US20100196895A1 - Method for determination of onset risk of glaucoma - Google Patents

Method for determination of onset risk of glaucoma Download PDF

Info

Publication number
US20100196895A1
US20100196895A1 US12/596,258 US59625808A US2010196895A1 US 20100196895 A1 US20100196895 A1 US 20100196895A1 US 59625808 A US59625808 A US 59625808A US 2010196895 A1 US2010196895 A1 US 2010196895A1
Authority
US
United States
Prior art keywords
seq
allele
risk
glaucoma
single nucleotide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/596,258
Other languages
English (en)
Inventor
Shigeru Kinoshita
Kei Tashiro
Masakazu Nakano
Tomohito Yagi
Kazuhiko Mori
Yoko Ikeda
Takazumi Taniguchi
Masaaki Kageyama
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Santen Pharmaceutical Co Ltd
Original Assignee
Santen Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Santen Pharmaceutical Co Ltd filed Critical Santen Pharmaceutical Co Ltd
Assigned to SANTEN PHARMACEUTICAL CO., LTD., SHIGERU KINOSHITA, KEI TASHIRO reassignment SANTEN PHARMACEUTICAL CO., LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NAKANO, MASAKAZU, TASHIRO, KEI, KINOSHITA, SHIGERU, IKEDA, YOKO, KAGEYAMA, MASAAKI, MORI, KAZUHIKO, TANIGUCHI, TAKAZUMI, YAGI, TOMOHITO
Publication of US20100196895A1 publication Critical patent/US20100196895A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • the present invention relates to a method of detecting the presence of a single nucleotide polymorphism associated with the onset of glaucoma, or a single nucleotide polymorphism with a high onset risk of glaucoma, and a kit used in the detection method.
  • Glaucoma is a disease which causes a characteristic optic nerve cupping and an impairment in a visual field by retinal ganglion cell death.
  • An elevation in an intraocular pressure is considered to be a major cause for the nerve cupping and the impairment in the visual field in glaucoma.
  • the basic treatment for glaucoma is to maintain an intraocular pressure at a low level, and it is necessary to consider the causes for a high intraocular pressure in order to maintain a low intraocular pressure. Therefore, in the diagnosis of glaucoma, it is important to classify the types of glaucoma in accordance with the levels of intraocular pressures and causes therefor. As the causes for an elevation in an intraocular pressure, the presence or absence of closure of angle which is a major drainage pathway for an aqueous humor filling an eye is important. From these viewpoints, the primary glaucoma is roughly classified into the two groups of closed-angle glaucoma accompanying angle closure and open-angle glaucoma without accompanying angle closure.
  • the open-angle glaucoma is classified into open-angle glaucoma, in a narrow sense, accompanying an elevation in an intraocular pressure, i.e. primary open-angle glaucoma, and normal tension glaucoma in which an intraocular pressure is held within a normal range.
  • Non-Patent Publication 1).
  • all the genetic causes of glaucoma cannot be explained only by these genes, and the presence of unknown glaucoma-related genes is expected.
  • a single nucleotide polymorphism means that a substitution mutation in which a single base is changed into another base is found in base sequences of the genome of an individual, and the mutation exists in a certain frequency, generally a frequency of about 1% or more, in the population of an organism species.
  • a single nucleotide polymorphism exists at intron or exon on genes, or any of the regions of the genome other than these.
  • Patent Publication 1 Japanese Patent-Laid Open No. 2000-306165
  • Patent Publication 1 Rezaie T and eleven others, Science, 2002, 295(5557), 1077-1079.
  • an intraocular pressure or an ocular fundus photograph is used as a simple examination for glaucoma; however, these examinations do not necessarily lead to a definite diagnosis for glaucoma.
  • visual field examinations are performed; however, there are some disadvantages that the examination is carried out for a long period of time, causing burdens on patients, and that one must be accustomed to the examination, so that initial examination results have low reliability.
  • the involvement of hereditary causes is strongly suspected in the onset of glaucoma, but critical responsible genes are not identified.
  • the involvement of a single gene to the disease cannot be explained by a mutation or polymorphism, it is considered that there are numerous mutations or polymorphisms of a gene of which involvement to glaucoma is relatively moderate, and the involvement of hereditary causes to the onset of glaucoma can be explained by a combined action of each of these mutations or polymorphisms.
  • the inventors have remarked on a polymorphism on the genome, especially a single nucleotide polymorphism, in order to find a gene associated with glaucoma.
  • the polymorphisms can be applied to screening of whether or not a visual field examination is required, in an early stage of glaucoma which is difficult to be detected by the simple determination of glaucoma, i.e. a method such as a measurement of intraocular pressure or an ocular fundus photograph, which is available to be diagnosed only by carrying out the visual field examination.
  • a sample donor can take a preventive measure for the onset of glaucoma by knowing the onset risk of glaucoma, and in addition, a necessary measure for preventing visual constriction such as a definite diagnosis and an initiation of treatment at an early stage according to a precision examination can be taken; therefore, it is important to find a polymorphism involved in the onset of glaucoma.
  • An object of the present invention is to provide a method of detecting a single nucleotide polymorphism involved in the onset of glaucoma, thereby predicting an onset risk of glaucoma, and a kit used in the detection method.
  • the present inventors have found a single nucleotide polymorphism associated with the onset of glaucoma by a comprehensive analysis of known polymorphic sites existing on the genome (autosome) in glaucoma patients and non-patients without a family history of glaucoma, and further found an allele identified in a high frequency in glaucoma patients and an opposite allele thereof, and a genotype identified in a high frequency in glaucoma patients, which is a combination of each of the alleles, in the single nucleotide polymorphism.
  • the present inventors have found that a determination on whether or not a sample donor is a person who is more likely to suffer from the onset of glaucoma can be made at an even higher precision by performing the determination in a combination of these plural single nucleotide polymorphisms associated with the onset of glaucoma.
  • the present invention has been perfected thereby.
  • the present invention relates to:
  • step A detecting in vitro an allele and/or a genotype of a single nucleotide polymorphism which is located on a 31st base of a base sequence, in a sample from a subject, wherein the base sequence is at least one base sequence selected from the group consisting of base sequences shown in SEQ ID NOs: 203 to 514 or a complementary sequence thereto (step A), and
  • step B comparing the allele and/or the genotype detected in the step A with at least one of an allele and/or a genotype, containing a high-risk allele, in the base sequences shown in SEQ ID NOs: 203 to 514 (step B),
  • nucleic acid molecule comprises at least one base sequence selected from the group consisting of base sequences shown in SEQ ID NOs: 203 to 514 or a complementary sequence thereto (step C1), or
  • step C2 detecting in vitro, in a sample from a subject, an allele and/or a genotype of a single nucleotide polymorphism, using a nucleic acid molecule comprising a base sequence containing at least one base sequence selected from the group consisting of base sequences shown in SEQ ID NOs: 515 to 694 or a complementary sequence thereto (step C2), and
  • step D) comparing the allele and/or the genotype detected in the step C1 or C2 with at least one nucleic acid molecule comprising an allele and/or a genotype, containing a high-risk allele, in the base sequences shown in the SEQ ID NOs: 203 to 514 (step D),
  • nucleic acid molecule comprising at least one base sequence selected from the group consisting of base sequences shown in SEQ ID NOs: 203 to 514 or a complementary sequence thereto, or a partial sequence thereof, wherein the nucleic acid molecule comprises a single nucleotide polymorphism which is located on a 31st base of a base sequence, and/or
  • nucleic acid molecule comprising a base sequence containing at least one base sequence selected from the group consisting of base sequences shown in SEQ ID NOs: 515 to 694 or a complementary sequence thereto,
  • step E detecting in vitro an allele and/or a genotype of a single nucleotide polymorphism which is located on a 31st base of a base sequence, in a sample from a subject, wherein the base sequence is at least one base sequence selected from the group consisting of base sequences shown in SEQ ID NOs: 203 to 514 or a complementary sequence thereto (step E), and
  • step F comparing the allele and/or the genotype detected in the step E with at least one of an allele and/or a genotype, containing a high-risk allele, in the base sequences shown in SEQ ID NOs: 203 to 514 (step F), wherein the subject is diagnosed as glaucoma in a case where the allele detected in the step E is the high-risk allele, or
  • the presence or the absence of the onset risk of glaucoma in a sample donor can be determined, and further the level of the risk can be predicted, by analyzing an allele or a genotype of a single nucleotide polymorphism in the present invention contained in a nucleic acid molecule derived from the genome existing in a sample.
  • a sample donor can be provided with a preventive measure for glaucoma, or can receive appropriate treatments, on the basis of this risk.
  • a sample donor who is suspected of glaucoma, having an allele or a genotype containing a single nucleotide polymorphism in the genome that is identified in a high frequency in glaucoma patients can be given a detailed examination on whether or not the donor is with early glaucoma, which is difficult to be determined sufficiently by an intraocular pressure or an ocular fundus photograph, and can be started with a treatment at an early stage in a case where the donor is diagnosed as glaucoma.
  • the present invention is a method of determining the presence or the absence of a glaucoma risk, including the step of detecting in vitro an allele and/or a genotype having at least one single nucleotide polymorphism using at least one single nucleotide polymorphism (hereinafter may be referred to as SNP) contained in a base sequence selected from the group consisting of specified base sequences or a complementary sequence thereto, wherein the method of determining the presence or the absence of a glaucoma risk further includes the step of:
  • a great feature of the present invention resides in that a single nucleotide polymorphism associated with the onset of glaucoma is found, further that in the single nucleotide polymorphism, an allele identified in a high frequency in glaucoma patients and an opposite allele thereof, and a genotype, which is a combination of each of the alleles identified in a high frequency in glaucoma patients are found, and used.
  • polymorphism refers to a matter that a diversity is found in sequences of a specified location on the genome in a certain organism species, and a site at which the polymorphism exists (hereinafter also referred to as polymorphic site) refers to a site on the genome that a single nucleotide polymorphism is found.
  • the allele as used herein refers to each of types having a different base from each other that can be taken in a certain polymorphic site.
  • the genotype as used herein refers to a combination of opposite alleles in a certain polymorphic site. Further, in a certain polymorphic site, there are three types for a genotype which is a combination of opposite alleles, wherein a combination of the same alleles is referred to as a homozygote, and a combination of different alleles is referred to as a heterozygote.
  • the opposite allele as used herein refers to another allele corresponding to a specified allele among the alleles constituting a certain single nucleotide polymorphism.
  • the single nucleotide polymorphism associated with glaucoma refers to a single nucleotide polymorphism associated with the onset of glaucoma or a single nucleotide polymorphism associated with the progression of glaucoma.
  • the single nucleotide polymorphism associated with the onset of glaucoma refers to a single nucleotide polymorphism in which each allele or each genotype frequency in the single nucleotide polymorphism significantly differs in a statistical analysis at a given p-value between glaucoma patients and non-patients; and the single nucleotide polymorphism associated with the progression of glaucoma refers to a single nucleotide polymorphism in which each allele or each genotype frequency in the single nucleotide polymorphism significantly differs in a statistical analysis at a given p-value between the progressive glaucoma cases and the nonprogressive glaucoma cases.
  • the high-risk allele refers to an allele having a higher frequency in a glaucoma patient group than that of a non-patient group among each of the alleles of the single nucleotide polymorphism associated with glaucoma.
  • the low-risk allele refers to an allele opposite to the high-risk allele in a certain polymorphic site.
  • homozygote and the heterozygote of a genotype are defined in the same manner as in the high-risk allele and the low-risk allele.
  • a combination of high-risk alleles or low-risk alleles themselves is referred to a homozygote
  • a combination of a high-risk allele and a low-risk allele is referred to as a heterozygote.
  • an allele model An embodiment where allele frequencies of the glaucoma patient group and the non-patient group are statistically compared is referred to as an allele model, and an embodiment where genotype frequencies thereof are compared is referred to as a genotype model.
  • genotype models There are a dominant genetic model and a recessive genetic model in the genotype models, wherein the former means an embodiment where both a homozygote of high-risk alleles and a heterozygote are involved with the onset risk, and the latter means an embodiment where a homozygote of a high-risk allele is involved with the onset risk.
  • the glaucoma risk refers to a risk concerning glaucoma.
  • the onset risk of glaucoma refers to a possibility of the future onset of glaucoma determined by susceptibility to a disease.
  • the prediction of a risk refers to a determination of the presence or the absence of a future risk at the present stage, or determining the level of a future risk at the present stage.
  • the glaucoma as used herein means preferably open-angle glaucoma (OAG) or normal tension glaucoma (NTG), and the open-angle glaucoma, when used without specifying otherwise, means primary open-angle glaucoma (POAG) in a narrow sense, without embracing normal tension glaucoma.
  • OAG open-angle glaucoma
  • NVG normal tension glaucoma
  • POAG primary open-angle glaucoma
  • a candidate single nucleotide polymorphism is selected by the steps including extracting a total DNA from blood of each of glaucoma patients diagnosed as primary open-angle glaucoma or normal tension glaucoma and non-patients diagnosed as not being with glaucoma and determined to have no family history of glaucoma according to a medical interview (also referred to as control individuals), and comparing allele or genotype frequencies of individual single nucleotide polymorphisms in the glaucoma patients and the non-patients using known single nucleotide polymorphisms of about 500,000 on the human genome as an index.
  • the allele or genotype frequencies of individual single nucleotide polymorphisms for the single nucleotide polymorphisms that are selected as candidates are obtained for glaucoma patients and non-patients that are different from the sample groups mentioned above.
  • a single nucleotide polymorphism of which difference in frequencies is recognized with high statistical significance is found.
  • a group composed of the glaucoma patients is referred to as a glaucoma patient group
  • a group composed of the non-patients is referred to as a non-patient group.
  • the determination of the presence or the absence of the onset risk of glaucoma, and the prediction of the level of an onset risk can be enabled.
  • a single nucleotide polymorphism associated with glaucoma disclosed in the present invention can be identified according to a method given below.
  • a total DNA is extracted from blood of each of patients diagnosed as glaucoma and non-patients determined to have no family history of glaucoma.
  • the total DNA in blood can be extracted by any known methods; for example, a DNA can be extracted by binding a DNA eluted by lysing cells to surfaces of magnetic beads coated with silica, and separating and collecting the DNA utilizing a magnetic force.
  • the kind of a base in a single nucleotide polymorphism in the extracted DNA sample i.e. an allele having a single nucleotide polymorphism can be identified by any methods, including, for example, a method using an immobilized probe described later, or the like.
  • a probe used in the detection can be designed on the basis of the sequence information of a single nucleotide polymorphism of interest and surrounding sequences thereof When the probe is designed, the sequence information obtained from the database for known single nucleotide polymorphisms such as dbSNP can be used as a reference.
  • the detection can be made with either a probe complementary to a sense strand of the genome, or a probe complementary to an antisense strand.
