. The method of claim 17 wherein the method is carried out by contacting the putative agent with a host cell or transgenic non-human animal comprising the polynucleotide comprising the variant allele or the polypeptide expressed from the polynucleotide comprising the variant allele.

21. A kit for the detection of a variant allele in the corneodesmosin gene comprising an agent capable of detecting one or more variant alleles in the corneodesmosin gene at one or more nucleotide positions selected from the group consisting of 3339, 3408, 4805, and 4875 of SEQ ID NO:1.

22. The kit of claim 21 wherein the one or more variant alleles are selected from C3339T, C3408T, 4805AAG (insert), and G4875T.

23. The kit of claim 21 wherein the agent is selected from the group consisting of:

(a) an anti-sense polynucleotide that is complementary to one or more of the variant alleles or to a region flanking one or more of the variant alleles;
(b) an antibody to an antigen of the protein expressed from the corneodesmosin gene comprising one or more of the variant alleles; and
(c) a restriction enzyme that is capable of detecting the presence of the polynucleotide comprising one or more of the variant alleles.

24. The kit of claim 21 further comprising means for the detection of a reaction selected from the group consisting of:

(a) nucleotide detection means;
(b) labeled secondary antibodies; and
(c) size detection means.

25. The kit of claim 21 for use in a method for the diagnosis of or determining the susceptibility of a subject to a corneodesmosin-mediated disease, and further comprising a key correlating the presence of the one or more variant alleles with the presence of, or susceptibility to, corneodesmosin-mediated disease.

Patent History
Publication number: 20080193939
Type: Application
Filed: Nov 29, 2007
Publication Date: Aug 14, 2008
Applicant: YORK PHARMA (R & D) LIMITED (London)
Inventors: Mark Olavesen (Abingdon), Nick Lench (Abingdon), Maxine Allen (Abingdon), Rachid Tazi-Ahnini (Sheffield)
Application Number: 11/947,600
Classifications
Current U.S. Class: 435/6
International Classification: C12Q 1/68 (20060101);