  • a kit in which probes capable of detecting single nucleotide polymorphisms existing on the human genome are immobilized in large amounts, thereby making it possible to determine alleles of numerous single nucleotide polymorphisms in a single operation is commercially available, and whereby an allele in a sample can be efficiently determined using the kit.
  • Many of the kits also have the constitution that the alleles that are opposite to each other contained in one sample are detected in a single operation, so that a genotype can be determined.
  • the single nucleotide polymorphism associated with glaucoma can be determined by previously identifying an allele existing on DNA from glaucoma patients and non-patients according to the method as mentioned above, statistically comparing each of the allele frequencies and the genotype frequencies in a glaucoma patient group against a non-patient group, and determining whether or not a difference that a p-value is below the significance level as defined by a given standard is caused in at least one of the allele frequencies and the genotype frequencies.
  • the allele frequencies or genotype frequencies for these factors in the glaucoma patient group and the non-patient group are compared to determine whether any of the alleles or genotypes are identified in a high frequency in the glaucoma patient group.
  • a chi-square test can be used.
  • Type I error caused by multiple comparisons can be corrected by a known correction method, for example, Bonferroni method.
  • a significance level can be obtained by dividing a p-value of 5 ⁇ 10 ⁇ 2 by the number of multiple comparisons, i.e. the number of polymorphisms to be compared in the chi-square test.
  • a single nucleotide polymorphism below the significance level determined in the manner described above can be selected as a more preferred single nucleotide polymorphism, and a method used in other known multiple corrections, for example, an FDR method or a permutation method may also be used in the selection of a preferred single nucleotide polymorphism.
  • a known multiple correction method such as Bonferroni correction is a method presupposing that the phenomenon of carrying out multiple analyses is completely independent; on the other hand, there are some cases where the phenomenon is not completely independent because linkage disequilibrium is found in a single nucleotide polymorphism as described later.
  • a significance level can be set at any appropriate levels within the range that would not be below the significance level according to the Bonferroni correction.
  • the significance level in a case where about 500,000 single nucleotide polymorphisms are analyzed repeatedly of 5 ⁇ 10 ⁇ 2 is used, more preferably 1 ⁇ 10 ⁇ 2 , even more preferably 1 ⁇ 10 ⁇ 3 , even more preferably 1 ⁇ 10 ⁇ 4 , even more preferably 3 ⁇ 10 ⁇ 5 , and even more preferably 1 ⁇ 10 ⁇ 5 .
  • the adjustment of the significance level as described above is useful from the fact that it is confirmed that a single nucleotide polymorphism identified to be associated with glaucoma in the present invention exists continuously in a certain region on the genome.
  • a method of maintaining the statistical power while lowering type I error includes a method of performing a single nucleotide polymorphism analysis in two divided steps (Skol A. D. et al., Nature Genetics. 2006: 38: 209-213).
  • a single nucleotide polymorphism analysis is carried out for a fixed number of samples, firstly, analysis of enormous single nucleotide polymorphisms over the whole genome for a part of samples thereof is carried out as primary analysis, and secondly, analysis of single nucleotide polymorphisms narrowed down in the first analysis to some degree is carried out for the remainder samples as secondary analysis.
  • a single nucleotide polymorphism may be selected so as to have a relatively low p-value, for example, 0.05; preferably, a single nucleotide polymorphism serving as a candidate in the first analysis may be selected at a given significance level, and the selected single nucleotide polymorphism may be further analyzed using another sample.
  • the results of the first analysis and the secondary analysis are not individually statistically analyzed but these results are combined and analyzed.
  • the two analytical results can be combined by a known method of meta-analysis, for example, Mantel-Haenszel method (Mantel N et al., Journal of the National Cancer Institute 1959: 22: 719-748).
  • Mantel-Haenszel method Mantel N et al., Journal of the National Cancer Institute 1959: 22: 719-748.
  • the significance level for the selection of a single nucleotide polymorphism in individual analysis is not needed to be at the level of Bonferroni correction, and the significance level may be set by taking narrowing-down efficiency or the like into consideration.
  • Mantel-Haenszel method refers to a method of combining analytical results by weighting the results obtained by multiple analyses when a chi-square test or the like is carried out.
  • a statistical parameter combined by Mantel-Haenszel method includes, in addition to the p-value, an odds ratio described later or the like.
  • a single nucleotide polymorphism for the detection of the allele or genotype associated with glaucoma is preferably a single nucleotide polymorphism having a p-value of 1 ⁇ 10 ⁇ 3 or less, more preferably a single nucleotide polymorphism having a p-value of 3 ⁇ 10 ⁇ 4 or less, even more preferably a single nucleotide polymorphism having a p-value of 1 ⁇ 10 ⁇ 4 or less, and even more preferably a single nucleotide polymorphism having a p-value of 3 ⁇ 10 ⁇ 5 or less, when the single nucleotide polymorphism for the detection is based on the results obtained in a single analysis using, for example, a microarray in which about 500,000 single nucleotide polymorphisms are detected in a single operation.
  • the single nucleotide polymorphism for the detection is preferably a single nucleotide polymorphism having a p-value of 1 ⁇ 10 ⁇ 2 or less, more preferably a single nucleotide polymorphism having a p-value of 3 ⁇ 10 ⁇ 3 or less, even more preferably a single nucleotide polymorphism having a p-value of 1 ⁇ 10 ⁇ 4 or less, and even more preferably a single nucleotide polymorphism having a p-value of 3 ⁇ 10 ⁇ 4 or less.
  • a sufficient number of single nucleotide polymorphisms are analyzed, in order to obtain highly reliable results upon analysis.
  • a polymorphic site having a low determination rate of each single nucleotide polymorphism to the whole sample i.e. a low call rate
  • the analysis is performed using a single nucleotide polymorphism having a sufficiently high call rate.
  • the call rate that serves as a standard of accepting or rejecting a single nucleotide polymorphism for example, it is preferable that a single nucleotide polymorphism showing a call rate of preferably 70%, more preferably 75%, even more preferably 80%, even more preferably 85%, and even more preferably 90% or more is employed.
  • Hardy-Weinberg's equilibrium is established in the general population, analysis of the Hardy-Weinberg's equilibrium is used for the purpose of detecting errors of genotype determination of a sample.
  • the Hardy-Weinberg's equilibrium is established as a whole, when a certain genotype is unevenly distributed in a disease group or a control group in a certain gene locus, for example, there are some cases where a certain genotype has a predominant influence on a disease, or the like; therefore, said analysis can be omitted, in a case where a search for disease-associated genes is carried out.
  • the minor allele frequency refers to an allele frequency with a lower frequency of the frequencies of two alleles in a case where single nucleotide polymorphisms are contained in two alleles. It is possible that a threshold thereof is arbitrarily set. As mentioned above, it is preferable that a single nucleotide polymorphism having a minor allele frequency of below 1% is rejected, because the concept of a single nucleotide polymorphism is in that the single nucleotide polymorphism has a minor allele frequency exceeding about 1%. On the other hand, there is a possibility that an allele having a very high or very low allele frequency in a disease group has a predominant influence on a disease.
  • polymorphisms of which relative involvement to a disease is relatively low are multiply involved in search of polymorphisms causative of multi-factorial diseases; therefore, for the purpose of searching the polymorphisms as mentioned above, an analysis excluding a frequency of a certain level or lower, for example, a minor allele of less than 5% can be a preferred means.
  • search polymorphisms that have predominant influences on a disease it is effective not to reject the polymorphisms of the minor allele frequency.
  • the information such as a location on the genome at which a single nucleotide polymorphism exists, the sequence information, a gene in which a single nucleotide polymorphism exists or a gene existing in the neighborhood, discrimination of intron or exon or a function thereof in a case where the single nucleotide polymorphism exists on the gene, and a homologous gene in other organism species can be obtained, by referring to the database of known sequences such as GenBank, or the database of known single nucleotide polymorphisms such as dbSNP, whereby a nucleic acid molecule used in the present invention is obtained, on the basis of the information, and a probe or the like used in the present invention can be designed.
  • a high-risk allele refers to an allele having a higher frequency in a glaucoma patient group than that of a non-patient group among each of the alleles of single nucleotide polymorphisms associated with glaucoma
  • the low-risk allele refers to an allele opposite to a high-risk allele in a certain polymorphic site.
  • the determination of the presence or the absence of an onset risk can be carried out according to an allele or a genotype.
  • the presence of the onset risk is determined for the single nucleotide polymorphism because of having a high-risk allele.
  • the onset risk is determined by taking into consideration whether the high-risk allele complies with a dominant genetic model, or with a recessive genetic model.
  • the frequency of a homozygote of the high-risk allele and a heterozygote is significantly high in a glaucoma patient group as compared to that of a non-patient group, it is said that these genotypes comply with a dominant genetic model.
  • the presence of an onset risk is determined for the single nucleotide polymorphism in a case where the genotype is a homozygote of the high-risk allele or a heterozygote, when the high-risk allele complies with a dominant genetic model.
  • the frequency of a homozygote of the high-risk allele is significantly high in a glaucoma patient group as compared to that of a non-patient group, it is said that these genotypes comply with a recessive genetic model.
  • the presence of an onset risk is determined for the single nucleotide polymorphism in a case where the genotype is a homozygote of the high-risk allele, when the high-risk allele complies with a recessive genetic model.
  • the determination of the presence or the absence of an onset risk can be also carried out according to a low-risk allele.
  • the low-risk allele is an allele opposite to a high-risk allele, i.e. an allele identified in a high frequency in a non-patient group.
  • the presence of an onset risk is determined for the single nucleotide polymorphism because of not having a low-risk allele.
  • an onset risk is determined by taking into consideration whether the low-risk allele complies with a dominant genetic model, or with a recessive genetic model.
  • a certain polymorphic site when the frequency of a homozygote of the low-risk allele and a heterozygote is significantly high in a non-patient group as compared to that of a glaucoma patient group, it is said that these genotypes comply with a dominant genetic model.
  • the presence of an onset risk is determined for the single nucleotide polymorphism in a case where the genotype is not a homozygote of the low-risk allele or a heterozygote, when the low-risk allele complies with a dominant genetic model.
  • the frequency of a homozygote of the low-risk allele is significantly high in a non-patient group as compared to that of a glaucoma patient group, it is said that these genotypes comply with a recessive genetic model.
  • the presence of an onset risk is determined for the single nucleotide polymorphism in a case where the genotype is not a homozygote of the low-risk allele, when the low-risk allele complies with a recessive genetic model.
  • the same method as in a method where a p-value judged to be significant is obtained can be used.
  • any of these methods may be used, and preferably, the same method as in a method where the lowest p-value is calculated is used.
  • a relative risk or an odds ratio is used as an index of an extent of the strength of the association that exists between one allele or genotype and the presence or the absence of a disease.
  • the relative risk refers to a ratio of an incidence rate in a group with a risk factor to an incidence rate in a group without a risk factor.
  • the odds ratio generally refers to a ratio obtained by dividing odds, which is a ratio of a proportion of individuals with a risk factor to a proportion of individuals without a risk factor in a patient group, by odds obtained in a non-patient group in the same manner, which is in many cases used in a case-control study as in the present invention.
  • the odds ratio in the present invention is determined on the basis of the allele frequency or the genotype frequency.
  • the odds ratio of a single nucleotide polymorphism associated with the onset refers to a value obtained by calculating a quotient obtained in a ratio of an allele or genotype frequency to another allele or genotype frequency in a glaucoma patient group, over a ratio of frequencies obtained in the same manner in a non-patient group.
  • an extent to which an onset risk of glaucoma increases can be predicted by comparing a case of having a certain allele or genotype to a case of having an allele or genotype other than the above, using these indices.
  • an odds ratio of a certain allele in a certain single nucleotide polymorphism is greater than 1, the allele is an allele found in a high frequency in a glaucoma patient group, in which the larger the odds ratio, the higher the onset risk of glaucoma for a sample donor having the allele.
  • an odds ratio of an allele is less than 1, the allele is an opposite allele of the allele that is identified in a high frequency in a disease, in which the smaller the odds ratio, the lower the onset risk of glaucoma for a sample donor having the allele.
  • the risk of a disease can also be predicted in the same manner for a genotype.
  • the value of the odds ratio would be always greater than 1 by obtaining an odds ratio based on the high-risk allele.
  • the risk prediction in a combination of plural single nucleotide polymorphisms is facilitated by defining so that the odds ratio is greater than 1 when having the high-risk allele as mentioned above.
  • a homozygote of the high-risk allele and a heterozygote becomes a risk factor when the high-risk allele complies with a dominant genetic model
  • a homozygote of the high-risk allele becomes a risk factor when the high-risk allele complies with a recessive genetic model
  • the odds ratio may be obtained by obtaining the sum of the homozygote frequency of the high-risk allele and the heterozygote frequency in a glaucoma patient group, and calculating a quotient obtained in a ratio of the above sum to the homozygote frequency of the low-risk allele, over a ratio of frequencies obtained in the same manner in a non-patient group.
  • the reliability of a single nucleotide polymorphism used in the prediction of a risk can be confirmed with an odds ratio.
  • the meaning for the prediction of a risk reverses in a case where the odds ratio is 1 or more and a case where the odds ratio is 1 or less. Therefore, in a case where a calculated 95% confidence interval of the odds ratio includes 1, it cannot be said that the reliability for the prediction of a risk for the odds ratio as mentioned above would be high.
  • the odds ratio is obtained by taking into consideration whether the high-risk allele complies with a dominant genetic model, or with a recessive genetic model, in the same manner as that alone.
  • the odds ratio of combined two or more single nucleotide polymorphisms can be calculated according to the following formula:
  • RGd1 comb a frequency at which at least one genotype is a homozygote of a high-risk allele or a heterozygote, in a glaucoma patient group;
  • the odds ratio of combined two or more single nucleotide polymorphisms can be calculated according to the following formula:
  • the odds ratio may be obtained by calculating a quotient obtained in a ratio of the frequency at which any of the two single nucleotide polymorphisms are a homozygote of a high-risk allele in a glaucoma patient group to the frequency at which both the two single nucleotide polymorphisms are a homozygote of a low-risk allele in the glaucoma patient group, over a ratio of frequencies of those obtained in the same manner in a non-patient group.
  • the odds ratio for a combination of single nucleotide polymorphisms can also be calculated by combining single nucleotide polymorphisms having different genetic forms.
  • the independent variables ⁇ used in analysis can be previously narrowed down using a stepwise method or the like.
  • the stepwise method refers to a method for selecting independent variables ⁇ so as to maximize the regression coefficients by adding an optional independent variable ⁇ . In other words, it means that after the regression coefficient is maximized by adding an arbitrary independent variable ⁇ , the same outcome is obtained even if another independent variable ⁇ is further added.
  • this sample donor is determined to be a glaucoma patient.
  • a given threshold for example, 0.5
  • this sample donor is determined to be a glaucoma patient.
  • the determination results are collated with the matter of whether the sample donor having a single nucleotide polymorphism was actually the glaucoma patient.
  • an improvement in a concordance proportion is confirmed, whereby the precision improvement by the combination can be confirmed.
  • the LD block can be determined by analyzing single nucleotide polymorphisms which exist in the region as many as possible by the method mentioned above, and applying an algorithm to determine an LD block, for example, an EM algorithm.
  • the LD block can be considered as an LD block associated with glaucoma.
  • Genome Browser provided on the internet web sites by California University at Santa Cruz, or the like can be consulted for a known LD block.
  • a single nucleotide polymorphism that belongs to an LD block associated with glaucoma is linked to a single nucleotide polymorphism associated with glaucoma identified according to the experiments of the present inventors, it can be considered that the single nucleotide polymorphism that belongs to an LD block associated with glaucoma also associates with glaucoma in the same manner; therefore, the single nucleotide polymorphism is used in the prediction of an onset risk or progressive risk of glaucoma.
  • a silent single nucleotide polymorphism refers to one without accompanying a change in an amino acid sequence, among those in which a single nucleotide polymorphism is identified in a coding region.
  • a genomic single nucleotide polymorphism refers to one in which a single nucleotide polymorphism exists in a region not encoding the gene on the genome.
  • a regulatory polymorphism refers to a single nucleotide polymorphism existing in a site that is thought to be involved in the transcriptional regulation.
  • a single nucleotide polymorphism may exist in any location on the genome, any cases of which can be associated with a disease. In a case where a single nucleotide polymorphism exists in the intron or a non-coding region, there may be some cases where the single nucleotide polymorphism may influence a gene expression control, or splicing that takes place after the gene transcription or stability of mRNA.
  • a codon corresponding to a certain amino acid may be changed to a codon corresponding to a different amino acid, or may undergo a change, for example, a change to a termination codon, or the like, which may lead to a change in the structure of a protein encoded thereby. Changes in expression levels or functions of genes by these changes consequently lead to changes in expression levels or functions of proteins encoded by the genes, which can be causes for various diseases.
  • a single nucleotide polymorphism other than the silent single nucleotide polymorphism even when the single nucleotide polymorphism itself is not a direct cause for glaucoma but links to a polymorphism which is the true cause for glaucoma existing in the surrounding, the association of these single nucleotide polymorphisms and glaucoma may be found in some cases.
  • a polymorphism which is causative of glaucoma can be found by re-sequencing the surrounding of the single nucleotide polymorphism in the present invention.
  • these single nucleotide polymorphisms can be also used for the purpose of predicting an onset risk of glaucoma, regardless of whether or not these would be the true causes for the disease.
  • the nucleic acid molecule comprising a single nucleotide polymorphism associated with glaucoma or the nucleic acid molecule having a sequence complementary to the nucleic acid molecule can be used as a marker for determining the level of the onset risk of glaucoma. Further, these nucleic acid molecules can be used as a probe for detecting an allele or an opposite allele thereof identified in a high frequency in glaucoma patients, or determining a genotype, in the single nucleotide polymorphism. In addition, in a case where the single nucleotide polymorphism exists on exon or in the neighborhood thereof, these nucleic acid molecules can be used in the detection of transcripts of genes.
  • the nucleic acid molecule constituting the genome of an eukaryote is constituted by double strands of a sense strand and an antisense strand complementary to the sense strand.
  • the single nucleotide polymorphism also exists on the sense strand and the antisense strand, and the nucleic acid molecule of the present invention embraces both of these strands because the detection of a single nucleotide polymorphism of both the strands is equally significant.
  • the nucleic acid molecule is a nucleic acid molecule comprising a sequence from a next base of a known single nucleotide polymorphism on an upstream side of the sense strand to a base before a known single nucleotide polymorphism on a downstream side, or a nucleic acid molecule comprising a sequence complementary thereto,
  • the nucleic acid molecule is a nucleic acid molecule comprising a full length of the gene on the genome including the single nucleotide polymorphism, a nucleic acid molecule comprising a sequence complementary thereto, and a nucleic acid molecule containing a complementary DNA (cDNA) molecule comprising the single nucleotide polymorphism or a sequence complementary thereto,
  • cDNA complementary DNA
  • the nucleic acid molecule is a nucleic acid molecule comprising a sequence from a next base of a known single nucleotide polymorphism on an upstream side of the sense strand to a full length of the gene existing downstream of a promoter region in which the single nucleotide polymorphism exists, or a nucleic acid molecule comprising a sequence complementary thereto.
  • the nucleic acid molecule in the present invention is not limited whether it is a deoxyribonucleic acid, a ribonucleic acid, or a peptide nucleic acid, and a nucleic acid molecule comprising a mixed sequence thereof is also embraced in the present invention.
  • a ribonucleic acid is used in the nucleic acid molecule in the present invention
  • thymine may be read as uracil.
  • these nucleic acid molecules may be subjected to chemical modifications as occasion demands, within the range that would not impair a function to be used in the present invention.
  • the function refers to a function of accomplishing the purpose of using the nucleic acid molecule.
  • the nucleic acid molecule in the present invention can be synthesized by a known method, for example, a phosphoramidite method, on the basis of the sequence information disclosed herein, or the sequence information obtained by searching the information disclosed herein with the database.
  • the nucleic acid molecule can be synthesized using a commercially available DNA synthesizer.
  • the nucleic acid molecule in the present invention can be obtained from a sample comprising DNA from human according to a known method such as a PCR method, or in some nucleic acid molecules, can be obtained from a sample containing RNA from human according to a known method such as an RT-PCR method.
  • primers that are necessary for the obtainment can design the primers on the basis of the sequence information disclosed herein, or the sequence information that can be searched from ID of the database disclosed herein.
  • primers having about 10 to about 30 bases that have sequences homologous to a part of the sequences of the nucleic acid molecule of interest can be used, and in a case where an RT-PCR method is used, the nucleic acid molecule can be obtained by carrying out reverse transcription reaction using an oligo dT primer, or a random hexamer, or the like to prepare cDNA, and amplifying a sequence of interest in the cDNA by the PCR method mentioned above.
  • the nucleic acid molecule has a length of preferably from 16 to 55 bases, and more preferably from 23 to 27 bases or 47 to 53 bases. It is preferable that the nucleic acid molecule is a nucleic acid molecule containing the polymorphic site mentioned above and a surrounding sequence thereof, or a sequence complementary thereto.
  • the nucleic acid molecule in the present invention is preferably a nucleic acid molecule having a p-value of 1 ⁇ 10 ⁇ 3 or less, more preferably a nucleic acid molecule having a p-value of 3 ⁇ 10 ⁇ 4 or less, even more preferably a nucleic acid molecule having a p-value of 1 ⁇ 10 ⁇ 4 or less, and even more preferably a nucleic acid molecule having a p-value of 3 ⁇ 10 ⁇ 5 or less.
  • the nucleic acid molecule is preferably a nucleic acid molecule having a p-value of 1 ⁇ 10 ⁇ 2 or less, more preferably a nucleic acid molecule having a p-value of 3 ⁇ 10 ⁇ 3 or less, even more preferably a nucleic acid molecule having a p-value of 1 ⁇ 10 ⁇ 3 or less, even more preferably a nucleic acid molecule having a p-value of 3 ⁇ 10 ⁇ 4 or less, and even more preferably a nucleic acid molecule having a p-value of 1 ⁇ 10 ⁇ 4 or less.
  • a significance level is set according to a known multiple correction method, whereby a preferred nucleic acid molecule can be selected.
  • a significance level can be obtained by dividing a p-value of 5 ⁇ 10 ⁇ 2 by the number of multiple comparisons, i.e. the number of polymorphisms to be compared in the chi-square test.
  • a nucleic acid molecule having a single nucleotide polymorphism below the significance level thus obtained may be selected as a more preferred nucleic acid molecule.
  • Bonferroni correction may be performed using a p-value that is combined according to a method of meta-analysis, such as Mantel-Haenszel method, and the number of single nucleotide polymorphisms to be subject for the meta-analysis.
  • a method of meta-analysis such as Mantel-Haenszel method
  • Other known methods used in multiple corrections for example, an FDR method or a permutation method may be used in the selection of a preferred nucleic acid molecule.
  • Another embodiment of the present invention provides a method of detecting the presence or absence of an allele or genotype having a high frequency in glaucoma patients in a sample containing a nucleic acid molecule from the genome.
  • the samples may be any ones so long as the nucleic acid molecules from the genome can be extracted, and for example, blood, white blood cells, hair root, hair, saliva, oral mucosa cells, skin, tissues such as muscles or organs obtained by biopsy, or the like can be used.
  • the nucleic acid molecule constituting the genome of an eukaryote is constituted by a sense strand and an antisense strand that are complementary to each other, and the determination of the allele of the single nucleotide polymorphism in the present invention can also be performed by detecting any one of the bases of the sense strand and the antisense strand of the polymorphic site.
  • any means can be used.
  • hybridization is carried out using a probe specific to each of the alleles, preferably a probe in the present invention described later, which is designed based on the sequence information disclosed in the present invention, and each of the alleles can be detected by detecting signals therefor.
  • each of the alleles opposite to each other in other words, an allele having a high association to a disease for a certain single nucleotide polymorphism and an allele having a low association thereto are each provided with different labels, and a probe capable of hybridizing these alleles to a polymorphic site, or an immobilized probe such as a microarray in which each of alleles opposite to each other is immobilized is used, whereby each of the alleles opposite to each other contained in the same sample can be detected.
  • a probe capable of hybridizing these alleles to a polymorphic site or an immobilized probe such as a microarray in which each of alleles opposite to each other is immobilized is used, whereby each of the alleles opposite to each other contained in the same sample can be detected.
  • the following method can be utilized.
  • a method of hybridization using a probe examples include Taqman method, Invader (registered trademark) method, LightCycler method, cyclin probe method, MPSS method, beads-array method, and the like, and any of these methods can be employed.
  • the probe for detecting the same allele a more preferred probe may differ in some cases depending upon a method used in the detection.
  • the determination of the allele or genotype of the single nucleotide polymorphism in the present invention does not depend upon the detection method, and it is preferable to use a suitable probe depending upon the detection method.
  • the Taqman method is a method of detecting a genetic polymorphism using an oligoprobe having a given length in which a fluorescent substance is bound to a 5′-side, and a quencher is bound to a 3′-side.
  • the presence or absence of the polymorphism is determined by hybridizing a probe to a nucleic acid molecule having a polymorphism of interest, cutting off a part of the probe on the 5′-side by a PCR reaction, and measuring a fluorescent amount emitted by a fluorescent substance.
  • the Invader method is a method of detecting a genetic polymorphism using a probe (reporter) which has a sequence common to a 3′-side of a nucleic acid molecule having a polymorphism, but the sequence on a 5′-side being completely different therefrom, and a probe (invader) having only a sequence common to a 5′-side.
  • nucleic acid molecules of interest and these two probes are hybridized, a product is then treated with a nuclease, a part of the cut-out reporter probe is hybridized with a probe for detection having a fluorescent substance and a quencher, a hybridization product is treated with a nuclease, and the fluorescent substance is released, whereby the presence or absence of the polymorphism is determined by a fluorescent amount thereof.
  • the LightCycler method is a method of detecting a polymorphism including the step of hybridizing a polymorphic detection probe having a fluorescent substance and an anchor probe having a quencher, to a nucleic acid molecule having a polymorphism previously amplified by PCR. If the hybridized DNA is gradually heated, the polymorphic detection probe is released when a given temperature is reached, and the presence or absence of the polymorphism is determined by measuring this fluorescent amount.
  • the cyclin probe method is a polymorphic analysis method utilizing a probe having a fluorescent substance or a quencher on each end of a DNA (DRD probe), wherein DNA sequences are bound in a manner that both ends of an RNA sequence having a sequence complementary to a polymorphic site of a nucleic acid molecule of interest are sandwiched.
  • a DRD probe is hybridized to a nucleic acid molecule of interest previously amplified by PCR or the like, RNase is allowed to act on this complex, and a fluorescent dye is released, whereby the presence or absence of the polymorphism is determined by measuring this fluorescent amount.
  • the MPSS method is a method of performing polymorphic analysis using an encoded adaptor probe and a decoder probe.
  • the encoded adaptor probe is an oligo DNA having a 4-bases long protruding end on a 5′-side, subsequently a recognition sequence for a restriction enzyme BbvI, and a single-stranded sequence bound to a decoder probe on a 3′-side.
  • the decoder probe is a single-stranded oligo DNA having a fluorescent substance on a 3′-side, and the decoder probe containing 4 different sequences, each sequence specifically hybridizing to a single encoded adaptor probe.
  • the nucleic acid molecule having a polymorphism is previously immobilized on beads, and an initiation adaptor containing a recognition sequence for BbvI is bound thereto, to digest with BbvI to form a 4-bases long protruding end.
  • the ligation with the encoded adaptor probe is carried out sequentially from a 3′-side of the protruding 4 bases, and the sequence of the bound encoded adaptor is detected with a specified decoder probe.
  • the beads array method is a method of performing the determination of a genotype including the step of combining beads to which a probe for allele detection and an oligonucleotide (address sequence) specifying the location information on the array of signals detected by the probe for allele detection are bound.
  • a probe for allele detection and an oligonucleotide address sequence
  • Infinium registered trademark
  • the method of the Golden Gate Assay will be shown hereinbelow.
  • two kinds of probes specifically hybridizing to each allele, and a probe capable of specifically hybridizing to a sequence located 1 to 20 bases downstream on the 3′-side of the single nucleotide polymorphism (downstream sequence recognition probe) are used.
  • the downstream sequence recognition probe an address sequence for specifying the location on the array is provided.
  • these three probes contain a sequence to which universal primers described later are bound. The three probes are annealed with a genomic DNA, and a DNA polymerase and a ligase are added thereto.
  • an allele-specific product ligating a gap between the allele-specific probe and the downstream sequence recognition probe is formed.
  • a reaction for PCR is carried out with this allele-specific product as a template using two kinds of fluorescent-labeled universal primers, each being specific to each allele, and a universal primer bound to the downstream sequence recognition probe.
  • a labeled PCR product is hybridized to an oligonucleotide immobilized on beads via an address sequence. The fluorescence on the beads is detected with a confocal laser scanner, thereby determining an allele and a genotype.
  • the probe and the fragmented genomic DNA are hybridized, and thereafter an allele-specific extension reaction takes place, whereby a base on the downstream (3′-side) by a single base of a polymorphic site labeled with a single kind of a fluorescent dye is incorporated corresponding to the probe.
  • one kind of probe without having an allele-specific sequence of a single nucleotide polymorphism in the probe is used (probe for allele detection of 50 bases, Infinium II type).
  • a 3′-end of this probe has a sequence up to a single base upstream (5′-side) from a polymorphic site.
  • the probe and the fragmented genomic DNA are hybridized, and according to a single base extension reaction, a base labeled with either one of two kinds of fluorescent dyes is incorporated corresponding to a single nucleotide polymorphic site of interest.
  • the fluorescence is detected by a confocal laser scanner, thereby determining an allele and a genotype.
  • a method without carrying out hybridization with a probe includes PCR-RFLP method, SSCP method, mass spectrometry and direct sequencing method.
  • the PCR-RFLP method is a method including the steps of forming different DNA fragments according to enzymatic digestion of a nucleic acid molecule having a polymorphism due to the existence of a polymorphism in a cleavage site of the restriction enzyme in the nucleic acid molecule, and determining the presence or absence of a polymorphism from a difference in electrophoretic patterns thereof.
  • a nucleic acid molecule of interest is amplified by PCR, this amplified fragment is cleaved with a restriction enzyme, and a fragment formed electrophoretically is analyzed.
  • the length of the nucleic acid molecule comprising an amplified polymorphism is usually from 50 to 10,000 base pairs, and more preferably from 100 to 1,000 base pairs.
  • the SSCP method is a method including the steps of amplifying a nucleic acid molecule having a polymorphism by PCR, forming a single-stranded DNA, electrophoresing the product, and determining the presence or absence of a polymorphism from a difference in the electrophoretic patterns thereof.
  • the nucleic acid molecule of interest is amplified by PCR, and a single-stranded DNA is formed by subjecting this amplified fragment to heat or an alkali treatment. This single-stranded DNA forms a base sequence-specific higher-order structure; therefore, if these amplified fragments are electrophoresed, a difference in the electrophoretic mobility is found due to the difference in its structure.
  • the primer used in PCR is labeled with a radioisotope or fluorescent substance.
  • the length of the nucleic acid molecule comprising an amplified polymorphism is usually from 50 to 10,000 base pairs, and more preferably from 100 to 1,000 base pairs.
  • the mass spectrometry is a method including the steps of ionizing a polymer with a matrix and a laser or the like, accelerating the ionized polymer in a high electric field to allow a flight to a detector, and identifying mass from a difference in the flight time, or the like. This mass spectrometry is combined with the above primer extension method or the like to detect a polymorphism.
  • a single base extension reaction is carried out with a primer complementary to a sequence up to a single base upstream of a polymorphic site of a nucleic acid molecule having a polymorphism, any one of 4 kinds of dideoxyribonucleotides, and deoxyribonucleic acids other than those corresponding the above, and a difference in mass of nucleic acid products having different sequences incorporated in a 3′-end is determined, whereby a polymorphism can be identified.
  • the direct sequencing method is a method of directly reading off a base sequence of a nucleic acid molecule having a polymorphism. Representative methods are called Sanger method (dideoxy method). A primer that is unlabeled or labeled with a radioisotope or a fluorescent substance is bound to a nucleic acid molecule of interest, an extension reaction with Klenow enzyme or the like is stopped with four kinds of dideoxyribonucleotides that are unlabeled or labeled with a radioisotope or a fluorescent substance, the product is digested with a restriction enzyme, and a DNA fragment generated is separated by electrophoresis.
  • the base sequence of a 3′-end is read off in the order of fragments having a lower molecular weight on the basis of an electrophoretic image, thereby a base sequence containing a few bases before and after a polymorphism is determined.
  • a primer extension method As a modified method thereof, there is a method called a primer extension method. This is a method including the steps of carrying out a single base extension reaction using a primer complementary to a sequence up to a single base upstream of a polymorphic site of a nucleic acid molecule having a polymorphism, and reading off any one of the sequences of the 4 kinds of dideoxyribonucleotides incorporated in the 3-end.
  • the length of the primer used in the extension reaction is usually from 10 to 300 base pairs, and preferably from 15 to 25 base pairs.
  • the hybridization means that a nucleic acid molecule having a certain sequence is associated with a nucleic acid molecule complementary to at least a part of the nucleic acid molecule via a hydrogen bond on the basis of base sequences that are complementary to each other.
  • the kind of the complementary nucleic acid molecule associated with the original nucleic acid molecule may be identical or different, and a nucleic acid constituting these nucleic acid molecules can be a deoxyribonucleic acid, a ribonucleic acid, or a peptide nucleic acid.
  • thymine may be read as uracil.
  • the stringent conditions in the present invention mean conditions in which a nucleic acid molecule having a sequence complementary to a partial sequence of a nucleic acid molecule having a certain sequence is specifically hybridized to the nucleic acid molecule (Fred M. Ausuble et al., Current Protocols in Molecular Biology, 2.10.1-2.10.16, John Wiley and Sons, Inc).
  • conditions as described above include conditions such as a temperature lower than a melting temperature (Tm) of a complex formed between a nucleic acid molecule having a certain sequence and a complementary nucleic acid molecule hybridized to the nucleic acid molecule by preferably from about 5° to about 30° C., and by more preferably about 10° to about 25° C., a reaction solution for hybridization, such as SSC (mixed solution of sodium chloride and sodium citrate) in a concentration of 0.01 to 6-folds, SSPE (mixed solution of sodium chloride, sodium dihydrogenphosphate, and EDTA) or MES (a mixed solution of 2-(N-morpholino)ethanesulfonic acid and tetramethylammonium chloride) buffer, and hydrogen ion concentrations of a pH of from 6 to 8.
  • Tm melting temperature
  • the stringent conditions in a case where an immobilized probe is prepared by immobilizing a 25 by DNA probe include conditions of hybridization at 49° C. in the MES buffer (hydrogen ion concentrations being from 6.5 to 6.7) in a 1-fold concentration, and sequentially washing with SSC (hydrogen ion concentrations being 8.0) in a 6-fold concentration at 25° C., and thereafter SSC (hydrogen ion concentrations being 8.0) in a 0.6-fold concentration at 45° C.
  • the term allele-specific means that the allele is contained in a sequence from the genome including the polymorphic site or in a prepared nucleic acid molecule including the polymorphic site, or a certain nucleic acid molecule is capable of specifically hybridizing under stringent conditions to a nucleic acid molecule having a sequence containing the allele in the polymorphic site, in other words, in the manner of being capable of discriminating the allele and the opposite allele.
  • Base sequences of 61 bases in length including a single nucleotide polymorphism associated with the onset of glaucoma, disclosed in the present invention are composed of two pairs of base sequences which differ only by a base in the center (i.e. 31st base) (i.e. those pairs are consisting of a sequence having odd number of SEQ ID No. and a sequence having even number of SEQ ID No.), and the 31st base is a polymorphic site.
  • the high-risk alleles in the polymorphic sites are listed in Tables 1 and 2 or Tables 52 to 63 given later.
  • any of these single nucleotide polymorphisms in a case where the existence of an allele that exists in a high frequency in glaucoma patients is determined, a high-risk allele in a sample is detected, whereby the existence of the allele that exists in a high frequency in glaucoma patients can be determined.
  • the genotype can be determined by detecting the presence or the absence of each of the alleles opposite to each other contained in one sample. In detail, in a case where only a certain allele is detected, the genotype is a homozygote of the allele, and in a case where two alleles are detected, the genotype is a heterozygote having the two alleles. In at least one of these single nucleotide polymorphisms, by detecting a genotype, it is determined whether or not the genotype that is identified in a higher frequency in a glaucoma patient group than that of a non-patient group exists in a sample.
  • a homozygote of the high-risk allele or a heterozygote is a genotype that is identified in a higher frequency in a glaucoma patient group than that of a non-patient group
  • a homozygote of the high-risk allele is a genotype that is identified in a higher frequency in a glaucoma patient group than that of a non-patient group. It is preferable that each of the opposite alleles is measured in a single operation, from the viewpoint of reducing judgmental error.
  • the sample is analyzed in the manner described above, and in a case where the allele or genotype that is identified in a higher frequency in a glaucoma patient group than that of a non-patient group exists in the sample, there are some high probabilities that an individual donating the sample not having glaucoma at the present point is predicted to have a high onset risk of glaucoma, or is determined that a precision examination for glaucoma such as visual field examination is necessary, and that an individual donating the sample who is suspected of having glaucoma should be diagnosed as glaucoma.
  • the single nucleotide polymorphism used in the detection is a single nucleotide polymorphism which is located on a 31st base of a base sequence, wherein the base sequence is at least one base sequence selected from the group consisting of base sequences shown in SEQ ID NOs: 203 to 514 or a complementary sequence thereto, more preferably a single nucleotide polymorphism which is located on a 31st base of a base sequence, wherein the base sequence is at least one base sequence selected from the group consisting of base sequences shown in
  • SEQ ID NOs: 203 to 238 or a complementary sequence thereto even more preferably a single nucleotide polymorphism which is located on a 31st base of a base sequence, wherein the base sequence is at least one base sequence selected from the group consisting of pairs of base sequences containing a single nucleotide polymorphism listed below or a complementary sequence thereto, wherein, as mentioned above, in the pairs of SEQ ID NOs: shown in a to r, each of the pairs of sequences corresponds to one single nucleotide polymorphism, and each of the base sequences is a base sequence containing an allele opposite to each other of the single nucleotide polymorphism in a 31st base:
  • any one of the single nucleotide polymorphisms is used, especially, it is preferable that an allele of a single nucleotide polymorphism located on a 31st base of a base sequence is used, wherein the base sequence is at least one base sequence selected from the group consisting of the following base sequences containing a single nucleotide polymorphism:
  • SEQ ID NO: 203 SEQ ID NO: 206, SEQ ID NO: 208, SEQ ID NO: 209, SEQ ID NO: 211, SEQ ID NO: 214, SEQ ID NO: 216, SEQ ID NO: 218, SEQ ID NO: 219, SEQ ID NO: 221, SEQ ID NO: 224, SEQ ID NO: 226, SEQ ID NO: 228, SEQ ID NO: 229, SEQ ID NO: 231, SEQ ID NO: 234, SEQ ID NO: 235, and SEQ ID NO: 238,
  • sequences are sequences containing a high-risk allele in each of polymorphic sites.
  • the precision of the determination of a future onset risk of glaucoma can be improved by detecting a combination of two or more of alleles or genotypes associated with glaucoma in the present invention, using one sample.
  • any ones can be used so long as they are a single nucleotide polymorphism in the present invention, preferably a single nucleotide polymorphism having a low p-value, and more preferably a single nucleotide polymorphism of which p-value obtained by combining the results obtained in two analyses by a meta-analysis method, such as Mantel-Haenszel method, is determined to be significant even below the level of Bonferroni correction.
  • a meta-analysis method such as Mantel-Haenszel method
  • a risk of a disease is predicted by combining the single nucleotide polymorphisms in the present invention
  • the prediction is carried out employing only one of the single nucleotide polymorphisms in the state of linkage disequilibrium, in a case that the plural single nucleotide polymorphisms that are in the state of linkage disequilibrium mentioned above are contained.
  • an onset risk of glaucoma can be predicted using the regression formula obtained by the logistic regression analysis.
  • a dependent variable ⁇ is calculated by substituting a value corresponding to each single nucleotide polymorphism into this formula.
  • a dependent variable ⁇ is greater than a given threshold (for example, 0.5), the determination can be made that this sample donor has an onset risk.
  • the single nucleotide polymorphisms used in the detection are preferably single nucleotide polymorphisms which are located on 31st bases of base sequences, wherein the base sequences are base sequences containing two or more different single nucleotide polymorphisms, selected from the group consisting of base sequences shown in SEQ ID NOs: 203 to 514 or a complementary sequence thereto,
  • base sequences are base sequences containing two or more different single nucleotide polymorphisms, selected from the group consisting of base sequences shown in SEQ ID NOs: 203 to 238 or a complementary sequence thereto,
  • base sequences are base sequences containing two or more different single nucleotide polymorphisms, selected from the group consisting of pairs of base sequences containing a single nucleotide polymorphism listed below or a complementary sequence thereto,
  • each of the pairs of sequences corresponds to one single nucleotide polymorphism
  • each of the base sequences is a base sequence containing an allele opposite to each other of the single nucleotide polymorphism in a 31st base:
  • the single nucleotide polymorphisms to be used in combination are those that are not in the state of linkage disequilibrium, and from this viewpoint, in all the embodiments of the combinations mentioned above, supposing that
  • an allele of a single nucleotide polymorphism located on a 31st base of a base sequence is used, wherein the base sequence is a base sequence containing two or more different single nucleotide polymorphisms, selected from the group consisting of the following base sequences containing a single nucleotide polymorphism:
  • an allele-specific nucleic acid molecule or probe capable of detecting an allele associated with glaucoma, and a method of detecting an allele or a genotype associated with glaucoma using the probe are provided.
  • any probes may be used so long as the probe is capable of hybridizing under the stringent conditions to an allele-specific sequence, in a polymorphic site of the single nucleotide polymorphism associated with glaucoma in the present invention.
  • the determination of the allele in a polymorphic site can be made by detecting any one of polymorphic sites of the sense strand and the antisense strand on the genome; therefore, the probe in the present invention embraces any one of sequences complementary to a sequence specific to an allele of the sense strand and sequences complementary to a sequence specific to an allele of the antisense strand, in other words, sequences specific to an allele of the sense strand.
  • the probe in the present invention can also be used in the detection of cDNA or mRNA, containing a single nucleotide polymorphism in the present invention.
  • a probe in which the single nucleotide polymorphism exists in exon or neighborhood thereof is used.
  • the probes capable of detecting each of alleles of the single nucleotide polymorphisms listed in Tables 1 and 2, Tables 5 to 25, Tables 26 to 28, Tables 29 to 51, or Tables 52 to 62 given later or a complementary strand thereto, and the probes capable of specifically detecting each of alleles of any single nucleotide polymorphisms that exist in a region associated with glaucoma listed in Tables 3 and 4 or Tables 63 to 70 given later or a complementary strand thereto are all embraced in the probe in the present invention.
  • the probe of the present invention is preferably a probe capable of specifically detecting each of alleles of a single nucleotide polymorphism or a complementary strand thereto, of which p-value is 1 ⁇ 10 ⁇ 3 or less, more preferably a probe capable of specifically detecting each of alleles of a single nucleotide polymorphism or a complementary strand thereto, of which p-value is 3 ⁇ 10 ⁇ 4 or less, even more preferably a probe capable of specifically detecting each of alleles of a single nucleotide polymorphism or a complementary strand thereto, of which p-value is 1 ⁇ 10 ⁇ 4 or less, and even more preferably a probe capable of specifically detecting
  • the probe is preferably a probe capable of specifically detecting each of alleles of a single nucleotide polymorphism or a complementary strand thereto, of which p-value is 1 ⁇ 10 ⁇ 2 or less, more preferably a probe capable of specifically detecting each of alleles of a single nucleotide polymorphism or a complementary strand thereto, of which p-value is 3 ⁇ 10 ⁇ 3 or less, even more preferably a probe capable of specifically detecting each of alleles of a single nucleotide polymorphism or a complementary strand thereto, of which p-value is 1 ⁇ 10 ⁇ 3 or less, even more preferably a probe capable of specifically detecting each of alleles of a single nucleotide polymorphism or a complementary strand thereto, of which p-value is 3 ⁇ 10 ⁇ 4 or less, and
  • the probe in the present invention preferably contains an allele-specific sequence or a complementary strand thereto, and even more preferably in the probe in the present invention, a sequence contributing to an allele-specific hybridization consists only of an allele-specific sequence or a complementary strand thereto.
  • a spacer or any sequences of several bases that are not from an allele-specific sequence for the purpose of providing stabilization or the like can be added in an end, within the range that the probe is capable of hybridizing to the sequence under the stringent conditions.
  • the added sequence is preferably a sequence that does not take a three-dimensional structure, such as a hairpin structure.
  • the probe can be provided with any labels for use in the detection. Any labels to be provided to the probe that are ordinarily used can be used, and in general, a fluorescent label such as FITC or Cy3, biotin, an enzyme label such as an alkaline phosphatase and horseradish peroxidase, or the like is usable.
  • a fluorescent label such as FITC or Cy3
  • biotin an enzyme label such as an alkaline phosphatase and horseradish peroxidase, or the like is usable.
  • streptavidin capable of specifically binding to biotin is previously provided with a further detectable label, and the labeled streptavidin is used as a secondary label.
  • a labeled anti-biotin antibody can also be used in place of the labeled streptavidin.
  • any known methods may be used, and the methods are well known to one of ordinary skill in the art.
  • An arbitrary sequence which serves as a spacer as mentioned above may be added to the probe, and the spacer may be provided with a label.
  • a reagent for labeling a probe, a labeled streptavidin, a labeled anti-biotin antibody or the like is commercially available as a reagent, and can also be purchased.
  • the probe in the present invention is not limited whether it is a deoxyribonucleic acid, a ribonucleic acid, or a peptide nucleic acid, and a probe containing a mixed sequence thereof is also embraced in the present invention, so long as the probe is capable of specifically hybridizing to a nucleic acid molecule having an allele of interest.
  • a probe containing a ribonucleic acid in the sequence of the probe in the present invention (including a sequence complementary thereto), thymine may read as uracil.
  • the probe in the present invention may be subjected to chemical modifications as needed, so long as the probe is capable of specifically hybridizing under stringent conditions to a nucleic acid molecule having an allele of interest.
  • any known methods may be used as the method of providing a chemical label.
  • the probe for the detection can be reacted with the sample in the state of solution and then detected by a known method, or previously immobilized to a carrier.
  • the probe can take the form of an immobilized probe obtained by previously immobilizing a probe corresponding to each of the alleles of several to several hundred-thousand different single nucleotide polymorphisms to a location defined on a solid carrier in the number of from one to dozen probes per one single nucleotide polymorphism, reacting a sample to the immobilized probes, scanning a signal generated from a hybridized probe, and analyzing the scanned data with a computer, which is a so-called microarray.
  • the probe takes the form of an immobilized probe, the largest number of the immobilized probes are limited by immobilization density and area of immobilized sites for the probes.
  • signals on the solid phase from the nucleic acid molecule having a labeled target allele can be detected by previously labeling a nucleic acid molecule in a sample by a known method, and binding the labeled nucleic acid molecule with an immobilized unlabeled probe in the present invention, or by binding a nucleic acid molecule having an allele to be detected to an immobilized unlabeled probe in the present invention, and thereafter labeling the product according to a known method.
  • the immobilization can be carried out by any of known method, and for example, a method such as synthetic oligoprint or spotting photolithograph can be used.
  • the material for the carrier is not limited, and a generally used material, for example, a polymer such as a polycarbonate or a polystyrene, glass, silicon crystal or the like can be used.
  • a carrier may be provided with a coating such as cationization before the immobilization.
  • blocking can be carried out with a known blocking agent after the immobilization.
  • the blocking agent as mentioned above may be any ones so long as the blocking agent is capable of controlling the nonspecific nucleic acids from being adsorbed to the carrier, and for example, salmon sperm DNA, Denhardt's solution, Cot-I DNA extracted from human placenta, an anionic surfactant such as sodium dodecyl sulfate, a nonionic surfactant such as polyoxyethylene sorbitan monolaurate, or the like can be used.
  • each of the opposite alleles contained in one sample is detected under the same operation by immobilizing a probe specific to each of the alleles opposite to each other on the same carrier.
  • immobilizing a probe specific to each of the alleles opposite to each other on the same carrier In the construction as described above, not only the alleles but also the genotypes in the samples can be determined.
  • the probe used in the detection of the allele is a probe having a length of preferably from 16 to 55 bases, more preferably from 23 to 27 bases or 47 to 53 bases, and even more preferably 25 bases in total of a length of the polymorphic site and some bases before and after the polymorphic site, the probe containing the polymorphic site mentioned above and a surrounding sequence thereof, or a sequence complementary thereto, that the probe is a probe containing the polymorphic site mentioned above and a 5′-upstream side thereof, preferably a sequence of 49 bases (i.e.
  • the probe containing the polymorphic site mentioned above and a surrounding sequence thereof, or a sequence complementary thereto, or that the probe is a probe containing a sequence of 50 bases on a 5′-upstream side of the polymorphic site mentioned above, the probe having a sequence adjoining the polymorphic site mentioned above, or a sequence complementary thereto.
  • An even more preferred probe used in the detection of the allele is:
  • the probe usable in the detection is a probe containing a single nucleotide polymorphism which is located on a 31st base of a base sequence, wherein the base sequence is at least one base sequence selected from the group consisting of base sequences shown in SEQ ID NOs: 203 to 514 or a complementary sequence thereto, or a partial sequence thereof, and/or a probe having a base sequence containing at least one base sequence selected from the group consisting of base sequences shown in SEQ ID NOs: 515 to 694 or a complementary sequence thereto, more preferably a probe containing a single nucleotide polymorphism which is located on a 31st base of a base sequence, wherein the base sequence is at least one base sequence selected from the group consisting of base sequences shown in SEQ ID NOs: 203 to 238
  • any one of the single nucleotide polymorphisms is used, especially, it is preferable that in Group A, a probe containing an allele of a single nucleotide polymorphism located on a 31st base of a base sequence is used, wherein the base sequence is at least one base sequence selected from the group consisting of the following base sequences containing a single nucleotide polymorphism:
  • the probes usable in the detection are preferably probes containing a single nucleotide polymorphism which is located on a 31st base of a base sequence, wherein the base sequence is a base sequence containing a single nucleotide polymorphism, selected from the group consisting of base sequences shown in SEQ ID NOs: 203 to 514 or a complementary sequence thereto, or a partial sequence thereof, and/or probes having a base sequence containing a base sequence selected from the group consisting of base sequences shown in SEQ ID NOs: 515 to 694 or a complementary sequence thereto, wherein the probes are probes corresponding to two or more different single nucleotide polymorphisms thereof,
  • the single nucleotide polymorphisms to be used in combination are those that are not in the state of linkage disequilibrium, and from this viewpoint, in all the embodiments of the combinations mentioned above, supposing that, in Group A, a group composed of a base sequence containing a single nucleotide polymorphism which is located on a 31st base of a base sequence, wherein the base sequence is a base sequence belonging to the group consisting of:
  • a probe containing an allele of a single nucleotide polymorphism located on a 31st base of a base sequence wherein the base sequence is a base sequence containing a single nucleotide polymorphism selected from the group consisting of the following base sequences containing a single nucleotide polymorphism:
  • the probe in a case where a Taqman method is used in the detection of an allele usually has a length of preferably from 10 to 300 bases, and contains the polymorphic site mentioned above and a surrounding sequence thereof, or a sequence complementary thereto, and the probe also contains a fluorescent substance and a quencher. More preferably, the probe has a length of 20 to 60 bases, and contains the polymorphic site mentioned above and a surrounding sequence thereof, or a sequence complementary thereto, and the probe contains a fluorescent substance and a quencher.
  • the probes in a case where an Invader method is used in the detection of an allele comprise a probe (reporter) which have a common sequence to a 3′-side of the polymorphic site mentioned above and a sequence on a 5′-side being completely different therefrom, and a probe (invader) only composed of the common sequence to a 5′-side.
  • These probes usually have a length of preferably from 10 to 300 bases, and more preferably a length of from 20 to 60 bases.
  • the probe in a case where a LightCycler method is used in the detection of an allele usually has a length of preferably from 10 to 300 bases, and contains the polymorphic site mentioned above and a surrounding sequence thereof, or a sequence complementary thereto, and the probe contains a fluorescent substance and a quencher. More preferably, the probe has a length of 20 to 60 bases, and contains the polymorphic site mentioned above and a surrounding sequence thereof, or a sequence complementary thereto, and the probe contains a fluorescent substance and a quencher.
  • the probe in a case where a cyclin probe method is used in the detection of an allele is a probe in which DNA sequences are bound in a manner that both ends of an RNA sequence having the polymorphic site and a surrounding sequence thereof, or a sequence complementary thereto, are sandwiched, and each of DNA ends has a fluorescent substance or a quencher.
  • These probes usually have a length of preferably from 10 to 300 bases, and contain the polymorphic site mentioned above and a surrounding sequence thereof, or a sequence complementary thereto. More preferably, the probe has a length of 20 to 60 bases, and contains the polymorphic site mentioned above and a surrounding sequence thereof, or a sequence complementary thereto.
  • the probes in a case where an MPSS method is used in the detection of an allele comprise an oligo DNA (encoded adaptor probe) having a protruding end of 4 bases on a 5′-side, subsequently a recognition sequence for a restriction enzyme BbvI, and a single-stranded sequence to which a decoder probe is bound on a 3′-side, and a single strand oligo DNA (decoder probe) which has fluorescent substance on a 3′-side, and containing 4 different sequences, each sequence specifically hybridizing to one of the encoded adaptor probes.
  • a DNA sequence is bound in a manner that both ends of an RNA sequence having the polymorphic site mentioned above and a surrounding sequence thereof, or a sequence complementary thereto, are sandwiched, and each of DNA ends has a fluorescent substance or a quencher.
  • the encoded adaptor probe usually has a length of preferably from 10 to 300 base pairs, and more preferably from 15 to 40 base pairs.
  • the decoder probe usually has a length of preferably from 10 to 300 base pairs, and more preferably from 5 to 30 base pairs.
  • kits of detecting a single nucleotide polymorphism associated with glaucoma is provided.
  • the kit of the present invention (or a composition for predicting a risk) embraces all those kits so long as the allele or genotype of any one of single nucleotide polymorphisms associated with glaucoma disclosed in the present invention can be detected in a nucleic acid molecule in a sample.
  • the kit of the present invention may be those that detect a base of either the sense strand or the antisense strand of the single nucleotide polymorphism, or those that detect bases of both the strands.
  • the kit of the present invention is based on the results obtained in a single analysis using a microarray for a kit of detecting an allele or genotype associated with glaucoma for detecting, for example, 500,000 single nucleotide polymorphisms in a single operation
  • the kit is preferably a kit of detecting an allele or genotype associated with glaucoma for single nucleotide polymorphisms having a p-value of 1 ⁇ 10 ⁇ 4 or less listed in Tables 1 and 2 set forth below, more preferably a kit of detecting an allele or genotype associated with glaucoma for single nucleotide polymorphisms having a p-value of 3 ⁇ 10 ⁇ 4 or less, even more preferably a kit of detecting an allele or genotype associated with glaucoma for single nucleotide polymorphisms having a p-value of 1 ⁇ 10 ⁇ 4 or less, and even more preferably a kit of detecting an allele or genotype
  • the kit is preferably a kit of detecting an allele or genotype associated with glaucoma for single nucleotide polymorphisms having a p-value listed in Tables 52 to B set forth below of 1 ⁇ 10 ⁇ 2 or less, more preferably a kit of detecting an allele or genotype associated with glaucoma for single nucleotide polymorphisms having a p-value of 3 ⁇ 10 ⁇ 3 or less, even more preferably a kit of detecting an allele or genotype associated with glaucoma for single nucleotide polymorphisms having a p-value of 1 ⁇ 10 ⁇ 3 or less, even more preferably a kit of detecting an allele or genotype associated with glaucoma for single nucleotide polymorphisms having a p-value of 3 ⁇ 10 ⁇ 4 or less,
  • a kit of detecting both an allele identified in a high frequency in the glaucoma patient group mentioned above and an allele opposite to the allele is also one embodiment of the present invention.
  • a genotype of each of the alleles can also be determined.
  • kits of the present invention By detecting the presence of an allele or a genotype that is identified in a high frequency in glaucoma patients in the sample using the kit of the present invention, a future onset risk of glaucoma of an individual not having glaucoma at the present stage can be predicted, whether or not precise visual field examinations for glaucoma are required can be determined, or the diagnosis of an individual who is suspected of glaucoma can be made for glaucoma.
  • kits for determining alleles that are opposite to each other in a single operation can be prepared by using a probe specific to each of the alleles that are opposite to each other, and providing different labels to the probes, or providing in the form of a microarray or beads array as mentioned above.
  • the precision for the prediction of the onset risk of glaucoma or the determination of whether or not precise visual field examinations are required can also be improved by providing a kit having the constitution of detecting these plural alleles or genotypes using one sample. Even in the constitution as described above, a constitution can be taken that the detection is carried out in a single operation by having the form of probes provided with different labels, or the form of the microarray or beads array mentioned above.
  • the kit usable in detecting or predicting a risk is
  • any one of the single nucleotide polymorphisms is used, especially, in Group A, preferred is a kit of detecting a single nucleotide polymorphism associated with the onset of glaucoma or predicting an onset risk of glaucoma, using a nucleic acid molecule containing an allele of a single nucleotide polymorphism located on a 31st base of a base sequence, wherein the base sequence is at least one base sequence selected from the group consisting of the following base sequences containing a single nucleotide polymorphism:
  • the kit usable in detecting or predicting a risk is
  • the single nucleotide polymorphisms to be used in combination are those that are not in the state of linkage disequilibrium, and from this viewpoint, in all the embodiments of the combinations mentioned above, supposing that, in Group A, a group composed of a base sequence containing a single nucleotide polymorphism which is located on a 31st base of a base sequence, wherein the base sequence is a base sequence belonging to the group consisting of:
  • kits of detecting a single nucleotide polymorphism associated with the onset of glaucoma or a kit of predicting an onset risk of glaucoma using a nucleic acid molecule comprising an allele of a single nucleotide polymorphism located on a 31st base of a base sequence, wherein the base sequence is a base sequence containing a single nucleotide polymorphism, selected from the group consisting of the following base sequences containing a single nucleotide polymorphism:
  • a prediction of an onset risk of glaucoma using a single nucleotide polymorphism in the present invention can be performed in two or more steps as follows; candidates who are considered that precise prediction of an onset risk of glaucoma is necessary are selected, and the candidates are subjected to detailed prediction of a risk.
  • prediction of a risk is performed in two or more multi-steps
  • prediction of an onset risk of glaucoma mentioned above is preformed on at least one single nucleotide polymorphism in the present invention, preferably any one or several single nucleotide polymorphisms
  • prediction of detailed risks may be performed using a combination of the single nucleotide polymorphisms of the present invention mentioned above.
  • the number of combinations may be further increased as occasion demands, whereby precision of the prediction of a risk can also be improved.
  • the reduction in costs for performing the prediction of a risk and the prediction of a risk in a high precision can be both accomplished.
  • the prediction of a risk in an initial step may be a convenient method of predicting a risk.
  • a method of predicting a risk so that an immobilized probe capable of detecting at least one of the single nucleotide polymorphisms, preferably any one or several single nucleotide polymorphisms, is immobilized in a manner that at least one of the single nucleotide polymorphisms in the present invention is detectable is a convenient method, and can be realized at a low cost.
  • a method for nucleic acid extraction in this case, a kit that can be realized according to a known technique, or a commercially available simple kit for nucleic acid extraction can be used.
  • a method including the steps of using, for example, an enzyme-labeled probe as the immobilized probe used in the prediction of a risk as described above, and detecting the probe according to a colorimetric method.
  • samples used in the detection those that are obtained in a relatively low penetration, such as saliva, oral mucosa cells, urine, hair root, blood or white blood cells are preferred.
  • the prediction of a risk in a next step may be a method of predicting a risk with an emphasis on precision.
  • the detection of a single nucleotide polymorphism associated with the onset of glaucoma is carried out by combining two or more single nucleotide polymorphisms in the present invention mentioned above, whereby prediction of a risk may be performed in a high precision.
  • the precision for prediction of a risk can be improved, while reducing the costs or lowering a burden on a subject at an initial step to a minimum level.
  • the determination can be made that an individual who has an allele or genotype on the genome that is identified in a high frequency in glaucoma patients disclosed in the present invention has a high risk of the onset of glaucoma in future, and that an individual who does not have an allele or genotype that is identified in a high frequency in the glaucoma patients has a low risk of the onset of glaucoma in future.
  • an individual having an allele or genotype on the genome that is identified in a high frequency in glaucoma patients disclosed in the present invention has a possibility of being in an early stage of glaucoma that is difficult to be diagnosed according to a simple method of determination of glaucoma, such as measurement of intraocular pressure or examination of ocular fundus, and that is diagnosed for the first time after performing visual field examination. Therefore, a single nucleotide polymorphism in the present invention is detected, whereby whether or not the visual field examination is required can be screened.
  • a total DNA was extracted from blood of each of patients diagnosed as glaucoma, and non-patients diagnosed as being not with glaucoma and determined not to have any family history in glaucoma according to a medical interview, and gene loci associated with the disease were analyzed based on about 500,000 known single nucleotide polymorphisms on the human genome as an index to determine an association of a single nucleotide polymorphism and the disease.
  • patients with fast progression of glaucoma i.e. progressive glaucoma cases
  • patients with slow progression of glaucoma i.e. nonprogressive glaucoma cases were subjected to the identification of a single nucleotide polymorphism and the association of the single nucleotide polymorphism with the progression in the same manner as above.
  • a commercially available automated nucleic acid extraction apparatus (QUIAGEN, BIOROBOT (registered trademark) EZ1), and a kit for extraction of a nucleic acid (EZ1 DNA Blood 350 ⁇ l Kit) compatible to the extraction apparatus and in which nucleic acids absorbed to magnetic beads were collected by a magnetic force were used.
  • a total DNA was extracted in accordance with the instruction manuals of the apparatus and kit. According to the present method, a total DNA of about 5 ⁇ g was obtained from 350 ⁇ L of a blood specimen.
  • microarray type single nucleotide polymorphism analysis kit (Affimetrix (GeneChip(registered trademark) Human Mapping 500K) (hereinafter also referred to as microarray) capable of analyzing about 500,000 known single nucleotide polymorphisms on the human genome was used.
  • a scanner Affimetrix (GeneChip(registered trademark) Scanner 3000)
  • a specialized analysis software (Affimetrix (GTYPE(registered trademark)) was used.
  • a fluorescence from phycoerythrin bound via biotin and streptavidin to DNA ends of samples hybridized with an immobilized allele-specific probe was read using the scanner mentioned above, and analyzed with the software mentioned above.
  • Probes corresponding to about 250,000 single nucleotide polymorphisms each are previously immobilized to the microarray for NspI-treated sample and the microarray for StyI-treated sample, respectively, and analytical results for about 500,000 single nucleotide polymorphisms per one sample were obtained by combination of both the results.
  • opposite alleles of each of the single nucleotide polymorphisms were read with a single operation, and consequently, a genotype was determined.
  • a probe for a sense strand or a probe for an antisense strand of the genome is used as the probe immobilized to the kit.
  • the determination results for the present kit using 270 samples and those in HapMap are compared for single nucleotide polymorphisms overlapping between single nucleotide polymorphisms reported in the HapMap project and single nucleotide polymorphisms in the kit. As a result, a concordance rate of the single nucleotide polymorphisms shows 99% or more.
  • the analytical results of a single nucleotide polymorphism obtained in each of the patients were stored in the Laboratory Information Management System (World Fusion, LaboServer) adopting a relational database.
  • a specialized analysis program for a single nucleotide polymorphism was created and loaded within the system, and the analysis was performed as follows: A single nucleotide polymorphism considered to have a high experimental reliability was extracted by rejecting a single nucleotide polymorphism having a call rate of less than 90% in both the glaucoma patient group and the non-patient group, a single nucleotide polymorphism having a difference in call rates between the glaucoma patient group and the non-patient group by 5% or more, a single nucleotide polymorphism having a minor allele frequency of less than 5%, and a single nucleotide polymorphism that is determined to deviate from the Hardy-Weinberg's equilibrium under conditions of a p-value of 1 ⁇ 10
  • the allele frequencies and the genotype frequencies were statistically compared according to the chi-square test.
  • cluster images serving as a basis for the determination of a genotype were confirmed.
  • the single nucleotide polymorphism was considered to be a non-subject of the analysis. In other words, the errors in the determination of a genotype were excluded by this step.
  • the evaluation of the cluster was performed without informing the names of single nucleotide polymorphisms and the critical rates.
  • Allele Frequency Number of Detection of an Allele in Group/Total Number of Detection of Alleles in Group formula (1)
  • Genotype Frequency Number of Detection of a Genotype in Group/Total Number of Detection of Genotypes in Group formula (2)
  • Odds Ratio for Allele [(Number of Detection of an Allele Identified in High Frequency in Glaucoma Patient Group, in Glaucoma Patient Group)/(Number of Detection of an Allele Opposite to the Allele Identified in High Frequency in Glaucoma Patient Group, in Glaucoma Patient Group)]/[(Number of Detection of the Allele Identified in High Frequency in Glaucoma Patient Group, in Non-Patient Group)/(Number of Detection of the Allele Opposite to the Allele Identified in High Frequency in Glaucoma Patient Group, in Non-Patient Group)] formula (3)
  • Tables 1 and 2 list dbSNP ID number or Affimetrix Array ID number specifying known single nucleotide polymorphisms obtained, each of bases constituting Allele 1 and Allele 2, the exon, intron information (in a case where a single nucleotide polymorphism exists on a gene, the gene name and the exon or intron in which SNP exists are shown, and in a case where a single nucleotide polymorphism does not exist on a gene, neighboring genes and a distance between the gene and the single nucleotide polymorphism are shown), the chromosome number at which a single nucleotide polymorphism exists, the physical location of a single nucleotide polymorphism, the p-value for an allele according to a chi-square test ( ⁇ log P), the high-risk allele frequencies in the glaucoma patient group and the non-patient group, the type of the high-risk allele (indicating whether the high-risk all
  • a single nucleotide polymorphism is associated with the onset of glaucoma, of which 31st base is either A (Allele 1) or G (Allele 2), wherein Allele 1 indicated as a high-risk allele, that is, an allele of being A in the single nucleotide polymorphism is identified in a high frequency in the glaucoma patient group.
  • the degree of which the risk of a disease increases can be predicted in a case of having the allele or genotype.
  • all the sequences disclosed in Tables 1 and 2 have a polymorphic site associated with glaucoma in the sequence, and one allele or at least one genotype in the polymorphic site is identified in a high frequency in the glaucoma patient group.
  • the allele or genotype identified in a high frequency in the glaucoma patient group of a single nucleotide polymorphism listed in Tables 1 and 2 can be used as a marker showing that an onset risk of glaucoma is high.
  • an allele that is opposite to the allele or a genotype other than the genotype can be used as a marker showing that an onset risk of glaucoma is low.
  • the surrounding regions and/or genes of the single nucleotide polymorphisms listed in Tables 1 and 2 were determined on the basis of the database provided by the HapMap project.
  • regions in which single nucleotide polymorphisms that were considered to be in a linkage disequilibrium with the single nucleotide polymorphisms listed in Tables 1 and 2 exist were determined, on the basis of the linkage disequilibrium data in combination of the Japanese and the Chinese in the HapMap project.
  • a single nucleotide polymorphism of which p-value is lowest in each region is considered to be a single nucleotide polymorphism representing the region.
  • Tables 3 and 4 list a single nucleotide polymorphism representing the region, the chromosome number at which the region exists, the physical location of the region (start point and end point) and the gene name contained in the region.
  • the region listed in Tables 3 and 4 is a region or gene considered to be linked with a single nucleotide polymorphism listed in Tables 3 and 4 which is associated with glaucoma in the present invention, and a single nucleotide polymorphism which exists in these regions or genes is considered to be linked with a single nucleotide polymorphism in the present invention.
  • any single nucleotide polymorphisms which exist in these regions are linked with the single nucleotide polymorphism which exists in the region as listed in Tables 3 and 4, and any of these single nucleotide polymorphisms can be used in the prediction of a risk of glaucoma in the same manner.
  • a single nucleotide polymorphism of which allele or genotype shows association with glaucoma at a p-value of 1 ⁇ 10 ⁇ 3 or less, i.e. ⁇ log P of 3 or more, is also listed in Tables 5 to 25.
  • Tables 5 to 25 list dbSNP ID number or Affimetrix Array ID number for specifying known single nucleotide polymorphisms obtained, the exon, intron information (in a case where a single nucleotide polymorphism exists on a gene, the gene name and the exon or intron in which SNP exists are shown, and in a case where a single nucleotide polymorphism does not exist on a gene, neighboring genes and a distance between the gene and the single nucleotide polymorphism are shown), the chromosome number at which the single nucleotide polymorphism exists, the physical location of the single nucleotide polymorphism, the p-value for an allele according to a chi-square test ( ⁇ log P), the high-risk allele frequencies in the glaucoma patient group and the non-patient group, the odds ratio for an allele, the p-value for a genotype according to a chi-square test ( ⁇ log
  • Odds Ratio for Allele [(Number of Detection of an Allele Identified in High Frequency in Progressive Glaucoma Group, in Progressive Glaucoma Group)/(Number of Detection of an Allele Opposite to the Allele Identified in High Frequency in Progressive Glaucoma Group, in Progressive Glaucoma Group)]/[(Number of Detection of the Allele Identified in High Frequency in Progressive Glaucoma Group, in Nonprogressive Glaucoma Group)/(Number of Detection of the Allele Opposite to the Allele Identified in High Frequency in Progressive Glaucoma Group, in Nonprogressive Glaucoma Group)] formula (6)
  • Odds Ratio for Genotype of Heterozygote [(Number of Detection of a Genotype of Heterozygote in Progressive Glaucoma Group)/(Number of Detection of a Genotype Having Homozygote of an Allele Identified in High Frequency in Nonprogressive Glaucoma Group, in Progressive Glaucoma Group)]/[(Number of Detection of the Genotype of Heterozygote in Nonprogressive Glaucoma Group)/(Number of Detection of the Genotype Having Homozygote of the Allele Identified in High Frequency in Nonprogressive Glaucoma Group, in Nonprogressive Glaucoma Group)] formula (8)
  • Tables 26 to 28 list dbSNP ID number or Affimetrix Array ID number specifying known single nucleotide polymorphisms obtained, each of bases constituting Allele 1 and Allele 2, the exon, intron information (in a case where a single nucleotide polymorphism exists on a gene, the gene name and the exon or intron in which SNP exists are shown, and in a case where a single nucleotide polymorphism does not exist on a gene, neighboring genes and a distance between the gene and the single nucleotide polymorphism are shown), the chromosome number at which the single nucleotide polymorphism exists, the physical location of the single nucleotide polymorphism, the p-value for an allele according to a chi-square test ( ⁇ log P), the high-risk allele frequencies in the progressive glaucoma group and the nonprogressive glaucoma group, the type of the high-risk allele (indicating whether the
  • the allele or genotype identified in a high frequency in the progressive glaucoma group for a single nucleotide polymorphism listed in Tables 26 to 28 can be used as a marker showing that a progressive risk of glaucoma is high.
  • an allele that is opposite to the allele or a genotype other than the genotype can be used as a marker showing that a progressive risk of glaucoma is low.
  • a single nucleotide polymorphism of which allele or genotype shows association with the progression of glaucoma at a p-value of 1 ⁇ 10 ⁇ 3 or less, i.e. ⁇ log P of 3 or more, is listed in Tables 29 to 51.
  • Odds Frequency Frequency Critical Ratio Ratio Critical in Pro- in Non- Odds rate, (Homo- (Hetero- Chro- rate, gressive progressive Ratio Geno- zygote) zygote) mo- Physical Allele Glaucoma Glaucoma (For- type (For- (For- DBSNP_ID Exon, Intron some Location ( ⁇ logP) Group Group mula 6) ( ⁇ logP) mula 7) mula 8) rs1358105 FLJ32955 +17818bp (NM_153041.1) 2 150,431,710 3.30 0.39 0.27 1.72 2.59 3.20 1.60 rs1724855 FLJ32955 Intron3 (NM_153041.1) 2 150,455,469 3.22 0.43 0.31 1.68 2.56 2.86 1.72 rs17589066 DNAH7 Intron48 (NM_018897.1) 2 196,522,
  • Odds Frequency in Non- Critical Ratio Ratio Critical in Pro- prog- Odds rate, (Homo- (Hetero- Chro- rate, gressive ressive Ratio Geno- zygote) zygote) mo- Physical Allele Glaucoma Glaucoma (For- type (For- (For- DBSNP_ID Exon, Intron some Location ( ⁇ logP) Group Group mula 6) ( ⁇ logP) mula 7) mula 8) rs6440874 LOC152118 ⁇ 101011bp (XM_098163) 3 154,583,971 3.33 0.92 0.83 2.21 2.45 4.99 2.44 rs9852831 LOC152118 ⁇ 133737bp (XM_098163) 3 154,551,245 3.19 0.90 0.82 2.08 2.43 5.05 2.57 rs9822326 LOC339894 Intron2
  • Odds Frequency Frequency Critical Ratio Ratio Critical in Pro- in Non- Odds rate, (Homo- (Hetero- Chro- rate, gressive progressive Ratio Geno- zygote) zygote) mo- Physical Allele Glaucoma Glaucoma (For- type (For- (For- DBSNP_ID Exon, Intron some Location ( ⁇ logP) Group Group mula 6) ( ⁇ logP) mula 7) mula 8) rs2055375 LOC441075 +72079bp (XM_499000) 5 60,566,977 3.56 0.26 0.15 1.97 3.03 7.10 1.84 rs1560026 LOC389319 ⁇ 225968bp (XM_374134) 5 125,497,684 0.82 0.74 0.69 1.26 3.00 0.67 0.33 rs16869864 PTGER4 ⁇ 300343bp (For- type (For- (For- DBSNP_ID Exon,
  • Odds Frequency Frequency Critical Ratio Ratio Critical in Pro- in Non- Odds rate, (Homo- (Hetero- Chro- rate, gressive progressive Ratio Geno- zygote) zygote) mo- Physical Allele Glaucoma Glaucoma (For- type (For- (For- DBSNP_ID Exon, Intron some Location ( ⁇ logP) Group Group mula 6) ( ⁇ logP) mula 7) mula 8) rs13213414 LOC441173 Intron1 (XM_496827) 6 142,262,919 3.60 0.31 0.19 1.87 3.15 2.59 2.17 rs16886390 TMEM30A Intron1 (NM_018247.1) 6 76,038,298 1.18 0.87 0.82 1.47 3.14 0.78 0.27 rs1322867 TBX18 ⁇ 222373bp (XM_496819
  • Odds Frequency Frequency Critical Ratio Ratio Critical in Pro- in Non- Odds rate, (Homo- (Hetero- Chro- rate, gressive progressive Ratio Geno- zygote) zygote) mo- Physical Allele Glaucoma Glaucoma (For- type (For- (For- DBSNP_ID Exon, Intron some Location ( ⁇ logP) Group Group mula 6) ( ⁇ logP) mula 7) 8) rs9403498 FUCA2 ⁇ 18431bp (NM_032020.3) 6 143,892,987 3.04 0.63 0.51 1.63 2.56 2.89 2.01 rs1402405 VMP ⁇ 35853bp (NM_080723.2) 6 24,198,476 3.13 0.58 0.45 1.63 2.53 2.63 1.91 rs7740547 SLC22A16 Intron1 (NM_033125.2) 6
  • Odds Frequency Frequency Critical Ratio Ratio Critical in Pro- in Non- Odds rate, (Homo- (Hetero- Chro- rate, gressive progressive Ratio Geno- zygote) zygote) mo- Physical Allele Glaucoma Glaucoma (For- type (For- (For- DBSNP_ID Exon, Intron some Location ( ⁇ logP) Group Group mula 6) ( ⁇ logP) mula 7) 8) rs7968509 FLJ40126 Intron18 (NM_173599.1), 12 38,541,115 3.46 0.24 0.13 1.99 2.82 7.74 1.66 SLC2A13 Intron6 (NM_052885.1) rs7956512 LOC401725 +199585bp (XM_377278) 12 82,248,022 3.71 0.46 0.32 1.75 2.79 2.82 1.72 rs4768188 FL
  • Odds Frequency Frequency Critical Ratio Ratio Critical in Pro- in Non- Odds rate, (Homo- (Hetero- Chro- rate, gressive progressive Ratio Geno- zygote) zygote) mo- Physical Allele Glaucoma Glaucoma (For- type (For- (For- DBSNP_ID Exon, Intron some Location ( ⁇ logP) Group Group mula 6) ( ⁇ logP) mula 7) mula 8) rs35146 CDH11 Intron12 (NM_001797.2), 16 63,541,382 2.24 0.86 0.79 1.70 3.43 0.76 0.31 CDH11 Intron12 (NM_033664.1) rs35572 LOC390735 ⁇ 442487bp (XM_497515) 16 62,385,041 1.02 0.85 0.81 1.38 3.42 ND ND rs35165 CDH11 +5537
  • Odds Frequency Frequency Critical Ratio Ratio Critical in Pro- in Non- Odds rate, (Homo- (Hetero- Chro- rate, gressive progressive Ratio Geno- zygote) zygote) mo- Physical Allele Glaucoma Glaucoma (For- type (For- (For- DBSNP_ID Exon, Intron some Location ( ⁇ logP) Group Group mula 6) ( ⁇ logP) mula 7) 8) rs35140 CDH11 Intron11 (NM_001797.2), 16 63,548,272 2.03 0.86 0.79 1.64 3.12 0.75 0.32 CDH11 Intron11 (NM_033664.1) rs9925034 A2BP1 Intron2 (NM_018723.2), 16 6,518,170 1.07 0.48 0.41 1.29 3.10 1.40 2.42 A2BP1 ⁇ 804582bp (NM_145891.1),
  • Tables 29 to 51 list dbSNP ID number or Affimetrix Array ID number specifying known single nucleotide polymorphisms obtained, the exon, intron information (in a case where a single nucleotide polymorphism exists on a gene, the gene name and the exon or intron in which SNP exists are shown, and in a case where a single nucleotide polymorphism does not exist on a gene, neighboring genes and a distance between the gene and the single nucleotide polymorphism are shown), the chromosome number at which the single nucleotide polymorphism exists, the physical location of the single nucleotide polymorphism, the p-value for an allele according to a chi-square test ( ⁇ log P), the high-risk allele frequencies in the progressive glaucoma group and the nonprogressive glaucoma group, the odds ratio for an allele, the p-value for a genotype according to a chi-
  • re-sequencing Surrounding sequences of single nucleotide polymorphisms described in Tables 1 to 2 or Tables 26 to 28 are subjected to re-sequencing, so that the detection of a single nucleotide polymorphism can be confirmed, and that an unknown single nucleotide polymorphism that possibly exists can be identified.
  • the re-sequencing can be performed according to any known methods, and for example, the re-sequencing can be performed by a direct sequencing method.
  • an immobilized probe can be prepared.
  • a known single nucleotide polymorphism can be referred to, for example, the database of dbSNP or J SNP.
  • an oligonucleotide probe designed so as to maximize its sensitivity, specificity or reproducibility for several probes to several hundred-thousand probes can be loaded.
  • the immobilized probe can be produced according to a method such as a method of synthesizing an oligonucleotide on a solid carrier or a method including the steps of previously synthesizing an oligonucleotide and immobilizing the oligonucleotide in a high density on a solid carrier.
  • the presence or the absence of the onset of glaucoma can be determined at a more accurate level using the immobilized probe produced in Example 6.
  • a probe for detecting a single nucleotide polymorphism associated with a disease is plurally combined, so that the level of which the onset risk of glaucoma increases is evaluated. In a case where a value exceeds a threshold, it is determined that the onset of glaucoma takes place.
  • the single nucleotide polymorphism existing on the genome of the glaucoma patients and that of the non-glaucoma patients can be compared.
  • single nucleotide polymorphisms existing in locations with an adjacent distance to each other are linked and inherited by linkage disequilibrium.
  • single nucleotide polymorphisms linked with the single nucleotide polymorphisms listed in Tables 1 and 2 or Tables 26 to 28 can be identified by the immobilized probe, so that it can be expected that a single nucleotide polymorphism having an even stronger association with glaucoma is found.
  • candidate single nucleotide polymorphisms associated with the onset of glaucoma identified in the primary analysis of Example 3 were subjected to a secondary analysis of a single nucleotide polymorphism in separately collected samples using an array for analyzing a single nucleotide polymorphism designed in an original style (hereinafter, referred to as a custom array).
  • a kit for analyzing a single nucleotide polymorphism commercially available from Illumina [Illumina, iSelectTM Genotyping BeadChip] was used.
  • Illumina, iSelectTM Genotyping BeadChip For 446 single nucleotide polymorphisms associated with the onset of glaucoma showing a p-value of 1 ⁇ 10 ⁇ 3 or less in Example 3, the designing of a probe for specifically detecting these single nucleotide polymorphisms was tried. Since these probes are randomly immobilized to the substrate via beads, the step of specifying a location of the beads (decoding) is needed.
  • a probe for detecting a single nucleotide polymorphism of which location was unable to be specified in a process of decoding was excluded from the subject for analysis.
  • the preparation of a custom array capable of typing 412 single nucleotide polymorphisms out of 446 single nucleotide polymorphisms is made possible, and the custom array was used in the analysis of a single nucleotide polymorphism described later.
  • Infinium (registered trademark) assay in a beads-array method there are two methods, i.e. a method using one kind of a probe and a method using two kinds of probes. Basically, in the detection of one single nucleotide polymorphism, one kind of the probe was used, and two probes were used for some single nucleotide polymorphisms.
  • the experiment was performed in accordance with the instruction manuals of the custom array kit and the analyzing instrument of Illumina, using specialized reagents contained in the kit. Briefly, the experimental procedures will be explained as follows. A reagent specialized in the treatment of the genome and a sodium hydroxide solution were added to 150 to 300 ng of the total DNA extracted in Example 1. Next, an enzyme for amplifying a whole genome was added thereto, and the mixture was incubated at 37° C. for 20 to 24 hours, and a whole genome was amplified. Further, an enzyme for fragmentation was added thereto, and the mixture was incubated at 37° C. for one hour.
  • an allele-specific extension reaction or a single base extension reaction was performed for each probe, and the fluorescent signals were amplified.
  • the signals were read with a scanner (Illumina, BeadArray Reader) compatible to the kit.
  • a specialized software (Illumina, BeadStudio 3.1) was used in the analysis of the single nucleotide polymorphisms. According to the present analytical method, the opposite alleles of a single nucleotide polymorphism can be determined simultaneously, and the genotypes were determined on the basis of the analytical results.
  • the genotype was determined to be a heterozygote when both the signals of each of the alleles constituting a single nucleotide polymorphism were detected, and the genotype was determined to be a homozygote of the detected allele when only one of the signals of the alleles was detected.
  • genotypes of the single nucleotide polymorphisms that are determined accurately are indicated on the image as three clusters of fluorescent signals that are completely separated from each other (two kinds of homozygotes and a heterozygote).
  • boundary lines of the three clusters become unclear for the single nucleotide polymorphisms that are not determined accurately.
  • the cluster image of the single nucleotide polymorphism was reconfirmed.
  • the sample was excluded from the subsequent analytical operations.
  • the confirmation of the cluster image was carried out under masking, in other words, in a state that the names of single nucleotide polymorphisms and p-values could not be compared with the single nucleotide polymorphisms.
  • the single nucleotide polymorphisms overlapping between the custom array used in the secondary analysis and GeneChip Human Mapping 500K of Affimetrix used in the primary analysis showed a concordance rate of 99% or more, when the concordance rates of the determination of genotypes were compared using 104 samples.
  • Example 9 Blood donated under the consent on free will of the participants after having sufficiently explained the contents of studies from 409 cases of the glaucoma patient group and 448 controls of the non-patient group, each group being different from those of Example 3 was used as a specimen, a total DNA was extracted according to the method described in Example 1, and the analysis of single nucleotide polymorphisms was performed according to the method described in Example 9.
  • the analytical results of a single nucleotide polymorphism obtained in each of the patients were stored in the Laboratory Information Management System (World Fusion, LaboServer) adopting a relational database.
  • a specialized analysis program for a single nucleotide polymorphism was created and loaded within the system, and the analysis described as follows was performed.
  • a single nucleotide polymorphism considered to have a high experimental reliability was extracted by rejecting a single nucleotide polymorphism having a call rate of less than 90% in both the glaucoma patient group and the non-patient group, and a single nucleotide polymorphism having a minor allele frequency of less than 5%.
  • a Mantel-Haenszel chi-square value was determined for the allele model, the dominant genetic model, and the recessive genetic model, and a p-value was calculated by comparing the value with the chi-square distribution of a degree of freedom of 1.
  • the Mantel-Haenszel chi-square value ( ⁇ A MH 2 ) of the allele model was calculated according to the following formulas.
  • the Mantel-Haenszel chi-square value ( ⁇ D MH 2 ) of the dominant genetic model was calculated according to the following formulas.
  • ED i xD i ⁇ mD i / ND i
  • the Mantel-Haenszel chi-square value ( ⁇ R MH 2 ) of the recessive genetic model was calculated according to the following formulas.
  • ER i xR i ⁇ mR i / NR i
  • the odds ratio in the Mantel-Haenszel test was calculated for the allele model, the dominant genetic model, and the recessive genetic model.
  • a 95% confidence interval of the odds ratio in the Mantel-Haenszel test was calculated for the allele model, the dominant genetic model, and the recessive genetic model.
  • the 95% confidence interval (95% CI A ) for the allele model was calculated according to the following formulas.
  • a 95% confidence interval (95% CI d ) for the dominant genetic model was calculated according to the following formulas.
  • PD i aD i + dD i zD i
  • QDi bD i + cD i zD i
  • ⁇ RD i aD i ⁇ dD i zD i
  • ⁇ SD i bD i ⁇ cD i zD i
  • the single nucleotide polymorphisms listed in Tables 53 to 62 can be also used as a marker for predicting an onset risk of glaucoma in the same manner.
  • regions and/or genes of the surrounding of single nucleotide polymorphism listed in Table 52 were determined by making reference to the database provided by the HapMap project.
  • regions in which the single nucleotide polymorphism considered to be in a linkage disequilibrium with the single nucleotide polymorphisms listed in Table 52 exists were determined, on the basis of the linkage disequilibrium data in combination of the Japanese and the Chinese in the HapMap project.
  • Table 52 exists in the linkage disequilibrium region containing genes, the physical location of the region and the gene name were determined. On the other hand, in a case where the single nucleotide polymorphism listed in Table 52 exists in the linkage disequilibrium region without containing the genes, only the physical location of the region was determined. In addition, in a case where the single nucleotide polymorphism listed in Table 52 exists on one gene beyond the linkage disequilibrium region, the gene name and the physical location of the gene were determined.
  • a single nucleotide polymorphism of which p-value is the lowest for each region is considered to be a single nucleotide polymorphism representing the region, and Tables 63 to 70 list a single nucleotide polymorphism representing the region, the chromosome number at which the region exists, the physical location of the region (start point and end point) and the gene name contained in the region.
  • the region listed in Tables 63 to 70 is a region or gene considered to be linked with a single nucleotide polymorphism associated with the onset of glaucoma in the present invention listed in Tables 53 to 62, and a single nucleotide polymorphism which exists in these regions or genes is considered to be linked with a single nucleotide polymorphism in the present invention.
  • any single nucleotide polymorphisms which exist in these regions are linked with the single nucleotide polymorphism which exists in the region listed in Tables 53 to 62, and any of these single nucleotide polymorphisms can be used in the prediction of an onset risk of glaucoma in the same manner.
  • 1/ ⁇ 1+exp[ ⁇ ( ⁇ 0+ ⁇ 1 ⁇ 1+ ⁇ 2 ⁇ 2+ ⁇ 3 ⁇ 3+ . . . )] ⁇
  • a value for risk prediction ( ⁇ ) was calculated by substituting a variable for each single nucleotide polymorphisms into this formula.
  • is greater than 0.5, this sample donor was determined to be with an onset risk.
  • a concordance rate was calculated by comparing the determination results with the matter of whether the sample donor having a single nucleotide polymorphism was actually a glaucoma patient.
  • the concordance rate was determined as mentioned above for each of the incorporated single nucleotide polymorphisms alone, and all the combinations of any two or more single nucleotide polymorphisms, and means and standard deviations of the concordance rate were obtained for each of the number of single nucleotide polymorphisms used in combination.
  • Table 71 lists the number of single nucleotide polymorphisms, alone or in a combination of arbitrary number, the number of combinations when arbitrary number of single nucleotide polymorphism is combined, and the relationship between the mean and the standard deviation of the concordance rate.
  • all the calculations were performed using SAS 9.1.3, Windows (registered trademark) Edition, SAS Institute Japan Corporation.
  • a value for risk prediction ( ⁇ ) of individual cases was calculated using a logistic regression formula, alone or in a combination of any two or more of these 10 single nucleotide polymorphisms.
  • a cut-off value for a value for risk prediction is defined as 0.5
  • mean ⁇ standard deviation of the concordance rate was 54.7 ⁇ 1.4% in a case that each of the single nucleotide polymorphisms was used alone.
  • This concordance rate was elevated as an increase in the number of single nucleotide polymorphisms used in combination, and reached the maximum of 59.9% in a case that all the ten were combined.
  • an individual who has an allele or a genotype that is identified in a high frequency in the glaucoma patients disclosed in the present invention on the genome has a high onset risk of glaucoma in future, and an individual who does not have an allele or a genotype that is identified in a high frequency in the glaucoma patients has a low onset risk of glaucoma in future.
  • the level of an onset risk of glaucoma of a sample donor can be determined by analyzing an allele or a genotype of a single nucleotide polymorphism in the present invention in a sample.
  • a sample donor can take a preventive measure of glaucoma, or can receive appropriate treatments, on the basis of this risk.
  • a sample donor who has an allele or a genotype that is identified in a high frequency in the glaucoma patients of a single nucleotide polymorphism in the present invention on the genome can be given a precision examination in whether or not the donor is with an early glaucoma which is difficult to be determined sufficiently by an intraocular pressure or an ocular fundus photograph, and can be started with a treatment at an early stage in a case where the donor is diagnosed as glaucoma.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
US12/596,258 2007-04-17 2008-04-17 Method for determination of onset risk of glaucoma Abandoned US20100196895A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2007108688 2007-04-17
JP2007-108688 2007-04-17
PCT/JP2008/057527 WO2008130008A1 (ja) 2007-04-17 2008-04-17 緑内障発症リスクの判定方法

Publications (1)

Publication Number Publication Date
US20100196895A1 true US20100196895A1 (en) 2010-08-05

Family

ID=39875536

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/596,258 Abandoned US20100196895A1 (en) 2007-04-17 2008-04-17 Method for determination of onset risk of glaucoma
US12/596,462 Expired - Fee Related US8431345B2 (en) 2007-04-17 2008-04-17 Method for determination of progression risk of glaucoma
US13/546,674 Abandoned US20130012408A1 (en) 2007-04-17 2012-07-11 Method for determination of onset risk of glaucoma
US13/850,453 Abandoned US20130210668A1 (en) 2007-04-17 2013-03-26 Method for determination of progression risk of glaucoma

Family Applications After (3)

Application Number Title Priority Date Filing Date
US12/596,462 Expired - Fee Related US8431345B2 (en) 2007-04-17 2008-04-17 Method for determination of progression risk of glaucoma
US13/546,674 Abandoned US20130012408A1 (en) 2007-04-17 2012-07-11 Method for determination of onset risk of glaucoma
US13/850,453 Abandoned US20130210668A1 (en) 2007-04-17 2013-03-26 Method for determination of progression risk of glaucoma

Country Status (12)

Country Link
US (4) US20100196895A1 (ja)
EP (4) EP2161334A4 (ja)
JP (5) JP5624763B2 (ja)
KR (2) KR20100016568A (ja)
CN (2) CN101679971A (ja)
AU (2) AU2008241867A1 (ja)
BR (2) BRPI0810425A2 (ja)
CA (2) CA2683836A1 (ja)
MX (2) MX2009011080A (ja)
RU (2) RU2009142223A (ja)
SG (2) SG177968A1 (ja)
WO (2) WO2008130008A1 (ja)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9684959B2 (en) 2012-08-24 2017-06-20 Agency For Science, Technology And Research Methods and systems for automatic location of optic structures in an image of an eye, and for automatic retina cup-to-disc ratio computation
US10648035B2 (en) 2012-11-26 2020-05-12 The Johns Hopkins University Methods and compositions for diagnosing and treating gastric cancer
US20220392639A1 (en) * 2019-10-31 2022-12-08 Google Llc Using Machine Learning-Based Trait Predictions For Genetic Association Discovery

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG158776A1 (en) * 2008-07-25 2010-02-26 Univ Kaohsiung Medical Using genetic polymorphisms of the bicd1 gene as a method for diagnosing and treating myopia
JP4515524B2 (ja) * 2008-11-27 2010-08-04 株式会社メニコン 正常眼圧緑内障疾患感受性遺伝子及びその利用
US10206813B2 (en) 2009-05-18 2019-02-19 Dose Medical Corporation Implants with controlled drug delivery features and methods of using same
AU2010267835C1 (en) * 2009-07-03 2015-02-26 University College Cardiff Consultants Limited Diagnosis and treatment of Alzheimer's disease
WO2011059409A1 (en) * 2009-11-16 2011-05-19 Jiang Liu Obtaining data for automatic glaucoma screening, and screening and diagnostic techniques and systems using the data
WO2011152098A1 (ja) * 2010-06-04 2011-12-08 コニカミノルタエムジー株式会社 イムノクロマトグラフ法のための展開液およびそれを用いた測定法
US9708664B2 (en) 2012-04-20 2017-07-18 University Of Virginia Patent Foundation Compositions and methods for identifying and diagnosing salt sensitivity of blood pressure
RU2508043C1 (ru) * 2012-12-20 2014-02-27 федеральное государственное бюджетное учреждение "Межотраслевой научно-технический комплекс "Микрохирургия глаза" имени академика С.Н. Федорова" Министерства здравоохранения Российской Федерации Способ прогнозирования развития открытоугольной глаукомы у пациентов с глазными проявлениями псевдоэксфолиативного синдрома
EP3140422A1 (en) * 2014-05-03 2017-03-15 The Regents of The University of California Methods of identifying biomarkers associated with or causative of the progression of disease, in particular for use in prognosticating primary open angle glaucoma
RU2580306C1 (ru) * 2015-03-26 2016-04-10 Федеральное государственное автономное образовательное учреждение высшего профессионального образования "Белгородский государственный национальный исследовательский университет" (НИУ "БелГУ") Способ прогнозирования риска развития первичной открытоугольной глаукомы у индивидуумов в зависимости от наличия/отсутствия сопутствующих неинфекционных заболеваний глаз
RU2585381C1 (ru) * 2015-03-27 2016-05-27 Федеральное государственное автономное образовательное учреждение высшего профессионального образования "Белгородский государственный национальный исследовательский университет" (НИУ "БелГУ") Способ прогнозирования показателей гидродинамики у больных первичной открытоугольной глаукомой
JP6542037B2 (ja) * 2015-06-04 2019-07-10 シスメックス株式会社 関節リウマチ患者におけるメトトレキサートの有効性の診断を補助する方法
WO2018003523A1 (ja) * 2016-06-30 2018-01-04 京都府公立大学法人 広義原発開放隅角緑内障の発症リスクの判定方法
JP7064215B2 (ja) * 2016-08-05 2022-05-10 京都府公立大学法人 落屑症候群又は落屑緑内障の発症リスクの判定方法
CN113444788B (zh) * 2021-08-27 2021-11-02 中国医学科学院北京协和医院 青光眼诊断产品及应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5474796A (en) * 1991-09-04 1995-12-12 Protogene Laboratories, Inc. Method and apparatus for conducting an array of chemical reactions on a support surface
US20010053519A1 (en) * 1990-12-06 2001-12-20 Fodor Stephen P.A. Oligonucleotides
US20060073506A1 (en) * 2004-09-17 2006-04-06 Affymetrix, Inc. Methods for identifying biological samples

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5686407A (en) * 1995-08-16 1997-11-11 Alcon Laboratories, Inc. Hepatocyte growth factor to treat glaucoma
US5962230A (en) 1997-02-13 1999-10-05 The University Of Connecticut Diagnosis and treatment of glaucoma
JP2000306165A (ja) 1999-02-19 2000-11-02 Toshiba Tec Corp 医療費控除用レシート発行装置および記録媒体
JP2002306165A (ja) 2000-05-17 2002-10-22 Tsubota:Kk 正常眼圧緑内障を含む開放隅角緑内障の関連遺伝子
JP5129563B2 (ja) * 2004-03-18 2013-01-30 ラスク・インテレクチュアル・リザーブ・アクチェンゲゼルシャフト 視神経症に対する感受性を診断または予測するための方法
US20100129799A1 (en) 2006-10-27 2010-05-27 Decode Genetics Ehf. Cancer susceptibility variants on chr8q24.21
WO2009061890A1 (en) * 2007-11-08 2009-05-14 St. Jude Children's Research Hospital Methods and compositions for the diagnosis, prognosis and treatment of chronic myeloid leukemia and acute lymphoblastic leukemia

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20010053519A1 (en) * 1990-12-06 2001-12-20 Fodor Stephen P.A. Oligonucleotides
US5474796A (en) * 1991-09-04 1995-12-12 Protogene Laboratories, Inc. Method and apparatus for conducting an array of chemical reactions on a support surface
US20060073506A1 (en) * 2004-09-17 2006-04-06 Affymetrix, Inc. Methods for identifying biological samples

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DB sNP ss718868(submitted July 27, 2000, www.ncbi.nlm.nih.gov/projects/SNP/snp_ss.cgi?subsnp_id=718868, downloaded 1/5/2012) *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9684959B2 (en) 2012-08-24 2017-06-20 Agency For Science, Technology And Research Methods and systems for automatic location of optic structures in an image of an eye, and for automatic retina cup-to-disc ratio computation
US10648035B2 (en) 2012-11-26 2020-05-12 The Johns Hopkins University Methods and compositions for diagnosing and treating gastric cancer
US20220392639A1 (en) * 2019-10-31 2022-12-08 Google Llc Using Machine Learning-Based Trait Predictions For Genetic Association Discovery

Also Published As

Publication number Publication date
US20110207122A1 (en) 2011-08-25
JP5759500B2 (ja) 2015-08-05
KR20100016568A (ko) 2010-02-12
JP6203217B2 (ja) 2017-09-27
SG177960A1 (en) 2012-02-28
CN101679970A (zh) 2010-03-24
WO2008130009A1 (ja) 2008-10-30
JP2013150628A (ja) 2013-08-08
SG177968A1 (en) 2012-02-28
AU2008241868A1 (en) 2008-10-30
EP2161334A1 (en) 2010-03-10
EP2147975A4 (en) 2010-11-10
US20130210668A1 (en) 2013-08-15
WO2008130008A1 (ja) 2008-10-30
CA2683836A1 (en) 2008-10-30
JPWO2008130008A1 (ja) 2010-07-22
RU2009142225A (ru) 2011-05-27
JP2015180219A (ja) 2015-10-15
KR20100016525A (ko) 2010-02-12
MX2009011079A (es) 2010-03-10
AU2008241867A1 (en) 2008-10-30
CN101679971A (zh) 2010-03-24
BRPI0810071A2 (pt) 2014-10-14
MX2009011080A (es) 2010-03-10
JP2017201995A (ja) 2017-11-16
US20130012408A1 (en) 2013-01-10
EP2161334A4 (en) 2010-11-10
EP2147975A1 (en) 2010-01-27
RU2009142223A (ru) 2011-05-27
JP5624763B2 (ja) 2014-11-12
EP2548961A1 (en) 2013-01-23
CA2683691A1 (en) 2008-10-30
US8431345B2 (en) 2013-04-30
EP2565270A1 (en) 2013-03-06
JPWO2008130009A1 (ja) 2010-07-22
BRPI0810425A2 (pt) 2014-10-07

Similar Documents

Publication Publication Date Title
US8431345B2 (en) Method for determination of progression risk of glaucoma
US10407738B2 (en) Markers for breast cancer
US8420315B2 (en) Single nucleotide polymorphisms and combinations of novel and known polymorphisms for determining the allele-specific expression of the IGF2 gene
EP2393939B1 (en) A snp marker of breast and ovarian cancer risk
AU2013203426B2 (en) Markers for breast cancer
AU2012202265B2 (en) Markers for breast cancer
KR20240057772A (ko) 우울증 진단용 단일염기다형성 및 이의 용도
Liu Identification of genetic and epigenetic risk factors for psoriasis and psoratic arthritis

Legal Events

Date Code Title Description
AS Assignment

Owner name: SANTEN PHARMACEUTICAL CO., LTD., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KINOSHITA, SHIGERU;TASHIRO, KEI;NAKANO, MASAKAZU;AND OTHERS;SIGNING DATES FROM 20090819 TO 20090825;REEL/FRAME:023400/0197

Owner name: KEI TASHIRO, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KINOSHITA, SHIGERU;TASHIRO, KEI;NAKANO, MASAKAZU;AND OTHERS;SIGNING DATES FROM 20090819 TO 20090825;REEL/FRAME:023400/0197

Owner name: SHIGERU KINOSHITA, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KINOSHITA, SHIGERU;TASHIRO, KEI;NAKANO, MASAKAZU;AND OTHERS;SIGNING DATES FROM 20090819 TO 20090825;REEL/FRAME:023400/0197

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION