US20110236370A1 - Genetic Alterations on Chromosomes 21q, 6q and 15q and Methods of Use Thereof for the Diagnosis and Treatment of Type I Diabetes - Google Patents

Genetic Alterations on Chromosomes 21q, 6q and 15q and Methods of Use Thereof for the Diagnosis and Treatment of Type I Diabetes Download PDF

Info

Publication number
US20110236370A1
US20110236370A1 US12/947,564 US94756410A US2011236370A1 US 20110236370 A1 US20110236370 A1 US 20110236370A1 US 94756410 A US94756410 A US 94756410A US 2011236370 A1 US2011236370 A1 US 2011236370A1
Authority
US
United States
Prior art keywords
seq
nucleic acid
gene
cells
diabetes
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
US12/947,564
Inventor
Hakon Hakonarson
Struan Grant
Jonathan Bradfield
Constantin Polychronakos
Hui-Qi Qu
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.)
Childrens Hospital of Philadelphia CHOP
Original Assignee
Childrens Hospital of Philadelphia CHOP
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 Childrens Hospital of Philadelphia CHOP filed Critical Childrens Hospital of Philadelphia CHOP
Priority to US12/947,564 priority Critical patent/US20110236370A1/en
Assigned to THE CHILDREN'S HOSPITAL OF PHILADELPHIA reassignment THE CHILDREN'S HOSPITAL OF PHILADELPHIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MCGILL UNIVERSITY
Publication of US20110236370A1 publication Critical patent/US20110236370A1/en
Assigned to THE CHILDREN'S HOSPITAL OF PHILADELPHIA reassignment THE CHILDREN'S HOSPITAL OF PHILADELPHIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRADFIELD, JONATHAN, GRANT, STRUAN, HAKONARSON, HAKON
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: CHILDREN'S HOSPITAL OF PHILADELPHIA
Priority to US14/628,147 priority patent/US10066266B2/en
Assigned to NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR reassignment NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: CHILDREN'S HOSPITAL OF PHILADELPHIA
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • C12Q2535/00Reactions characterised by the assay type for determining the identity of a nucleotide base or a sequence of oligonucleotides
    • C12Q2535/131Allele specific probes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/172Haplotypes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/14Heterocyclic carbon compound [i.e., O, S, N, Se, Te, as only ring hetero atom]
    • Y10T436/142222Hetero-O [e.g., ascorbic acid, etc.]
    • Y10T436/143333Saccharide [e.g., DNA, etc.]

Definitions

  • This invention relates to the fields of glucose metabolism, genetics and pathology associated with diabetes, particularly type I diabetes. More specifically, the invention provides a panel of genes containing genetic alterations, e.g., single nucleotide polymorphisms, which had heretofore not been associated with this disease. Methods and kits for using the sequences so identified for diagnostic and therapeutic treatment purposes are also provided, as are therapeutic compositions for management of diabetes.
  • Type I diabetes results from the autoimmune destruction of pancreatic beta cells, a process believed to be strongly influenced by multiple genes and environmental factors.
  • the incidence of T1D has been increasing in Western countries and has more than doubled in the United States over the past 30 years.
  • the disease shows a strong familial component, with first-degree relatives of cases being at 15 times greater risk of T1D than a randomly selected member of the general population and monozygotic twins being concordant for T1D at a frequency of approximately 50%.
  • the genetic evidence is strong, the latter data suggests that an interplay with environmental factors also plays a key role in influencing T1D outcome.
  • the familial clustering of T1D is influenced by multiple genes. Variation in four loci has already been established to account for a significant proportion of the familial aggregation of T1D. These include the major histocompatibility complex (MHC) region on 6p21 (including the HLA-DRBJ, -DQA1 and -DRQJ genes 1 ); the insulin/insulin-like growth factor 2 gene complex (INS-IGF2) on 11p15 24 , the protein tyrosine phosphatase-22 (PTPN22) gene on 1p13 5, 6 and the gene encoding cytotoxic T-lymphocyte-associated protein 4 (CTLA4) on 2q31 7, 8 .
  • MHC major histocompatibility complex
  • INS-IGF2 insulin/insulin-like growth factor 2 gene complex
  • PTPN22 protein tyrosine phosphatase-22
  • the interleukin-2 receptor alpha (CD25) locus on 10p15 9 has also been implicated in the pathogenesis of T1D but remains to be replicated by independent studies.
  • spontaneous mouse model studies of T1D have implicated numerous other regions that have been confirmed in replication studies 10 .
  • Several other loci have also been implicated in human association studies with T1D but the effects of these implicated genes remain controversial and are subject to confirmation in independent studies utilizing sufficient sample sizes. Together, these studies suggest that many more T1D susceptibility genes remain to be discovered.
  • T1D-associated SNPs have been identified which are indicative of an increased or reduced risk of developing T1D.
  • nucleic acids comprising at least one genetic alteration identified in Tables 1, 2, 4 and 5 are provided. Such nucleic acids and the proteins encoded thereby have utility in the diagnosis and management of type 1 diabetes (T1D).
  • An exemplary method entails providing a target nucleic acid from a patient sample, said target nucleic acid having a predetermined sequence in the normal population, and assessing said target nucleic acid for the presence of at least one genetic alteration, e.g., a single nucleotide polymorphism, which is indicative of an increased or decreased risk of developing T1D.
  • genetic alteration e.g., a single nucleotide polymorphism
  • the genetic alteration is a single nucleotide polymorphism present in UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7 encoding nucleic acids and genetic regions associated therewith.
  • Such genetic regions include the linkage disequilibrium blocks provided in Table 3 and the method entails detecting any variant associated with diabetes in such blocks.
  • the SNP is rs9976767 present on chromosome 21 at position 42709459 within the UBASH3A gene
  • the SNP is rs3757247 present on chromosome 6 at position 91014184 in the BACH2 gene
  • the SNP is rs7171171 at position 36694333 on chromosome 15 in the RASGRP1 gene.
  • the methods of the invention also include the detection of any of the T1D associated genetic alterations comprising the single nucleotide polymorphisms set forth in Tables 1, 2, 4 or 5 for the diagnosis of T1D.
  • genetic alterations associated with T1D present in the linkage disequilibrium blocks set forth in Table 3 can be detected. Kits and microarrays for practicing the foregoing methods are also provided.
  • a method of managing T1D which entails administering a therapeutic agent to a patient in need thereof.
  • the therapeutic agent can be a small molecule, an antibody, a protein, an oligonucleotide, or a siRNA molecule.
  • a method for identifying agents that bind and/or modulate UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7 functional activity is provided, as well as pharmaceutical compositions comprising said agent in a biologically acceptable carrier.
  • FIG. 1 The LD plot of the RASGRP1 SNPs, based on the HapMap European data.
  • the top panel shows the constrained elements and conservation scores called by the DNA sequence alignments of 29 eutherian mammals (Ensembl, on the world wide web at .ensembl.org. See Cooper et al. Genome Research 2005; 15:901-913.
  • the LD map is made by Haploview v4.0 software available on the world wide web at road.mit.edu/personal/jcbarret/haploview. D′ values (%) are shown in the boxes, and r 2 values are represented by the grey scale.
  • the red arrows highlight the SNPs genotyped in this study. Inside the red circle, it is the SNP described in Example I.
  • FIG. 2 BACH-2 related protein protein interaction network of differentially expressed genes for T1D.
  • FIG. 3 EDG7/LPAR3 related protein protein interaction network of differentially expressed genes for T1D.
  • Type 1 diabetes is a common and strongly heritable disease that most often manifests in childhood. Recent genome wide association studies have revealed a number of new genes associated with the disease. We carried out a follow-up strategy to our T1D GWA study in an attempt to uncover additional novel T1D risk loci.
  • SNPs single nucleotide polymorphisms
  • T1D probands from the DCCT/EDIC study including 1,303 T1D patients using an independent matched control dataset of diabetes free individuals from Philadelphia which were genotyped on the 1M and HumanHap550K SNP BeadChips, respectively.
  • a or “an” entity refers to one or more of that entity; for example, “a cDNA” refers to one or more cDNA or at least one cDNA.
  • a cDNA refers to one or more cDNA or at least one cDNA.
  • the terms “a” or “an,” “one or more” and “at least one” can be used interchangeably herein.
  • the terms “comprising,” “including,” and “having” can be used interchangeably.
  • a compound “selected from the group consisting of refers to one or more of the compounds in the list that follows, including mixtures (i.e. combinations) of two or more of the compounds.
  • an isolated, or biologically pure molecule is a compound that has been removed from its natural milieu. As such, Aisolated@ and Abiologically pure@ do not necessarily reflect the extent to which the compound has been purified.
  • An isolated compound of the present invention can be obtained from its natural source, can be produced using laboratory synthetic techniques or can be produced by any such chemical
  • SNP single nucleotide polymorphism
  • genetic alteration refers to a change from the wild-type or reference sequence of one or more nucleic acid molecules. Genetic alterations include without limitation, base pair substitutions, additions and deletions of at least one nucleotide from a nucleic acid molecule of known sequence.
  • Type 1 diabetes refers to a chronic (lifelong) disease that occurs when the pancreas produces too little insulin to regulate blood sugar levels appropriately.
  • T1D often called juvenile or insulin-dependent diabetes results from altered metabolism of carbohydrates (including sugars such as glucose), proteins, and fats.
  • the beta cells of the pancreas produce little or no insulin, the hormone that allows glucose to enter body cells. Once glucose enters a cell, it is used as fuel. Without adequate insulin, glucose builds up in the bloodstream instead of going into the cells. The body is unable to use this glucose for energy despite high levels in the bloodstream, leading to increased hunger.
  • the high levels of glucose in the blood cause the patient to urinate more, which in turn causes excessive thirst.
  • the insulin-producing beta cells of the pancreas are completely destroyed, and no more insulin is produced.
  • T1D-associated SNP or specific marker is a SNP or marker which is associated with an increased or decreased risk of developing TID not found normal patients who do not have this disease. Such markers may include but are not limited to nucleic acids, proteins encoded thereby, or other small molecules.
  • Type 1 diabetes can occur at any age, but it usually starts in people younger than 30. Symptoms are usually severe and occur rapidly. The exact cause of type 1 diabetes is not known. Type 1 diabetes accounts for 3% of all new cases of diabetes each year. There is 1 new case per every 7,000 children per year. New cases are less common among adults older than 20.
  • solid matrix refers to any format, such as beads, microparticles, a microarray, the surface of a microtitration well or a test tube, a dipstick or a filter.
  • the material of the matrix may be polystyrene, cellulose, latex, nitrocellulose, nylon, polyacrylamide, dextran or agarose.
  • Sample or “patient sample” or “biological sample” generally refers to a sample which may be tested for a particular molecule, preferably an T1D specific marker molecule, such as a marker shown in the tables provided below. Samples may include but are not limited to cells, body fluids, including blood, serum, plasma, urine, saliva, tears, pleural fluid and the like.
  • phrases “consisting essentially of when referring to a particular nucleotide or amino acid means a sequence having the properties of a given SEQ ID NO.
  • the phrase when used in reference to an amino acid sequence, the phrase includes the sequence per se and molecular modifications that would not affect the functional and novel characteristics of the sequence.
  • Linkage describes the tendency of genes, alleles, loci or genetic markers to be inherited together as a result of their location on the same chromosome, and is measured by percent recombination (also called recombination fraction, or ⁇ ) between the two genes, alleles, loci or genetic markers. The closer two loci physically are on the chromosome, the lower the recombination fraction will be. Normally, when a polymorphic site from within a disease-causing gene is tested for linkage with the disease, the recombination fraction will be zero, indicating that the disease and the disease-causing gene are always co-inherited.
  • “Centimorgan” is a unit of genetic distance signifying linkage between two genetic markers, alleles, genes or loci, corresponding to a probability of recombination between the two markers or loci of 1% for any meiotic event.
  • Linkage disequilibrium or “allelic association” means the preferential association of a particular allele, locus, gene or genetic marker with a specific allele, locus, gene or genetic marker at a nearby chromosomal location more frequently than expected by chance for any particular allele frequency in the population.
  • Target nucleic acid refers to a previously defined region of a nucleic acid present in a complex nucleic acid mixture wherein the defined wild-type region contains at least one known nucleotide variation which may or may not be associated with T1D.
  • the nucleic acid molecule may be isolated from a natural source by cDNA cloning or subtractive hybridization or synthesized manually.
  • the nucleic acid molecule may be synthesized manually by the triester synthetic method or by using an automated DNA synthesizer.
  • the term “isolated nucleic acid” is sometimes employed. This term, when applied to DNA, refers to a DNA molecule that is separated from sequences with which it is immediately contiguous (in the 5′ and 3′ directions) in the naturally occurring genome of the organism from which it was derived.
  • the “isolated nucleic acid” may comprise a DNA molecule inserted into a vector, such as a plasmid or virus vector, or integrated into the genomic DNA of a prokaryote or eukaryote.
  • An “isolated nucleic acid molecule” may also comprise a cDNA molecule.
  • An isolated nucleic acid molecule inserted into a vector is also sometimes referred to herein as a recombinant nucleic acid molecule.
  • isolated nucleic acid primarily refers to an RNA molecule encoded by an isolated DNA molecule as defined above.
  • the term may refer to an RNA molecule that has been sufficiently separated from RNA molecules with which it would be associated in its natural state (i.e., in cells or tissues), such that it exists in a “substantially pure” form.
  • enriched in reference to nucleic acid it is meant that the specific DNA or RNA sequence constitutes a significantly higher fraction (2-5 fold) of the total DNA or RNA present in the cells or solution of interest than in normal cells or in the cells from which the sequence was taken.
  • nucleotide sequence be in purified form.
  • purified in reference to nucleic acid does not require absolute purity (such as a homogeneous preparation); instead, it represents an indication that the sequence is relatively purer than in the natural environment (compared to the natural level, this level should be at least 2-5 fold greater, e.g., in terms of mg/ml).
  • Individual clones isolated from a cDNA library may be purified to electrophoretic homogeneity.
  • the claimed DNA molecules obtained from these clones can be obtained directly from total DNA or from total RNA.
  • the cDNA clones are not naturally occurring, but rather are preferably obtained via manipulation of a partially purified naturally occurring substance (messenger RNA).
  • a cDNA library from mRNA involves the creation of a synthetic substance (cDNA) and pure individual cDNA clones can be isolated from the synthetic library by clonal selection of the cells carrying the cDNA library.
  • the process which includes the construction of a cDNA library from mRNA and isolation of distinct cDNA clones yields an approximately 10 ⁇ 6 -fold purification of the native message.
  • purification of at least one order of magnitude, preferably two or three orders, and more preferably four or five orders of magnitude is expressly contemplated.
  • the term “substantially pure” refers to a preparation comprising at least 50-60% by weight the compound of interest (e.g., nucleic acid, oligonucleotide, etc.). More preferably, the preparation comprises at least 75% by weight, and most preferably 90-99% by weight, the compound of interest. Purity is measured by methods appropriate for the compound of interest.
  • complementary describes two nucleotides that can form multiple favorable interactions with one another.
  • adenine is complementary to thymine as they can form two hydrogen bonds.
  • guanine and cytosine are complementary since they can form three hydrogen bonds.
  • a “complement” of this nucleic acid molecule would be a molecule containing adenine in the place of thymine, thymine in the place of adenine, cytosine in the place of guanine, and guanine in the place of cytosine.
  • the complement can contain a nucleic acid sequence that forms optimal interactions with the parent nucleic acid molecule, such a complement can bind with high affinity to its parent molecule.
  • the term “specifically hybridizing” refers to the association between two single-stranded nucleotide molecules of sufficiently complementary sequence to permit such hybridization under pre-determined conditions generally used in the art (sometimes termed “substantially complementary”).
  • the term refers to hybridization of an oligonucleotide with a substantially complementary sequence contained within a single-stranded DNA or RNA molecule of the invention, to the substantial exclusion of hybridization of the oligonucleotide with single-stranded nucleic acids of non-complementary sequence.
  • specific hybridization can refer to a sequence which hybridizes to any T1D specific marker gene or nucleic acid, but does not hybridize to other human nucleotides.
  • polynucleotide which Aspecifically hybridizes@ may hybridize only to a T1D specific marker, such a T1D-specific marker shown in Tables 1-3. Appropriate conditions enabling specific hybridization of single stranded nucleic acid molecules of varying complementarity are well known in the art.
  • T m 81.5′′ C+16.6 Log[Na+]+0.41(% G+C ) ⁇ 0.63 (% formamide) ⁇ 600/#bp in duplex
  • the T m is 57′′ C.
  • the T m of a DNA duplex decreases by 1-1.5′′ C with every 1% decrease in homology.
  • targets with greater than about 75% sequence identity would be observed using a hybridization temperature of 42′′ C.
  • the stringency of the hybridization and wash depend primarily on the salt concentration and temperature of the solutions. In general, to maximize the rate of annealing of the probe with its target, the hybridization is usually carried out at salt and temperature conditions that are 20-25° C. below the calculated T m of the hybrid. Wash conditions should be as stringent as possible for the degree of identity of the probe for the target. In general, wash conditions are selected to be approximately 12-20° C. below the T m of the hybrid.
  • a moderate stringency hybridization is defined as hybridization in 6 ⁇ SSC, 5 ⁇ Denhardt's solution, 0.5% SDS and 100 ⁇ g/ml denatured salmon sperm DNA at 42° C., and washed in 2 ⁇ SSC and 0.5% SDS at 55° C. for 15 minutes.
  • a high stringency hybridization is defined as hybridization in 6 ⁇ SSC, 5 ⁇ Denhardt's solution, 0.5% SDS and 100 ⁇ g/ml denatured salmon sperm DNA at 42° C., and washed in 1 ⁇ SSC and 0.5% SDS at 65° C. for 15 minutes.
  • a very high stringency hybridization is defined as hybridization in 6 ⁇ SSC, 5 ⁇ Denhardt's solution, 0.5% SDS and 100 ⁇ g/ml denatured salmon sperm DNA at 42° C., and washed in 0.1 ⁇ SSC and 0.5% SDS at 65° C. for 15 minutes.
  • oligonucleotide or “oligo” as used herein means a short sequence of DNA or DNA derivatives typically 8 to 35 nucleotides in length, primers, or probes.
  • An oligonucleotide can be derived synthetically, by cloning or by amplification.
  • An oligo is defined as a nucleic acid molecule comprised of two or more ribo- or deoxyribonucleotides, preferably more than three. The exact size of the oligonucleotide will depend on various factors and on the particular application and use of the oligonucleotide.
  • derivative is intended to include any of the above described variants when comprising an additional chemical moiety not normally a part of these molecules. These chemical moieties can have varying purposes including, improving solubility, absorption, biological half life, decreasing toxicity and eliminating or decreasing undesirable side effects.
  • probe refers to an oligonucleotide, polynucleotide or nucleic acid, either RNA or DNA, whether occurring naturally as in a purified restriction enzyme digest or produced synthetically, which is capable of annealing with or specifically hybridizing to a nucleic acid with sequences complementary to the probe.
  • a probe may be either single-stranded or double-stranded. The exact length of the probe will depend upon many factors, including temperature, source of probe and use of the method. For example, for diagnostic applications, depending on the complexity of the target sequence, the oligonucleotide probe typically contains 15-25 or more nucleotides, although it may contain fewer nucleotides.
  • the probes herein are selected to be complementary to different strands of a particular target nucleic acid sequence. This means that the probes must be sufficiently complementary so as to be able to “specifically hybridize” or anneal with their respective target strands under a set of pre-determined conditions. Therefore, the probe sequence need not reflect the exact complementary sequence of the target. For example, a non-complementary nucleotide fragment may be attached to the 5′ or 3′ end of the probe, with the remainder of the probe sequence being complementary to the target strand. Alternatively, non-complementary bases or longer sequences can be interspersed into the probe, provided that the probe sequence has sufficient complementarity with the sequence of the target nucleic acid to anneal therewith specifically.
  • primer refers to an oligonucleotide, either RNA or DNA, either single-stranded or double-stranded, either derived from a biological system, generated by restriction enzyme digestion, or produced synthetically which, when placed in the proper environment, is able to functionally act as an initiator of template-dependent nucleic acid synthesis.
  • suitable nucleoside triphosphate precursors of nucleic acids, a polymerase enzyme, suitable cofactors and conditions such as a suitable temperature and pH
  • the primer may be extended at its 3′ terminus by the addition of nucleotides by the action of a polymerase or similar activity to yield a primer extension product.
  • the primer may vary in length depending on the particular conditions and requirement of the application.
  • the oligonucleotide primer is typically 15-25 or more nucleotides in length.
  • the primer must be of sufficient complementarity to the desired template to prime the synthesis of the desired extension product, that is, to be able anneal with the desired template strand in a manner sufficient to provide the 3′ hydroxyl moiety of the primer in appropriate juxtaposition for use in the initiation of synthesis by a polymerase or similar enzyme. It is not required that the primer sequence represent an exact complement of the desired template.
  • a non-complementary nucleotide sequence may be attached to the 5′ end of an otherwise complementary primer.
  • non-complementary bases may be interspersed within the oligonucleotide primer sequence, provided that the primer sequence has sufficient complementarity with the sequence of the desired template strand to functionally provide a template-primer complex for the synthesis of the extension product.
  • PCR Polymerase chain reaction
  • siRNA refers to a molecule involved in the RNA interference process for a sequence-specific post-transcriptional gene silencing or gene knockdown by providing small interfering RNAs (siRNAs) that has homology with the sequence of the targeted gene.
  • small interfering RNAs can be synthesized in vitro or generated by ribonuclease III cleavage from longer dsRNA and are the mediators of sequence-specific mRNA degradation.
  • the siRNA of the invention are chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional DNA/RNA synthesizer.
  • the siRNA can be synthesized as two separate, complementary RNA molecules, or as a single RNA molecule with two complementary regions.
  • Commercial suppliers of synthetic RNA molecules or synthesis reagents include Applied Biosystems (Foster City, Calif., USA), Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, Colo., USA), Pierce Chemical (part of Perbio Science, Rockford, Ill., USA), Glen Research (Sterling, Va., USA), ChemGenes (Ashland, Mass., USA) and Cruachem (Glasgow, UK).
  • siRNA constructs for inhibiting UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7 mRNA may be between 15-35 nucleotides in length, and more typically about 21 nucleotides in length.
  • Exemplary siRNA molecules which downregulate the aforementioned gene targets are provided in Tables 6-10.
  • vector relates to a single or double stranded circular nucleic acid molecule that can be infected, transfected or transformed into cells and replicate independently or within the host cell genome.
  • a circular double stranded nucleic acid molecule can be cut and thereby linearized upon treatment with restriction enzymes.
  • restriction enzymes An assortment of vectors, restriction enzymes, and the knowledge of the nucleotide sequences that are targeted by restriction enzymes are readily available to those skilled in the art, and include any replicon, such as a plasmid, cosmid, bacmid, phage or virus, to which another genetic sequence or element (either DNA or RNA) may be attached so as to bring about the replication of the attached sequence or element.
  • a nucleic acid molecule of the invention can be inserted into a vector by cutting the vector with restriction enzymes and ligating the two pieces together.
  • transformation transformation
  • transfection transduction
  • Atransduction@ refer to methods of inserting a nucleic acid and/or expression construct into a cell or host organism. These methods involve a variety of techniques, such as treating the cells with high concentrations of salt, an electric field, or detergent, to render the host cell outer membrane or wall permeable to nucleic acid molecules of interest, microinjection, peptide-tethering, PEG-fusion, and the like.
  • promoter element describes a nucleotide sequence that is incorporated into a vector that, once inside an appropriate cell, can facilitate transcription factor and/or polymerase binding and subsequent transcription of portions of the vector DNA into mRNA.
  • the promoter element of the present invention precedes the 5′ end of the T1D specific marker nucleic acid molecule such that the latter is transcribed into mRNA. Host cell machinery then translates mRNA into a polypeptide.
  • nucleic acid vector can contain nucleic acid elements other than the promoter element and the T1D specific marker gene nucleic acid molecule.
  • nucleic acid elements include, but are not limited to, origins of replication, ribosomal binding sites, nucleic acid sequences encoding drug resistance enzymes or amino acid metabolic enzymes, and nucleic acid sequences encoding secretion signals, localization signals, or signals useful for polypeptide purification.
  • a “replicon” is any genetic element, for example, a plasmid, cosmid, bacmid, plastid, phage or virus that is capable of replication largely under its own control.
  • a replicon may be either RNA or DNA and may be single or double stranded.
  • an “expression operon” refers to a nucleic acid segment that may possess transcriptional and translational control sequences, such as promoters, enhancers, translational start signals (e.g., ATG or AUG codons), polyadenylation signals, terminators, and the like, and which facilitate the expression of a polypeptide coding sequence in a host cell or organism.
  • transcriptional and translational control sequences such as promoters, enhancers, translational start signals (e.g., ATG or AUG codons), polyadenylation signals, terminators, and the like, and which facilitate the expression of a polypeptide coding sequence in a host cell or organism.
  • reporter As used herein, the terms “reporter,” “reporter system”, “reporter gene,” or “reporter gene product” shall mean an operative genetic system in which a nucleic acid comprises a gene that encodes a product that when expressed produces a reporter signal that is a readily measurable, e.g., by biological assay, immunoassay, radio immunoassay, or by colorimetric, fluorogenic, chemiluminescent or other methods.
  • the nucleic acid may be either RNA or DNA, linear or circular, single or double stranded, antisense or sense polarity, and is operatively linked to the necessary control elements for the expression of the reporter gene product.
  • the required control elements will vary according to the nature of the reporter system and whether the reporter gene is in the form of DNA or RNA, but may include, but not be limited to, such elements as promoters, enhancers, translational control sequences, poly A addition signals, transcriptional termination signals and the like.
  • the introduced nucleic acid may or may not be integrated (covalently linked) into nucleic acid of the recipient cell or organism.
  • the introduced nucleic acid may be maintained as an episomal element or independent replicon such as a plasmid.
  • the introduced nucleic acid may become integrated into the nucleic acid of the recipient cell or organism and be stably maintained in that cell or organism and further passed on or inherited to progeny cells or organisms of the recipient cell or organism.
  • the introduced nucleic acid may exist in the recipient cell or host organism only transiently.
  • selectable marker gene@ refers to a gene that when expressed confers a selectable phenotype, such as antibiotic resistance, on a transformed cell.
  • operably linked means that the regulatory sequences necessary for expression of the coding sequence are placed in the DNA molecule in the appropriate positions relative to the coding sequence so as to effect expression of the coding sequence. This same definition is sometimes applied to the arrangement of transcription units and other transcription control elements (e.g. enhancers) in an expression vector.
  • Arecombinant organism or Atransgenic organism@ refer to organisms which have a new combination of genes or nucleic acid molecules. A new combination of genes or nucleic acid molecules can be introduced into an organism using a wide array of nucleic acid manipulation techniques available to those skilled in the art.
  • the term “organism” relates to any living being comprised of a least one cell. An organism can be as simple as one eukaryotic cell or as complex as a mammal. Therefore, the phrase Aa recombinant organism@ encompasses a recombinant cell, as well as eukaryotic and prokaryotic organism.
  • isolated protein or “isolated and purified protein” is sometimes used herein. This term refers primarily to a protein produced by expression of an isolated nucleic acid molecule of the invention. Alternatively, this term may refer to a protein that has been sufficiently separated from other proteins with which it would naturally be associated, so as to exist in “substantially pure” form. “Isolated” is not meant to exclude artificial or synthetic mixtures with other compounds or materials, or the presence of impurities that do not interfere with the fundamental activity, and that may be present, for example, due to incomplete purification, addition of stabilizers, or compounding into, for example, immunogenic preparations or pharmaceutically acceptable preparations.
  • a “specific binding pair” comprises a specific binding member (sbm) and a binding partner (bp) which have a particular specificity for each other and which in normal conditions bind to each other in preference to other molecules.
  • specific binding pairs are antigens and antibodies, ligands and receptors and complementary nucleotide sequences.
  • Aspecific binding pair@ is also applicable where either or both of the specific binding member and the binding partner comprise a part of a large molecule.
  • the specific binding pair comprises nucleic acid sequences, they will be of a length to hybridize to each other under conditions of the assay, preferably greater than 10 nucleotides long, more preferably greater than 15 or 20 nucleotides long.
  • Sample or “patient sample” or “biological sample” generally refers to a sample which may be tested for a particular molecule, preferably a T1D specific marker molecule, such as a marker shown in Tables 1-4. Samples may include but are not limited to cells, body fluids, including blood, serum, plasma, urine, saliva, tears, pleural fluid and the like.
  • agent and “test compound” are used interchangeably herein and denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials such as bacteria, plants, fungi, or animal (particularly mammalian) cells or tissues.
  • Biological macromolecules include siRNA, shRNA, antisense oligonucleotides, small molecules, antibodies, peptides, peptide/DNA complexes, and any nucleic acid based molecule, for example an oligo, which exhibits the capacity to modulate the activity of the SNP containing nucleic acids described herein or their encoded proteins. Agents are evaluated for potential biological activity by inclusion in screening assays described herein below.
  • modulate refers increasing or decreasing.
  • modulate refers to the ability of a compound or test agent to interfere with signaling or activity of a gene or protein of the present invention. Therefore, modulating the signaling mediated by the target genes disclosed herein (e.g., UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7) means that an agent or compound inhibits or enhances the activity of the proteins encoded by the gene. This includes altering the activity of natural killer cells, and preventing autoimmune beta cell destruction.
  • T1D SNP containing nucleic acids may be used for a variety of purposes in accordance with the present invention.
  • T1D-associated SNP containing DNA, RNA, or fragments thereof may be used as probes to detect the presence of and/or expression of T1D specific markers.
  • Methods in which T1D specific marker nucleic acids may be utilized as probes for such assays include, but are not limited to: (1) in situ hybridization; (2) Southern hybridization (3) northern hybridization; and (4) assorted amplification reactions such as polymerase chain reactions (PCR).
  • assays for detecting T1D-associated SNPs may be conducted on any type of biological sample, including but not limited to body fluids (including blood, urine, serum, gastric lavage), any type of cell (such as white blood cells, mononuclear cells) or body tissue.
  • T1D associated SNP containing nucleic acids, vectors expressing the same, T1D SNP containing marker proteins and anti-T1D specific marker antibodies of the invention can be used to detect T1D associated SNPs in body tissue, cells, or fluid, and alter T1D SNP containing marker protein expression for purposes of assessing the genetic and protein interactions involved in T1D.
  • the T1D-associated SNP containing nucleic acid in the sample will initially be amplified, e.g. using PCR, to increase the amount of the template as compared to other sequences present in the sample. This allows the target sequences to be detected with a high degree of sensitivity if they are present in the sample. This initial step may be avoided by using highly sensitive array techniques that are becoming increasingly important in the art.
  • new detection technologies can overcome this limitation and enable analysis of small samples containing as little as 1 ⁇ g of total RNA.
  • RLS Resonance Light Scattering
  • PWG planar wave guide technology
  • any of the aforementioned techniques may be used to detect or quantify T1D-associated SNP marker expression and accordingly, detect patient susceptibility for developing T1D.
  • kits which may contain an T1D-associated SNP specific marker polynucleotide or one or more such markers immobilized on a Gene Chip, an oligonucleotide, a polypeptide, a peptide, an antibody, a label, marker, or reporter, a pharmaceutically acceptable carrier, a physiologically acceptable carrier, instructions for use, a container, a vessel for administration, an assay substrate, or any combination thereof.
  • SNPs identified herein have been associated with the etiology of T1D, methods for identifying agents that modulate the activity of the genes and their encoded products containing such SNPs should result in the generation of efficacious therapeutic agents for the treatment of a variety of disorders associated with this condition.
  • Chromosomes 21, 6, 15, 9 and 1 contain regions which provide suitable targets for the rational design of therapeutic agents which modulate the activity of proteins encoded by these sequences. Small nucleic acid molecules or peptides corresponding to these regions may be used to advantage in the design of therapeutic agents which effectively modulate the activity of the encoded proteins.
  • candidate drugs can be screened from large libraries of synthetic or natural compounds.
  • One example is an FDA approved library of compounds that can be used by humans.
  • compound libraries are commercially available from a number of companies including but not limited to Maybridge Chemical Co.
  • the polypeptides or fragments employed in drug screening assays may either be free in solution, affixed to a solid support or within a cell.
  • One method of drug screening utilizes eukaryotic or prokaryotic host cells which are stably transformed with recombinant polynucleotides expressing the polypeptide or fragment, preferably in competitive binding assays.
  • primary cells may be isolated from donors expressing the minor or major SNP alleles associated with the T1D described herein.
  • Such cells either in viable or fixed form, can be used for standard binding assays.
  • One may determine, for example, formation of complexes between the polypeptide or fragment and the agent being tested, or examine the degree to which the formation of a complex between the polypeptide or fragment and a known substrate is interfered with by the agent being tested.
  • Another technique for drug screening provides high throughput screening for compounds having suitable binding affinity for the encoded polypeptides and is described in detail in Geysen, PCT published application WO 84/03564, published on Sep. 13, 1984. Briefly stated, large numbers of different, small peptide test compounds, such as those described above, are synthesized on a solid substrate, such as plastic pins or some other surface. The peptide test compounds are reacted with the target polypeptide and washed. Bound polypeptide is then detected by methods well known in the art.
  • a further technique for drug screening involves the use of host eukaryotic cell lines or cells (such as described above) which have a nonfunctional or altered T1D associated gene. These host cell lines or cells are defective at the polypeptide level. The host cell lines or cells are grown in the presence of drug compound. The rate of cellular metabolism of the host cells is measured to determine if the compound is capable of regulating cellular metabolism in the defective cells.
  • Host cells contemplated for use in the present invention include but are not limited to bacterial cells, fungal cells, insect cells, mammalian cells, and plant cells.
  • the T1D-associated SNP encoding DNA molecules may be introduced singly into such host cells or in combination to assess the phenotype of cells conferred by such expression.
  • donor cells expressing the alleles described herein may be employed.
  • Methods for introducing DNA molecules are also well known to those of ordinary skill in the art. Such methods are set forth in Ausubel et al. eds., Current Protocols in Molecular Biology, John Wiley & Sons, NY, N.Y. 1995, the disclosure of which is incorporated by reference herein.
  • Cells and cell lines suitable for studying the effects of the SNP encoding nucleic acids on glucose metabolism and methods of use thereof for drug discovery are provided. Such cells and cell lines will either already express the SNP or be transfected with the SNP encoding nucleic acids described herein and the effects on glucagon secretion, insulin secretion and/or beta cell apoptosis can be determined. Such cells and cell lines will also be contacted with the siRNA molecules provided herein to assess the effects thereof on glucagon secretion, insulin secretion and/or beta cell apoptosis.
  • siRNA molecules will be tested alone and in combination of 2, 3, 4, and 5 siRNAs to identify the most efficacious combination for down regulating at least one target gene, e.g., UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7.
  • Cells suitable for these purposes include, without limitation, INS cells (ATCC CRL 11605), PC12 cells (ATCC CRL 1721), MIN6 cells, alpha-TC6 cells and INS-1 832/13 cells (Fernandez et al., J. of Proteome Res. (2007). 7:400-411).
  • Pancreatic islet cells can be isolated and cultured as described in Joseph, J. et al., (J. Biol. Chem. (2004) 279:51049).
  • Diao et al. (J. Biol. Chem. (2005) 280:33487-33496), provide methodology for assessing the effects of the SNP encoding nucleic acids and/or the siRNAs provided herein on glucagon secretion and insulin secretion.
  • Park, J. et al. (J. of Bioch. and Mol. Biol. (2007) 40:1058-68) provide methodology for assessing the effect of these nucleic acid molecules on glucosamine induced beta cell apoptosis in pancreatic islet cells.
  • Suitable vectors for use in practicing the invention include prokaryotic vectors such as the pNH vectors (Stratagene Inc., 11099 N. Torrey Pines Rd., La Jolla, Calif. 92037), pET vectors (Novogen Inc., 565 Science Dr., Madison, Wis. 53711) and the pGEX vectors (Pharmacia LKB Biotechnology Inc., Piscataway, N.J. 08854).
  • Examples of eukaryotic vectors useful in practicing the present invention include the vectors pRc/CMV, pRc/RSV, and pREP (Invitrogen, 11588 Sorrento Valley Rd., San Diego, Calif.
  • pcDNA3.1/V5&His Invitrogen
  • baculovirus vectors such as pVL1392, pVL1393, or pAC360 (Invitrogen)
  • yeast vectors such as YRP17, YIPS, and YEP24 (New England Biolabs, Beverly, Mass.), as well as pRS403 and pRS413 Stratagene Inc.
  • Picchia vectors such as pHIL-D1 (Phillips Petroleum Co., Bartlesville, Okla. 74004)
  • retroviral vectors such as PLNCX and pLPCX (Clontech)
  • adenoviral and adeno-associated viral vectors adenoviral and adeno-associated viral vectors.
  • Promoters for use in expression vectors of this invention include promoters that are operable in prokaryotic or eukaryotic cells. Promoters that are operable in prokaryotic cells include lactose (lac) control elements, bacteriophage lambda (pL) control elements, arabinose control elements, tryptophan (trp) control elements, bacteriophage T7 control elements, and hybrids thereof.
  • lac lactose
  • pL bacteriophage lambda
  • trp tryptophan
  • Promoters that are operable in eukaryotic cells include Epstein Barr virus promoters, adenovirus promoters, SV40 promoters, Rous Sarcoma Virus promoters, cytomegalovirus (CMV) promoters, baculovirus promoters such as AcMNPV polyhedrin promoter, Picchia promoters such as the alcohol oxidase promoter, and Saccharomyces promoters such as the gal4 inducible promoter and the PGK constitutive promoter, as well as neuronal-specific platelet-derived growth factor promoter (PDGF), and the Thy-1 promoter.
  • Epstein Barr virus promoters include Epstein Barr virus promoters, adenovirus promoters, SV40 promoters, Rous Sarcoma Virus promoters, cytomegalovirus (CMV) promoters, baculovirus promoters such as AcMNPV polyhedrin promoter, Picchia promoters such as the alcohol oxidase promoter
  • a vector of this invention may contain any one of a number of various markers facilitating the selection of a transformed host cell.
  • markers include genes associated with temperature sensitivity, drug resistance, or enzymes associated with phenotypic characteristics of the host organisms.
  • Host cells expressing the T1D-associated SNPs of the present invention or functional fragments thereof provide a system in which to screen potential compounds or agents for the ability to modulate the development of T1D.
  • the nucleic acid molecules of the invention may be used to create recombinant cell lines for use in assays to identify agents which modulate aspects of the diabetic phenotype. Also provided herein are methods to screen for compounds capable of modulating the function of proteins encoded by the SNP containing nucleic acids described below.
  • Another approach entails the use of phage display libraries engineered to express fragment of the polypeptides encoded by the SNP containing nucleic acids on the phage surface. Such libraries are then contacted with a combinatorial chemical library under conditions wherein binding affinity between the expressed peptide and the components of the chemical library may be detected.
  • U.S. Pat. Nos. 6,057,098 and 5,965,456 provide methods and apparatus for performing such assays.
  • the goal of rational drug design is to produce structural analogs of biologically active polypeptides of interest or of small molecules with which they interact (e.g., agonists, antagonists, inhibitors) in order to fashion drugs which are, for example, more active or stable forms of the polypeptide, or which, e.g., enhance or interfere with the function of a polypeptide in vivo. See, e.g., Hodgson, (1991) Bio/Technology 9:19-21.
  • the three-dimensional structure of a protein of interest or, for example, of the protein-substrate complex is solved by x-ray crystallography, by nuclear magnetic resonance, by computer modeling or most typically, by a combination of approaches.
  • peptides may be analyzed by an alanine scan (Wells, (1991) Meth. Enzym. 202:390-411). In this technique, an amino acid residue is replaced by Ala, and its effect on the peptide's activity is determined. Each of the amino acid residues of the peptide is analyzed in this manner to determine the important regions of the peptide.
  • anti-idiotypic antibodies As a mirror image of a mirror image, the binding site of the anti-ids would be expected to be an analog of the original molecule.
  • the anti-id could then be used to identify and isolate peptides from banks of chemically or biologically produced banks of peptides. Selected peptides would then act as the pharmacophore.
  • drugs which have, e.g., improved polypeptide activity or stability or which act as inhibitors, agonists, antagonists, etc. of polypeptide activity.
  • SNP containing nucleic acid sequences described herein sufficient amounts of the encoded polypeptide may be made available to perform such analytical studies as x-ray crystallography.
  • the knowledge of the protein sequence provided herein will guide those employing computer modeling techniques in place of, or in addition to x-ray crystallography.
  • T1D-associated SNP containing nucleic acids enables the production of strains of laboratory mice carrying the T1D-associated SNPs of the invention.
  • Transgenic mice expressing the T1D-associated SNP of the invention provide a model system in which to examine the role of the protein encoded by the SNP containing nucleic acid in the development and progression towards T1D.
  • Methods of introducing transgenes in laboratory mice are known to those of skill in the art. Three common methods include: (1) integration of retroviral vectors encoding the foreign gene of interest into an early embryo; (2) injection of DNA into the pronucleus of a newly fertilized egg; and (3) the incorporation of genetically manipulated embryonic stem cells into an early embryo.
  • mice described above will facilitate the molecular elucidation of the role that a target protein plays in various cellular metabolic processes, including: aberrant lipid deposition, altered cellular metabolism and glucose regulation.
  • Such mice provide an in vivo screening tool to study putative therapeutic drugs in a whole animal model and are encompassed by the present invention.
  • animal is used herein to include all vertebrate animals, except humans. It also includes an individual animal in all stages of development, including embryonic and fetal stages.
  • a “transgenic animal” is any animal containing one or more cells bearing genetic information altered or received, directly or indirectly, by deliberate genetic manipulation at the subcellular level, such as by targeted recombination or microinjection or infection with recombinant virus.
  • transgenic animal is not meant to encompass classical cross-breeding or in vitro fertilization, but rather is meant to encompass animals in which one or more cells are altered by or receive a recombinant DNA molecule.
  • This molecule may be specifically targeted to a defined genetic locus, be randomly integrated within a chromosome, or it may be extra-chromosomally replicating DNA.
  • the term “germ cell line transgenic animal” refers to a transgenic animal in which the genetic alteration or genetic information was introduced into a germ line cell, thereby conferring the ability to transfer the genetic information to offspring. If such offspring, in fact, possess some or all of that alteration or genetic information, then they, too, are transgenic animals.
  • the alteration of genetic information may be foreign to the species of animal to which the recipient belongs, or foreign only to the particular individual recipient, or may be genetic information already possessed by the recipient. In the last case, the altered or introduced gene may be expressed differently than the native gene. Such altered or foreign genetic information would encompass the introduction of T1D-associated SNP containing nucleotide sequences.
  • the DNA used for altering a target gene may be obtained by a wide variety of techniques that include, but are not limited to, isolation from genomic sources, preparation of cDNAs from isolated mRNA templates, direct synthesis, or a combination thereof.
  • ES cells may be obtained from pre-implantation embryos cultured in vitro (Evans et al., (1981) Nature 292:154-156; Bradley et al., (1984) Nature 309:255-258; Gossler et al., (1986) Proc. Natl. Acad. Sci. 83:9065-9069).
  • Transgenes can be efficiently introduced into the ES cells by standard techniques such as DNA transfection or by retrovirus-mediated transduction.
  • the resultant transformed ES cells can thereafter be combined with blastocysts from a non-human animal.
  • the introduced ES cells thereafter colonize the embryo and contribute to the germ line of the resulting chimeric animal.
  • T1D-associated SNP genes as insertional cassettes to selectively inactivate a wild-type gene in totipotent ES cells (such as those described above) and then generate transgenic mice.
  • the use of gene-targeted ES cells in the generation of gene-targeted transgenic mice was described, and is reviewed elsewhere (Frohman et al., (1989) Cell 56:145-147; Bradley et al., (1992) Bio/Technology 10:534-539).
  • Non-homologous recombinants are selected against by using the Herpes Simplex virus thymidine kinase (HSV-TK) gene and selecting against its nonhomologous insertion with effective herpes drugs such as gancyclovir (GANC) or (1-(2-deoxy-2-fluoro-B-D arabinofluranosyl)-5-iodou-racil, (FIAU).
  • GANC Herpes Simplex virus thymidine kinase
  • FIAU (1-(2-deoxy-2-fluoro-B-D arabinofluranosyl)-5-iodou-racil
  • T1D-associated SNP containing nucleic acid as a targeted insertional cassette provides means to detect a successful insertion as visualized, for example, by acquisition of immunoreactivity to an antibody immunologically specific for the polypeptide encoded by T1D-associated SNP nucleic acid and, therefore, facilitates screening/selection of ES cells with the desired genotype.
  • a knock-in animal is one in which the endogenous murine gene, for example, has been replaced with human T1D-associated SNP containing gene of the invention. Such knock-in animals provide an ideal model system for studying the development of T1D.
  • T1D-associated SNP containing nucleic acid, fragment thereof, or a T1D-associated SNP fusion protein can be targeted in a “tissue specific manner” or “cell type specific manner” using a vector in which nucleic acid sequences encoding all or a portion of T1D-associated SNP are operably linked to regulatory sequences (e.g., promoters and/or enhancers) that direct expression of the encoded protein in a particular tissue or cell type.
  • regulatory sequences e.g., promoters and/or enhancers
  • Such regulatory elements may be used to advantage for both in vitro and in vivo applications. Promoters for directing tissue specific expression of proteins are well known in the art and described herein.
  • the nucleic acid sequence encoding the T1D-associated SNP of the invention may be operably linked to a variety of different promoter sequences for expression in transgenic animals.
  • promoters include, but are not limited to a prion gene promoter such as hamster and mouse Prion promoter (MoPrP), described in U.S. Pat. No. 5,877,399 and in Borchelt et al., Genet. Anal. 13(6) (1996) pages 159-163; a rat neuronal specific enolase promoter, described in U.S. Pat. Nos. 5,612,486, and 5,387,742; a platelet-derived growth factor B gene promoter, described in U.S. Pat. No.
  • a brain specific dystrophin promoter described in U.S. Pat. No. 5,849,999
  • a Thy-1 promoter a PGK promoter
  • a CMV promoter a neuronal-specific platelet-derived growth factor B gene promoter
  • Glial fibrillar acidic protein (GFAP) promoter for the expression of transgenes in glial cells.
  • Transgenic mice into which a nucleic acid containing the T1D-associated SNP or its encoded protein have been introduced are useful, for example, to develop screening methods to screen therapeutic agents to identify those capable of modulating the development of T1D.
  • compositions useful for treatment and diagnosis of T1D may comprise, in addition to one of the above substances, a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • administration is preferably in a “prophylactically effective amount” or a “therapeutically effective amount” (as the case may be, although prophylaxis may be considered therapy), this being sufficient to show benefit to the individual.
  • RNA interference involves a multi-step process. Double stranded RNAs are cleaved by the endonuclease Dicer to generate nucleotide fragments (siRNA). The siRNA duplex is resolved into 2 single stranded RNAs, one strand being incorporated into a protein-containing complex where it functions as guide RNA to direct cleavage of the target RNA (Schwarz et al, Mol. Cell. 10:537 548 (2002), Zamore et al, Cell 101:25 33 (2000)), thus silencing a specific genetic message (see also Zeng et al, Proc. Natl. Acad. Sci. 100:9779 (2003)).
  • the invention includes a method of treating T1D in a mammal.
  • An exemplary method entails administering to the mammal a pharmaceutically effective amount of an siRNA molecule directed to a gene target selected from the group consisting of UBASH3A (GenBank No.: NM — 018961; SEQ ID NO: 1), GLIS3, (GenBank No.: NM — 001042413; SEQ ID NO: 2), RASGRP1 (GenBank No.: NM — 005739; SEQ ID NO: 3), BACH2 (GenBank No.: NM — 021813; SEQ ID NO: 4) and EDG7 (GenBank Acc. No.: AY322547; SEQ ID NO: 5).
  • the siRNA inhibits the expression of the aforementioned genes.
  • the mammal is a human.
  • the term “patient” as used herein refers to a human.
  • siRNA preparations directed at inhibiting the expression of UBASH3A, GLIS3, RASGRP1, BACH2 and EDG7, as well as delivery methods are provided as a novel therapy to treat T1D. See Tables 6-10.
  • the siRNA can be delivered to a patient in vivo either systemically or locally with carriers, as discussed below.
  • the compositions of the invention may be used alone or in combination with other agents or genes encoding proteins to augment the efficacy of the compositions.
  • a “membrane permeant peptide sequence” refers to a peptide sequence which is able to facilitate penetration and entry of the siRNA inhibitor across the cell membrane.
  • Exemplary peptides include with out limitation, the signal sequence from Karposi fibroblast growth factor exemplified herein, the HIV tat peptide (Vives et al., J Biol. Chem., 272:16010-16017, 1997), Nontoxic membrane translocation peptide from protamine (Park et al., FASEB J. 19(11):1555-7, 2005), CHARIOT® delivery reagent (Active Motif; U.S. Pat. No. 6,841,535) and the antimicrobial peptide Buforin 2.
  • siRNAs are delivered for therapeutic benefit.
  • methods to administer the siRNA of the invention to in vivo to treat T1D including, but not limited to, naked siRNA delivery, siRNA conjugation and delivery, liposome carrier-mediated delivery, polymer carrier delivery, nanoparticle compositions, plasmid-based methods, and the use of viruses.
  • siRNA composition of the invention can comprise a delivery vehicle, including liposomes, for administration to a subject, carriers and diluents and their salts, and/or can be present in pharmaceutically acceptable formulations. This can be necessary to allow the siRNA to cross the cell membrane and escape degradation.
  • a delivery vehicle including liposomes
  • This can be necessary to allow the siRNA to cross the cell membrane and escape degradation.
  • Methods for the delivery of nucleic acid molecules are described in Akhtar et al., 1992, Trends Cell Bio., 2, 139; Delivery Strategies for Antisense Oligonucleotide Therapeutics, ed. Akhtar, 1995, Maurer et al., 1999, Mol. Membr. Biol., 16, 129-140; Hofland and Huang, 1999, Handb. Exp.
  • the frequency of administration of the siRNA to a patient will also vary depending on several factors including, but not limited to, the type and severity of the T1D to be treated, the route of administration, the age and overall health of the individual, the nature of the siRNA, and the like. It is contemplated that the frequency of administration of the siRNA to the patient may vary from about once every few months to about once a month, to about once a week, to about once per day, to about several times daily.
  • compositions that are useful in the methods of the invention may be administered systemically in parenteral, oral solid and liquid formulations, ophthalmic, suppository, aerosol, topical or other similar formulations.
  • these pharmaceutical compositions may contain pharmaceutically-acceptable carriers and other ingredients known to enhance and facilitate drug administration.
  • Such compositions may optionally contain other components, such as adjuvants, e.g., aqueous suspensions of aluminum and magnesium hydroxides, and/or other pharmaceutically acceptable carriers, such as saline.
  • nanoparticles such as nanoparticles, liposomes, resealed erythrocytes, and immunologically based systems may also be used to administer the appropriate siRNA to a patient according to the methods of the invention.
  • nanoparticles to deliver siRNAs, as well as cell membrane permeable peptide carriers that can be used are described in Crombez et al., Biochemical Society Transactions v35:p44 (2007).
  • Methods of the invention directed to treating T1D involve the administration of at least one UBASH3A, GLIS3, RASGRP1, BACH2 and EDG7 siRNA in a pharmaceutical composition.
  • the siRNA is administered to an individual as a pharmaceutical composition comprising the siRNA and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers include aqueous solutions such as physiologically buffered saline, other solvents or vehicles such as glycols, glycerol, oils such as olive oil or injectable organic esters.
  • a pharmaceutically acceptable carrier can contain physiologically acceptable compounds that act, for example, to stabilize the siRNA or increase the absorption of the agent.
  • physiologically acceptable compounds include, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients.
  • carbohydrates such as glucose, sucrose or dextrans
  • antioxidants such as ascorbic acid or glutathione
  • chelating agents such as ascorbic acid or glutathione
  • low molecular weight proteins or other stabilizers or excipients include, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients.
  • a pharmaceutical composition comprising siRNA can be administered to a subject by various routes including, for example, orally or parenterally, such as intravenously (i.v.), intramuscularly, subcutaneously, intraorbitally, intranasally, intracapsularly, intraperitoneally (i.p.), intracisternally, intratracheally (i.t.), or intra-articularly or by passive or facilitated absorption.
  • routes of administration can be used other pharmaceutically useful compounds, for example, small molecules, nucleic acid molecules, peptides, antibodies and polypeptides as discussed hereinabove.
  • a pharmaceutical composition comprising siRNA inhibitor also can be incorporated, if desired, into liposomes, microspheres, microbubbles, or other polymer matrices (Gregoriadis, Liposome Technology, Vols. Ito III, 2nd ed., CRC Press, Boca Raton Fla. (1993)).
  • Liposomes for example, which consist of phospholipids or other lipids, are nontoxic, physiologically acceptable and metabolizable carriers that are relatively simple to make and administer.
  • the pharmaceutical preparation comprises a siRNA targeting the SNP containing sequences described herein or an expression vector encoding for the siRNA.
  • Such pharmaceutical preparations can be administered to a patient for treating T1D.
  • Expression vectors for the expression of siRNA molecules preferably employ a strong promoter which may be constitutive or regulated.
  • promoters are well known in the art and include, but are not limited to, RNA polymerase II promoters, the T7 RNA polymerase promoter, and the RNA polymerase III promoters U6 and H1 (see, e.g., Myslinski et al. (2001) Nucl. Acids Res., 29:2502 09).
  • a formulated siRNA composition can be a composition comprising one or more siRNA molecules or a vector encoding one or more siRNA molecules independently or in combination with a cationic lipid, a neutral lipid, and/or a polyethyleneglycol-diacylglycerol (PEG-DAG) or PEG-cholesterol (PEG-Chol) conjugate.
  • PEG-DAG polyethyleneglycol-diacylglycerol
  • PEG-Chol PEG-cholesterol
  • a lipid nanoparticle composition is a composition comprising one or more biologically active molecules independently or in combination with a cationic lipid, a neutral lipid, and/or a polyethyleneglycol-diacylglycerol (i.e., polyethyleneglycol diacylglycerol (PEG-DAG), PEG-cholesterol, or PEG-DMB) conjugate.
  • a polyethyleneglycol-diacylglycerol i.e., polyethyleneglycol diacylglycerol (PEG-DAG), PEG-cholesterol, or PEG-DMB
  • the biologically active molecule is encapsulated in the lipid nanoparticle as a result of the process of providing and aqueous solution comprising a biologically active molecule of the invention (i.e., siRNA), providing an organic solution comprising lipid nanoparticle, mixing the two solutions, incubating the solutions, dilution, ultrafiltration, resulting in concentrations suitable to produce nanoparticle compositions.
  • a biologically active molecule of the invention i.e., siRNA
  • Nucleic acid molecules can be administered to cells by incorporation into other vehicles, such as biodegradable polymers, hydrogels, cyclodextrins. (see for example Gonzalez et al., 1999, Bioconjugate Chem., 10, 1068-1074; Wang et al., International PCT publication Nos. WO 03/47518 and WO 03/46185), poly(lactic-co-glycolic)acid (PLGA) and PLCA microspheres (see for example U.S. Pat. No. 6,447,796 and US Patent Application Publication No. US 2002130430), biodegradable nanocapsules, and bioadhesive microspheres, or by proteinaceous vectors (O′Hare and Normand, International PCT Publication No. WO 00/53722)
  • Cationic lipids and polymers are two classes of non-viral siRNA delivery which can form complexes with negatively charged siRNA.
  • the self-assembly PEG-ylated polycation polyethylenimine (PEI) has also been used to condense and protect siRNAs (Schiffelers et al., 2004, Nuc. Acids Res. 32: 141-110).
  • PEI polycation polyethylenimine
  • the siRNA complex can be condensed into a nanoparticle to allow efficient uptake of the siRNA through endocytosis.
  • the nucleic acid-condensing property of protamine has been combined with specific antibodies to deliver siRNAs and can be used in the invention (Song et al., 2005, Nat Biotech. 23:709-717).
  • siRNA should be administered in an effective dose.
  • the total treatment dose can be administered to a subject as a single dose or can be administered using a fractionated treatment protocol, in which multiple doses are administered over a more prolonged period of time, for example, over the period of a day to allow administration of a daily dosage or over a longer period of time to administer a dose over a desired period of time.
  • a fractionated treatment protocol in which multiple doses are administered over a more prolonged period of time, for example, over the period of a day to allow administration of a daily dosage or over a longer period of time to administer a dose over a desired period of time.
  • the amount of siRNA required to obtain an effective dose in a subject depends on many factors, including the age, weight and general health of the subject, as well as the route of administration and the number of treatments to be administered. In view of these factors, the skilled artisan would adjust the particular dose so as to obtain an effective dose for treating an individual having T1D.
  • the effective dose of siRNA will depend on the mode of administration, and the weight of the individual being treated.
  • the dosages described herein are generally those for an average adult but can be adjusted for the treatment of children.
  • the dose will generally range from about 0.001 mg to about 1000 mg.
  • the concentration of siRNA in a particular formulation will depend on the mode and frequency of administration.
  • a given daily dosage can be administered in a single dose or in multiple doses so long as the siRNA concentration in the formulation results in the desired daily dosage.
  • One skilled in the art can adjust the amount of siRNA in the formulation to allow administration of a single dose or in multiple doses that provide the desired concentration of siRNA over a given period of time.
  • siRNA can be particularly useful when administered in combination, for example, with a conventional agent for treating such a disease.
  • a conventional agent for treating such a disease The skilled artisan would administer siRNA, alone or in combination and would monitor the effectiveness of such treatment using routine methods such as pancreatic beta cell function determination, radiologic, immunologic or, where indicated, histopathologic methods.
  • Other conventional agents for the treatment of diabetes include insulin administration, glucagon administration or agents that alter levels of either of these two molecules. Glucophage®, Avandia®, Actos®, Januvia® and Glucovance® are examples of such agents.
  • Administration of the pharmaceutical preparation is preferably in an “effective amount” this being sufficient to show benefit to the individual. This amount prevents, alleviates, abates, or otherwise reduces the severity of T1D symptoms in a patient.
  • Dosage unit form refers to a physically discrete unit of the pharmaceutical preparation appropriate for the patient undergoing treatment. Each dosage should contain a quantity of active ingredient calculated to produce the desired effect in association with the selected pharmaceutical carrier. Procedures for determining the appropriate dosage unit are well known to those skilled in the art.
  • Dosage units may be proportionately increased or decreased based on the weight of the patient. Appropriate concentrations for alleviation of a particular pathological condition may be determined by dosage concentration curve calculations, as known in the art.
  • Type 1 Diabetes Cohort from Canada 1. Type 1 Diabetes Cohort from Canada:
  • the Canadian cohort consisted of 1,120 nuclear family trios (one affected child and two parents) and 267 independent T1D cases, collected in pediatric diabetes clinics in Montreal, Toronto, Ottawa and Winnipeg.
  • the median age at onset is 8 with lower and upper quartiles at 4.6 years and 11 years. All patients were diagnosed under the age of 18, were treated with insulin since diagnosis and none has stopped treatment for any reason since. Disease diagnosis was based on these clinical criteria, rather than any laboratory tests.
  • Ethnic backgrounds were of mixed European descent, with the largest single subset (409 families) being French Canadian.
  • the Research Ethics Board of the Montreal Children's Hospital and other participating centers approved the study, and written informed consent was obtained from all subjects.
  • the Type 1 Diabetes Genetics Consortium cohort consisted of 549 families (2350 individuals) with at least two children diagnosed with diabetes and both parents available as of the July 2005 data freeze. Criteria were age at diagnosis below 35 years and uninterrupted treatment with insulin within six months of diagnosis. For siblings of probands diagnosed under the age of 35, the age-at-diagnosis limit was extended to 45 if they were lean and had positive antibodies and/or low C-peptide levels at diagnosis. The median age is 8 with quartiles at 4 years and 13 years. The samples were collected in Europe, North America and Australia and most subjects were of European ancestry. Autoantibody results are available but were not used to substantiate the diagnosis, except as noted above.
  • the T1D cohort consisted of 103 children recruited at the Children's Hospital of Philadelphia (CHOP), since September, 2006. All patients were diagnosed under the age of 18. Of those, 49 T1D patients (32 female, 17 male) were Caucasians by self-report (average age of onset 7.07 years; range 9 months-14 years) and were included in the analysis. All were treated with insulin since diagnosis and none has stopped treatment for any reason since.
  • the Research Ethics Board of CHOP approved the study and written informed consent was obtained from all subjects.
  • Type 1 diabetes cohort The DCCT was a multi-center randomized clinical trial to determine the effect of intensive insulin treatment with respect to reduced development and progression of retinopathy and nephropathy complications in patients with type 1 diabetes 19, 20 . A total of 1,441 subjects with type 1 diabetes were recruited from 29 centers across North America into the DCCT between 1983 and 1989; they were between 13 and 39 years of age and 53% were male.
  • the primary prevention cohort consisted of 726 subjects with no retinopathy, an albumin excretion rate ⁇ 28 ⁇ g per minute, and diabetes duration of 1 to 5 years and were used to determine if intensive therapy prevented the development of diabetic retinopathy in patients with no retinopathy
  • the secondary intervention cohort consisted of 715 subjects who had non-proliferative retinopathy, a urinary albumin excretion rate ⁇ 140 ⁇ g per minute, and diabetes duration of 1 to 15 years were studied to determine whether intensive therapy would affect the progression of early retinopathy 19 .
  • Approval for the DCCT/EDIC Genetics study was provided by the Research Ethics Board of the Hospital for Sick Children, Toronto.
  • the control group included 1,146 children with self reported Caucasian status, mean age 9.42 years; 53.05% male and 46.95% female, who did not have diabetes or a first-degree relative with T1D.
  • the control group used to match with the 1,100 DCCT/EDIC T1D probands included 2,024 children with self reported Caucasian ethnicity, mean age 8.82 years; 50.83% male and 49.17% female, who did not have diabetes or a first-degree relative with T1D. These individual were recruited by CHOP's clinicians and nursing staff within the CHOP's Health Care Network, including four primary care clinics and several group practices and outpatient practices that included well child visits.
  • Genotypes for this study were obtained using the Infinium and GoldenGate platforms from Illumina.
  • DCCT/EDIC samples were genotyped on the Illumina 1M chip at Illumina (San Diego, Calif.).
  • Type 1 diabetes is a multifactorial disease with a strong genetic component that results from autoimmune destruction of the pancreatic ⁇ -cells.
  • the major T1D susceptibility locus mapping to the HLA class II genes at 6p21 1 and encoding highly polymorphic antigen-presenting proteins, accounts for almost 50% of the genetic risk for T1D 2 .
  • Several other loci with more modest effects account for another 10-20% of the risk.
  • INS insulin
  • VNTR 3 insulin VNTR 3
  • SNP Arg620Trp single-nucleotide polymorphism
  • IL2RA 7-9 Non-coding SNPs at IL2RA 7-9 , which encodes the ⁇ chain of the IL2 receptor complex (CD25), an important modulator of immunity
  • CTLA4 locus 10 whose protein product transmits inhibitory signals to attenuate T-cell activation. It is worth noting that all of these T1D-associated genes are expressed in cells with immune function and all except INS have been associated with other autoimmune disorders.
  • T1D single nucleotide polymorphism
  • T1D risk loci we carried out a follow-up strategy to uncover additional novel T1D risk loci.
  • loci that were significantly associated with T1D during this process, both of which reside in genes that are biologically relevant to autoimmunity.
  • These genes encode ubiquitin-associated and SH3 domain-containing protein A (UBASH3A) and BTB and CNC homology 2 (BACH2), respectively, both of which are known to be involved in T cell signaling.
  • UASH3A ubiquitin-associated and SH3 domain-containing protein A
  • BACH2 CNC homology 2
  • UBASH3A is the only gene in this region of linkage disequilibrium. Mice lacking Sts2 (the mouse homologue for UBASH3A) have been shown to be normal in all respects, including T-cell function 23 . Mice lacking both Sts1 and Sts2 do have increased splenocyte numbers and are hyperresponsive to T-cell receptor stimulation. It has been suggested that STS1 and STS2 are critical regulators of the signaling pathways that regulate T-cell activation 23 .
  • BACH2 is also the only gene at this locus.
  • the gene product is a member of the small Maf family which are basic region leucine zipper proteins that function either as transcriptional activators or repressors, depending on the proteins they heterodimerize with.
  • Muto et al 24 found that Bach2 ⁇ / ⁇ mice had relatively high levels of serum IgM but low levels of IgA and IgG subclasses.
  • the Bach2 ⁇ / ⁇ mice have also been reported to present with deficient T cell-independent and T cell-dependent IgG responses, leading the authors to conclude that BACH2 was a regulator of the antibody response 24 .
  • IPA software we generated a BACH-2 related protein-protein interaction network of differentially expressed genes (DEG) for T1D.
  • Grey represents genes that are up regulated in TID group versus controls.
  • White represents genes that are down regulated in T1D.
  • Solid lines show direct protein-protein interactions whereas dotted lines show indirect interactions. Different shapes represent different molecule types. See
  • rs1983853 yielded nominally significant association with T1D in all the cohorts but did not survive correction for multiple testing in the final replication attempt in the Toronto dataset.
  • This SNP resides in endothelial differentiation gene 7 (EDG7; formerly LPA3), which has been implicated in mechanisms of embryo implantation 25 .
  • EDG7 endothelial differentiation gene 7
  • IPA software we generated an EDG7-LPAR3-related protein-protein interaction network of differentially expressed genes (DEG) for T1D.
  • Grey represents genes that are up regulated in TID group versus controls.
  • White represents genes that are down regulated in T1D.
  • Solid lines show direct protein-protein interactions whereas dotted lines show indirect interactions. Different shapes represent different molecule types. See FIG. 3 .
  • the Diabetes Control and Complications Trial Research Group The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus.
  • the Diabetes Control and Complications Trial Research Group N Engl J Med 329, 977-86 (1993).
  • T1DGC Type 1 Diabetes Genetics Consortium
  • Example 1 identifying a novel type 1 diabetes locus involving the RASGRP1 gene. This gene is known to play a crucial role in thymocyte differentiation and TCR signaling by activating the Ras signaling pathway.
  • Example II The following materials and methods are similar to those described in Example I and are provided to facilitate the practice of Example 2.
  • SNP single-nucleotide polymorphisms
  • Type 1 diabetes association was tested by the Family Based Association Test (FBAT) software available on the world wide web at biostat.harvard.edu/ ⁇ fbat/fbat.htm 8 .
  • FBAT Family Based Association Test
  • the option of the empirical variance was used in the FBAT statistics to permit a robust but unbiased test of genetic association.
  • SNPs were tested in the RR2 study, we used a conserved significance threshold corrected for multiple comparisons at 3.26 ⁇ 10 ⁇ 5 .
  • the genotype calling rate of rs17574546 in the RR2 samples is 99.8%, and for rs7171171 in our own samples is 99.9%. No Mendelian error was found in either.
  • This novel type 1 diabetes association signal maps to a LD block at Chr15q14, ⁇ 13 kb upstream of the transcription start site of the RASGRP1 gene, and has no LD with any known type 1 diabetes locus. See FIG. 1 .
  • type 1 diabetes is caused by the autoimmune destruction of pancreatic ⁇ -cells, it is interesting that the RASGRP1 gene has an important immune function.
  • RASGRP1 (NCBI GeneID: 10125) encodes calcium and DAG-regulated RAS guanyl releasing protein 1 (RasGRP1) 9 .
  • RasGRP1 plays crucial roles in thymocyte differentiation and TCR signaling by activating the Ras signaling pathway.
  • RasGRP1-null mutant mice have approximately normal numbers of immature thymocytes but a marked deficiency of mature, single-positive (CD4 + CD8 ⁇ and CD4 ⁇ CD8 + ) thymocytes 10 .
  • Transgenic expression of RasGRP1 induces the maturation of double-negative thymocytes and enhances the production of CD4 ⁇ CD8 + thymocytes 11 .
  • RasGRP1 has dramatic effect on the development and function of CD4 + CD25 + regulatory T-cells (T reg ).
  • CD4 + CD25 + T reg plays a critical role in maintaining immune homeostasis and inhibiting autoimmune reaction of type 1 diabetes and other autoimmune diseases 13 .
  • transfer of CD4 + CD25 + CD4 + CD25 + T reg cells can prevent type 1 diabetes in the recipient NOD mice 14 , knowledge of the role of genes involved in the generation of this subset in type 1 diabetes may play an important role in the development of preventive interventions.
  • a preferred embodiment of the invention comprises clinical application of the information described herein to a patient.
  • Diagnostic compositions, including microarrays, and methods can be designed to identify the genetic alterations described herein in nucleic acids from a patient to assess susceptibility for developing T1D. This can occur after a patient arrives in the clinic; the patient has blood drawn, and using the diagnostic methods described herein, a clinician can detect a SNP in the regions of chromosome 21, 15, 6, 9 and 1 described herein.
  • the typical age range for a patient to be screened is between 9 and 12 years of age.
  • kits for performing the diagnostic method of the invention comprise a microarray comprising at least one of the SNPs provided herein in and the necessary reagents for assessing the patient samples as described above.
  • T1D-involved genes and the patient results will indicate which variants are present, and will identify those that possess an altered risk for developing T1D.
  • the information provided herein allows for therapeutic intervention at earlier times in disease progression that previously possible.
  • UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7 provide novel targets for the development of new therapeutic agents efficacious for the treatment of T1D.
  • the therapeutic siRNAs described herein can be used to block expression of the gene product based on the patient signal, thereby inhibiting the pancreatic 3-cell destruction that occurs in T1D.
  • Candidate siRNA compositions for use in the invention are provided in Tables 6-10.
  • the sequences in Tables 6-10 include several siRNAs (i.e., sense sequences for a target region).
  • siRNAs i.e., sense sequences for a target region.
  • Those of skill in the art can determine the sequence of an antisense siRNA strand based on the disclosure of the sense strand, and will appreciate the difference between any “U” and “T” designations in the sequences which correspond to RNA and DNA molecules, respectively.
  • methods of using known inhibitors of UBASH3A, GLIS3, RASGRP1, BACH2 and EDG7 to treat T1D are also provided. See the tables below.
  • shRNA constructs can be designed based on the sense sequence provided in Tables 4-8, and may be effective to inhibit UBASH3A, GLIS3, RASGRP1, BACH2 and EDG7.
  • the shRNA constructs utilizing the sense strand from Tables 6-10 for the respective targets would include a hairpin loop 3′ to the sense sequence (e.g., suitable hairpins include, but are not limited to: TCAAGAG, TTCAAGAGA, GAAGCTTG, and TTCG) followed by the corresponding antisense sequence from the sense strand provided in Tables 6-10.
  • Table 10B provides known small molecule antagonists of EDG7 which should have utility for the treatment of T1D.
  • PC AM966 is a LPA1 (lysophosphatidic acid receptor 1) Pulmonary Lysophosphatidic Acid Pharmaceuticals inhibitor.
  • Lysophosphatidic acid receptor 1 LPAR1; Fibrosis Type 1 Receptor (LPA1) (Private) EDG2; LPA1 is an EDG receptor that is part of the (Idiopathic Antagonist phosphatidic acid/lysophosphatidylcholine Pulmonary pathway.
  • Lysophosphatidic acid is an anionic Fibrosis (LPA) bioactive lipid that acts through EDG receptors to mediate cell proliferation, platelet aggregation, smooth muscle contraction, inhibition of neuroblastoma cell differentiation, chemotaxis and tumor cell invasion. LPA also elicits angiogenesis. AM966 is being developed for the treatment of idiopathic pulmonary fibrosis (IPF).
  • Amira LPA1 PC LPA1 Receptor Antagonist acts by blocking the Cancer Lysophosphatidic Acid Pharmaceuticals Receptor LPA-induced migration of cancer cells and is being Type 1 Receptor (LPA1) (Private) Antagonist developed for the treatment of cancer.
  • Antagonist Amira LPA1 — PC LPA1 Receptor Antagonist is an anti-fibrotic and Kidney Disease Lysophosphatidic Acid Pharmaceuticals Receptor anti-inflammatory agent which is being developed (Kidney Fibrosis) Type 1 Receptor (LPA1) (Private) Antagonist for the treatment of kidney fibrosis.
  • Antagonist Catena VPC51299 — PC VPC51299 is a potent lysophosphatidic acid (LPA1) Solid Tumors Lysophosphatidic Acid Pharmaceuticals, receptor antagonist which targets LPA G-protein Type 1 Receptor (LPA1) Inc. (Private) coupled receptors (GPCRs) with high potency.
  • VPC51299 antagonizes (blocks) this signaling cascade.
  • VPC51299 is being developed as oral formulation for the treatment of solid tumors.
  • Debiopharm Debio0719 PC Debio 0719 is a highly potent, selective, small Bone Metastases Lysophosphatidic Acid Group (Private) molecule inhibitor of type 1 lysophosphatidic acid (Bone Damage Type 1 Receptor (LPA1) receptor (LPA1) and type 3 (LPA3).
  • Debio 0719 is Induced by Antagonist; being developed for the treatment of osteolytic Metastatic Lysophosphatidic bone damage induced by breast tumour Breast Cancer) Acid Type 3 Receptor metastasis.
  • PC Debio0719 is a highly potent, selective, small Bone Metastases Lysophosphatidic Acid Kirin Co., Ltd. molecule inhibitor of type 1 lysophosphatidic acid (Bone Damage Type 1 Receptor (LPA1) (formerly Kyowa receptor (LPA1) and type 3 (LPA3).
  • Debio0719 is Induced by Antagonist; Hakko Kogyo being developed for the treatment of bone damage Metastatic Lysophosphatidic Co., Ltd.) (Stock induced by metastatic breast cancer. Note: This Breast Cancer) Acid Type 3 Receptor Code Number: product is added upon merger of Kyowa Hakko (LPA3) Antagonist 4151 (TYO)) Kogyo Co., Ltd.
  • AM152 1 AM152 is a lysophosphatidic acid (LPA1) Pulmonary Lysophosphatidic Acid Pharmaceuticals antagonist that acts by blocking phosphatidic Fibrosis Type 1 Receptor (LPA1) (Private) acid/lysophosphatidylcholine pathway involved in (Idiopathic Antagonist fibrotic disease and is being developed for the Pulmonary treatment of idiopathic pulmonary fibrosis. Fibrosis)

Abstract

Compositions and methods for the detection and treatment of T1D are provided.

Description

  • This application is a §365 application of PCT/US2009/044356 filed May 19, 2009 which claims priority to U.S. Provisional Application 61/054,040 filed May 16, 2008, each of the aforementioned applications being incorporated herein by reference as though set forth in full.
  • FIELD OF THE INVENTION
  • This invention relates to the fields of glucose metabolism, genetics and pathology associated with diabetes, particularly type I diabetes. More specifically, the invention provides a panel of genes containing genetic alterations, e.g., single nucleotide polymorphisms, which had heretofore not been associated with this disease. Methods and kits for using the sequences so identified for diagnostic and therapeutic treatment purposes are also provided, as are therapeutic compositions for management of diabetes.
  • BACKGROUND OF THE INVENTION
  • Several publications and patent documents are cited throughout the specification in order to describe the state of the art to which this invention pertains. Each of these citations is incorporated herein by reference as though set forth in full.
  • Type I diabetes (T1D) results from the autoimmune destruction of pancreatic beta cells, a process believed to be strongly influenced by multiple genes and environmental factors. The incidence of T1D has been increasing in Western countries and has more than doubled in the United States over the past 30 years. The disease shows a strong familial component, with first-degree relatives of cases being at 15 times greater risk of T1D than a randomly selected member of the general population and monozygotic twins being concordant for T1D at a frequency of approximately 50%. However, while the genetic evidence is strong, the latter data suggests that an interplay with environmental factors also plays a key role in influencing T1D outcome.
  • The familial clustering of T1D is influenced by multiple genes. Variation in four loci has already been established to account for a significant proportion of the familial aggregation of T1D. These include the major histocompatibility complex (MHC) region on 6p21 (including the HLA-DRBJ, -DQA1 and -DRQJ genes1); the insulin/insulin-like growth factor 2 gene complex (INS-IGF2) on 11p1524, the protein tyrosine phosphatase-22 (PTPN22) gene on 1p135, 6 and the gene encoding cytotoxic T-lymphocyte-associated protein 4 (CTLA4) on 2q317, 8. The interleukin-2 receptor alpha (CD25) locus on 10p159 has also been implicated in the pathogenesis of T1D but remains to be replicated by independent studies. In addition, spontaneous mouse model studies of T1D have implicated numerous other regions that have been confirmed in replication studies10. Several other loci have also been implicated in human association studies with T1D but the effects of these implicated genes remain controversial and are subject to confirmation in independent studies utilizing sufficient sample sizes. Together, these studies suggest that many more T1D susceptibility genes remain to be discovered.
  • SUMMARY OF THE INVENTION
  • In accordance with the present invention, T1D-associated SNPs have been identified which are indicative of an increased or reduced risk of developing T1D. Thus, in one aspect, nucleic acids comprising at least one genetic alteration identified in Tables 1, 2, 4 and 5 are provided. Such nucleic acids and the proteins encoded thereby have utility in the diagnosis and management of type 1 diabetes (T1D).
  • In another aspect of the invention, methods for assessing susceptibility for developing T1D are provided. An exemplary method entails providing a target nucleic acid from a patient sample, said target nucleic acid having a predetermined sequence in the normal population, and assessing said target nucleic acid for the presence of at least one genetic alteration, e.g., a single nucleotide polymorphism, which is indicative of an increased or decreased risk of developing T1D. Such genetic alterations include, without limitation, inversion, deletion, duplication, and insertion of at least one nucleotide in said sequence.
  • Preferably, the genetic alteration is a single nucleotide polymorphism present in UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7 encoding nucleic acids and genetic regions associated therewith. Such genetic regions include the linkage disequilibrium blocks provided in Table 3 and the method entails detecting any variant associated with diabetes in such blocks. Preferably, the SNP is rs9976767 present on chromosome 21 at position 42709459 within the UBASH3A gene, the SNP is rs3757247 present on chromosome 6 at position 91014184 in the BACH2 gene or the SNP is rs7171171 at position 36694333 on chromosome 15 in the RASGRP1 gene.
  • The methods of the invention also include the detection of any of the T1D associated genetic alterations comprising the single nucleotide polymorphisms set forth in Tables 1, 2, 4 or 5 for the diagnosis of T1D. Alternatively or in addition, genetic alterations associated with T1D present in the linkage disequilibrium blocks set forth in Table 3 can be detected. Kits and microarrays for practicing the foregoing methods are also provided.
  • In yet another embodiment, a method of managing T1D is provided which entails administering a therapeutic agent to a patient in need thereof. The therapeutic agent can be a small molecule, an antibody, a protein, an oligonucleotide, or a siRNA molecule.
  • In another aspect of the invention, a method for identifying agents that bind and/or modulate UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7 functional activity is provided, as well as pharmaceutical compositions comprising said agent in a biologically acceptable carrier.
  • BRIEF DESCRIPTION OF THE DRAWING
  • FIG. 1. The LD plot of the RASGRP1 SNPs, based on the HapMap European data. The top panel shows the constrained elements and conservation scores called by the DNA sequence alignments of 29 eutherian mammals (Ensembl, on the world wide web at .ensembl.org. See Cooper et al. Genome Research 2005; 15:901-913. The LD map is made by Haploview v4.0 software available on the world wide web at road.mit.edu/personal/jcbarret/haploview. D′ values (%) are shown in the boxes, and r2 values are represented by the grey scale. The red arrows highlight the SNPs genotyped in this study. Inside the red circle, it is the SNP described in Example I.
  • FIG. 2. BACH-2 related protein protein interaction network of differentially expressed genes for T1D.
  • FIG. 3. EDG7/LPAR3 related protein protein interaction network of differentially expressed genes for T1D.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Type 1 diabetes (T1D) is a common and strongly heritable disease that most often manifests in childhood. Recent genome wide association studies have revealed a number of new genes associated with the disease. We carried out a follow-up strategy to our T1D GWA study in an attempt to uncover additional novel T1D risk loci. We selected 982 single nucleotide polymorphisms (SNPs) with at least a nominally significant P-value (but excluding SNPs in the major histocompatibility complex region) from a combination of our data generated on 563 T1D probands and 1,146 controls plus 483 complete T1D family trios of the same ancestry, using the Illumina HumanHap550 BeadChip. We then genotyped these SNPs in an independent cohort of 939 nuclear T1D families from Montreal and the type 1 diabetes genetics consortium. Subsequently, we looked across all three cohorts plus the Wellcome Trust Case Control Consortium dataset for T1D to identify SNPs in loci that were both not previously described and nominally significant across all cohorts. We selected five loci for further investigation, which we queried in T1D probands from the DCCT/EDIC study including 1,303 T1D patients using an independent matched control dataset of diabetes free individuals from Philadelphia which were genotyped on the 1M and HumanHap550K SNP BeadChips, respectively. Two of the five variants (rs9976767 and rs3757247) were also significantly associated with T1D in this cohort; these SNPs reside in the UBASH3A (OR: 1.16; five cohorts combined P=2.33×10−8) and BACH2 (OR: 1.13; combined P=1.25×10−6) genes respectively, both of which are biologically relevant to autoimmunity. In summary, we have identified two novel loci on 21q and 6q that are associated with T1D across five different cohorts of European decent.
  • The following definitions are provided to facilitate an understanding of the present invention:
  • For purposes of the present invention, “a” or “an” entity refers to one or more of that entity; for example, “a cDNA” refers to one or more cDNA or at least one cDNA. As such, the terms “a” or “an,” “one or more” and “at least one” can be used interchangeably herein. It is also noted that the terms “comprising,” “including,” and “having” can be used interchangeably. Furthermore, a compound “selected from the group consisting of refers to one or more of the compounds in the list that follows, including mixtures (i.e. combinations) of two or more of the compounds. According to the present invention, an isolated, or biologically pure molecule is a compound that has been removed from its natural milieu. As such, Aisolated@ and Abiologically pure@ do not necessarily reflect the extent to which the compound has been purified. An isolated compound of the present invention can be obtained from its natural source, can be produced using laboratory synthetic techniques or can be produced by any such chemical synthetic route.
  • A “single nucleotide polymorphism (SNP)” refers to a change in which a single base in the DNA differs from the usual base at that position. These single base changes are called SNPs or “snips.” Millions of SNP's have been cataloged in the human genome. Some SNPs such as that which causes sickle cell are responsible for disease. Other SNPs are normal variations in the genome.
  • The term “genetic alteration” as used herein refers to a change from the wild-type or reference sequence of one or more nucleic acid molecules. Genetic alterations include without limitation, base pair substitutions, additions and deletions of at least one nucleotide from a nucleic acid molecule of known sequence.
  • The phrase “Type 1 diabetes (T1D)” refers to a chronic (lifelong) disease that occurs when the pancreas produces too little insulin to regulate blood sugar levels appropriately. T1D, often called juvenile or insulin-dependent diabetes results from altered metabolism of carbohydrates (including sugars such as glucose), proteins, and fats. In type 1 diabetes, the beta cells of the pancreas produce little or no insulin, the hormone that allows glucose to enter body cells. Once glucose enters a cell, it is used as fuel. Without adequate insulin, glucose builds up in the bloodstream instead of going into the cells. The body is unable to use this glucose for energy despite high levels in the bloodstream, leading to increased hunger. In addition, the high levels of glucose in the blood cause the patient to urinate more, which in turn causes excessive thirst. Within 5 to 10 years after diagnosis, the insulin-producing beta cells of the pancreas are completely destroyed, and no more insulin is produced.
  • “T1D-associated SNP or specific marker” is a SNP or marker which is associated with an increased or decreased risk of developing TID not found normal patients who do not have this disease. Such markers may include but are not limited to nucleic acids, proteins encoded thereby, or other small molecules. Type 1 diabetes can occur at any age, but it usually starts in people younger than 30. Symptoms are usually severe and occur rapidly. The exact cause of type 1 diabetes is not known. Type 1 diabetes accounts for 3% of all new cases of diabetes each year. There is 1 new case per every 7,000 children per year. New cases are less common among adults older than 20.
  • The term “solid matrix” as used herein refers to any format, such as beads, microparticles, a microarray, the surface of a microtitration well or a test tube, a dipstick or a filter. The material of the matrix may be polystyrene, cellulose, latex, nitrocellulose, nylon, polyacrylamide, dextran or agarose. “Sample” or “patient sample” or “biological sample” generally refers to a sample which may be tested for a particular molecule, preferably an T1D specific marker molecule, such as a marker shown in the tables provided below. Samples may include but are not limited to cells, body fluids, including blood, serum, plasma, urine, saliva, tears, pleural fluid and the like.
  • The phrase “consisting essentially of when referring to a particular nucleotide or amino acid means a sequence having the properties of a given SEQ ID NO. For example, when used in reference to an amino acid sequence, the phrase includes the sequence per se and molecular modifications that would not affect the functional and novel characteristics of the sequence.
  • “Linkage” describes the tendency of genes, alleles, loci or genetic markers to be inherited together as a result of their location on the same chromosome, and is measured by percent recombination (also called recombination fraction, or θ) between the two genes, alleles, loci or genetic markers. The closer two loci physically are on the chromosome, the lower the recombination fraction will be. Normally, when a polymorphic site from within a disease-causing gene is tested for linkage with the disease, the recombination fraction will be zero, indicating that the disease and the disease-causing gene are always co-inherited. In rare cases, when a gene spans a very large segment of the genome, it may be possible to observe recombination between polymorphic sites on one end of the gene and causative mutations on the other. However, if the causative mutation is the polymorphism being tested for linkage with the disease, no recombination will be observed.
  • “Centimorgan” is a unit of genetic distance signifying linkage between two genetic markers, alleles, genes or loci, corresponding to a probability of recombination between the two markers or loci of 1% for any meiotic event.
  • “Linkage disequilibrium” or “allelic association” means the preferential association of a particular allele, locus, gene or genetic marker with a specific allele, locus, gene or genetic marker at a nearby chromosomal location more frequently than expected by chance for any particular allele frequency in the population.
  • “Target nucleic acid” as used herein refers to a previously defined region of a nucleic acid present in a complex nucleic acid mixture wherein the defined wild-type region contains at least one known nucleotide variation which may or may not be associated with T1D. The nucleic acid molecule may be isolated from a natural source by cDNA cloning or subtractive hybridization or synthesized manually. The nucleic acid molecule may be synthesized manually by the triester synthetic method or by using an automated DNA synthesizer.
  • With regard to nucleic acids used in the invention, the term “isolated nucleic acid” is sometimes employed. This term, when applied to DNA, refers to a DNA molecule that is separated from sequences with which it is immediately contiguous (in the 5′ and 3′ directions) in the naturally occurring genome of the organism from which it was derived. For example, the “isolated nucleic acid” may comprise a DNA molecule inserted into a vector, such as a plasmid or virus vector, or integrated into the genomic DNA of a prokaryote or eukaryote. An “isolated nucleic acid molecule” may also comprise a cDNA molecule. An isolated nucleic acid molecule inserted into a vector is also sometimes referred to herein as a recombinant nucleic acid molecule.
  • With respect to RNA molecules, the term “isolated nucleic acid” primarily refers to an RNA molecule encoded by an isolated DNA molecule as defined above. Alternatively, the term may refer to an RNA molecule that has been sufficiently separated from RNA molecules with which it would be associated in its natural state (i.e., in cells or tissues), such that it exists in a “substantially pure” form. By the use of the term “enriched” in reference to nucleic acid it is meant that the specific DNA or RNA sequence constitutes a significantly higher fraction (2-5 fold) of the total DNA or RNA present in the cells or solution of interest than in normal cells or in the cells from which the sequence was taken. This could be caused by a person by preferential reduction in the amount of other DNA or RNA present, or by a preferential increase in the amount of the specific DNA or RNA sequence, or by a combination of the two. However, it should be noted that “enriched” does not imply that there are no other DNA or RNA sequences present, just that the relative amount of the sequence of interest has been significantly increased.
  • It is also advantageous for some purposes that a nucleotide sequence be in purified form. The term “purified” in reference to nucleic acid does not require absolute purity (such as a homogeneous preparation); instead, it represents an indication that the sequence is relatively purer than in the natural environment (compared to the natural level, this level should be at least 2-5 fold greater, e.g., in terms of mg/ml). Individual clones isolated from a cDNA library may be purified to electrophoretic homogeneity. The claimed DNA molecules obtained from these clones can be obtained directly from total DNA or from total RNA. The cDNA clones are not naturally occurring, but rather are preferably obtained via manipulation of a partially purified naturally occurring substance (messenger RNA). The construction of a cDNA library from mRNA involves the creation of a synthetic substance (cDNA) and pure individual cDNA clones can be isolated from the synthetic library by clonal selection of the cells carrying the cDNA library. Thus, the process which includes the construction of a cDNA library from mRNA and isolation of distinct cDNA clones yields an approximately 10−6-fold purification of the native message. Thus, purification of at least one order of magnitude, preferably two or three orders, and more preferably four or five orders of magnitude is expressly contemplated. Thus, the term “substantially pure” refers to a preparation comprising at least 50-60% by weight the compound of interest (e.g., nucleic acid, oligonucleotide, etc.). More preferably, the preparation comprises at least 75% by weight, and most preferably 90-99% by weight, the compound of interest. Purity is measured by methods appropriate for the compound of interest.
  • The term “complementary” describes two nucleotides that can form multiple favorable interactions with one another. For example, adenine is complementary to thymine as they can form two hydrogen bonds. Similarly, guanine and cytosine are complementary since they can form three hydrogen bonds. Thus if a nucleic acid sequence contains the following sequence of bases, thymine, adenine, guanine and cytosine, a “complement” of this nucleic acid molecule would be a molecule containing adenine in the place of thymine, thymine in the place of adenine, cytosine in the place of guanine, and guanine in the place of cytosine. Because the complement can contain a nucleic acid sequence that forms optimal interactions with the parent nucleic acid molecule, such a complement can bind with high affinity to its parent molecule.
  • With respect to single stranded nucleic acids, particularly oligonucleotides, the term “specifically hybridizing” refers to the association between two single-stranded nucleotide molecules of sufficiently complementary sequence to permit such hybridization under pre-determined conditions generally used in the art (sometimes termed “substantially complementary”). In particular, the term refers to hybridization of an oligonucleotide with a substantially complementary sequence contained within a single-stranded DNA or RNA molecule of the invention, to the substantial exclusion of hybridization of the oligonucleotide with single-stranded nucleic acids of non-complementary sequence. For example, specific hybridization can refer to a sequence which hybridizes to any T1D specific marker gene or nucleic acid, but does not hybridize to other human nucleotides. Also polynucleotide which Aspecifically hybridizes@ may hybridize only to a T1D specific marker, such a T1D-specific marker shown in Tables 1-3. Appropriate conditions enabling specific hybridization of single stranded nucleic acid molecules of varying complementarity are well known in the art.
  • For instance, one common formula for calculating the stringency conditions required to achieve hybridization between nucleic acid molecules of a specified sequence homology is set forth below (Sambrook et al., Molecular Cloning, Cold Spring Harbor Laboratory (1989):

  • T m=81.5″ C+16.6 Log[Na+]+0.41(% G+C)−0.63 (% formamide)−600/#bp in duplex
  • As an illustration of the above formula, using [Na+]=[0.368] and 50% formamide, with GC content of 42% and an average probe size of 200 bases, the Tm is 57″ C. The Tm of a DNA duplex decreases by 1-1.5″ C with every 1% decrease in homology. Thus, targets with greater than about 75% sequence identity would be observed using a hybridization temperature of 42″ C.
  • The stringency of the hybridization and wash depend primarily on the salt concentration and temperature of the solutions. In general, to maximize the rate of annealing of the probe with its target, the hybridization is usually carried out at salt and temperature conditions that are 20-25° C. below the calculated Tm of the hybrid. Wash conditions should be as stringent as possible for the degree of identity of the probe for the target. In general, wash conditions are selected to be approximately 12-20° C. below the Tm of the hybrid. In regards to the nucleic acids of the current invention, a moderate stringency hybridization is defined as hybridization in 6×SSC, 5× Denhardt's solution, 0.5% SDS and 100 μg/ml denatured salmon sperm DNA at 42° C., and washed in 2×SSC and 0.5% SDS at 55° C. for 15 minutes. A high stringency hybridization is defined as hybridization in 6×SSC, 5× Denhardt's solution, 0.5% SDS and 100 μg/ml denatured salmon sperm DNA at 42° C., and washed in 1×SSC and 0.5% SDS at 65° C. for 15 minutes. A very high stringency hybridization is defined as hybridization in 6×SSC, 5× Denhardt's solution, 0.5% SDS and 100 μg/ml denatured salmon sperm DNA at 42° C., and washed in 0.1×SSC and 0.5% SDS at 65° C. for 15 minutes.
  • The term “oligonucleotide” or “oligo” as used herein means a short sequence of DNA or DNA derivatives typically 8 to 35 nucleotides in length, primers, or probes. An oligonucleotide can be derived synthetically, by cloning or by amplification. An oligo is defined as a nucleic acid molecule comprised of two or more ribo- or deoxyribonucleotides, preferably more than three. The exact size of the oligonucleotide will depend on various factors and on the particular application and use of the oligonucleotide. The term “derivative” is intended to include any of the above described variants when comprising an additional chemical moiety not normally a part of these molecules. These chemical moieties can have varying purposes including, improving solubility, absorption, biological half life, decreasing toxicity and eliminating or decreasing undesirable side effects.
  • The term “probe” as used herein refers to an oligonucleotide, polynucleotide or nucleic acid, either RNA or DNA, whether occurring naturally as in a purified restriction enzyme digest or produced synthetically, which is capable of annealing with or specifically hybridizing to a nucleic acid with sequences complementary to the probe. A probe may be either single-stranded or double-stranded. The exact length of the probe will depend upon many factors, including temperature, source of probe and use of the method. For example, for diagnostic applications, depending on the complexity of the target sequence, the oligonucleotide probe typically contains 15-25 or more nucleotides, although it may contain fewer nucleotides. The probes herein are selected to be complementary to different strands of a particular target nucleic acid sequence. This means that the probes must be sufficiently complementary so as to be able to “specifically hybridize” or anneal with their respective target strands under a set of pre-determined conditions. Therefore, the probe sequence need not reflect the exact complementary sequence of the target. For example, a non-complementary nucleotide fragment may be attached to the 5′ or 3′ end of the probe, with the remainder of the probe sequence being complementary to the target strand. Alternatively, non-complementary bases or longer sequences can be interspersed into the probe, provided that the probe sequence has sufficient complementarity with the sequence of the target nucleic acid to anneal therewith specifically.
  • The term “primer” as used herein refers to an oligonucleotide, either RNA or DNA, either single-stranded or double-stranded, either derived from a biological system, generated by restriction enzyme digestion, or produced synthetically which, when placed in the proper environment, is able to functionally act as an initiator of template-dependent nucleic acid synthesis. When presented with an appropriate nucleic acid template, suitable nucleoside triphosphate precursors of nucleic acids, a polymerase enzyme, suitable cofactors and conditions such as a suitable temperature and pH, the primer may be extended at its 3′ terminus by the addition of nucleotides by the action of a polymerase or similar activity to yield a primer extension product. The primer may vary in length depending on the particular conditions and requirement of the application. For example, in diagnostic applications, the oligonucleotide primer is typically 15-25 or more nucleotides in length. The primer must be of sufficient complementarity to the desired template to prime the synthesis of the desired extension product, that is, to be able anneal with the desired template strand in a manner sufficient to provide the 3′ hydroxyl moiety of the primer in appropriate juxtaposition for use in the initiation of synthesis by a polymerase or similar enzyme. It is not required that the primer sequence represent an exact complement of the desired template. For example, a non-complementary nucleotide sequence may be attached to the 5′ end of an otherwise complementary primer. Alternatively, non-complementary bases may be interspersed within the oligonucleotide primer sequence, provided that the primer sequence has sufficient complementarity with the sequence of the desired template strand to functionally provide a template-primer complex for the synthesis of the extension product.
  • Polymerase chain reaction (PCR) has been described in U.S. Pat. Nos. 4,683,195, 4,800,195, and 4,965,188, the entire disclosures of which are incorporated by reference herein.
  • An “siRNA” refers to a molecule involved in the RNA interference process for a sequence-specific post-transcriptional gene silencing or gene knockdown by providing small interfering RNAs (siRNAs) that has homology with the sequence of the targeted gene. Small interfering RNAs (siRNAs) can be synthesized in vitro or generated by ribonuclease III cleavage from longer dsRNA and are the mediators of sequence-specific mRNA degradation. Preferably, the siRNA of the invention are chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional DNA/RNA synthesizer. The siRNA can be synthesized as two separate, complementary RNA molecules, or as a single RNA molecule with two complementary regions. Commercial suppliers of synthetic RNA molecules or synthesis reagents include Applied Biosystems (Foster City, Calif., USA), Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, Colo., USA), Pierce Chemical (part of Perbio Science, Rockford, Ill., USA), Glen Research (Sterling, Va., USA), ChemGenes (Ashland, Mass., USA) and Cruachem (Glasgow, UK). Specific siRNA constructs for inhibiting UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7 mRNA may be between 15-35 nucleotides in length, and more typically about 21 nucleotides in length. Exemplary siRNA molecules which downregulate the aforementioned gene targets are provided in Tables 6-10.
  • The term “vector” relates to a single or double stranded circular nucleic acid molecule that can be infected, transfected or transformed into cells and replicate independently or within the host cell genome. A circular double stranded nucleic acid molecule can be cut and thereby linearized upon treatment with restriction enzymes. An assortment of vectors, restriction enzymes, and the knowledge of the nucleotide sequences that are targeted by restriction enzymes are readily available to those skilled in the art, and include any replicon, such as a plasmid, cosmid, bacmid, phage or virus, to which another genetic sequence or element (either DNA or RNA) may be attached so as to bring about the replication of the attached sequence or element. A nucleic acid molecule of the invention can be inserted into a vector by cutting the vector with restriction enzymes and ligating the two pieces together.
  • Many techniques are available to those skilled in the art to facilitate transformation, transfection, or transduction of the expression construct into a prokaryotic or eukaryotic organism. The terms “transformation”, “transfection”, and Atransduction@ refer to methods of inserting a nucleic acid and/or expression construct into a cell or host organism. These methods involve a variety of techniques, such as treating the cells with high concentrations of salt, an electric field, or detergent, to render the host cell outer membrane or wall permeable to nucleic acid molecules of interest, microinjection, peptide-tethering, PEG-fusion, and the like.
  • The term “promoter element” describes a nucleotide sequence that is incorporated into a vector that, once inside an appropriate cell, can facilitate transcription factor and/or polymerase binding and subsequent transcription of portions of the vector DNA into mRNA. In one embodiment, the promoter element of the present invention precedes the 5′ end of the T1D specific marker nucleic acid molecule such that the latter is transcribed into mRNA. Host cell machinery then translates mRNA into a polypeptide.
  • Those skilled in the art will recognize that a nucleic acid vector can contain nucleic acid elements other than the promoter element and the T1D specific marker gene nucleic acid molecule. These other nucleic acid elements include, but are not limited to, origins of replication, ribosomal binding sites, nucleic acid sequences encoding drug resistance enzymes or amino acid metabolic enzymes, and nucleic acid sequences encoding secretion signals, localization signals, or signals useful for polypeptide purification.
  • A “replicon” is any genetic element, for example, a plasmid, cosmid, bacmid, plastid, phage or virus that is capable of replication largely under its own control. A replicon may be either RNA or DNA and may be single or double stranded.
  • An “expression operon” refers to a nucleic acid segment that may possess transcriptional and translational control sequences, such as promoters, enhancers, translational start signals (e.g., ATG or AUG codons), polyadenylation signals, terminators, and the like, and which facilitate the expression of a polypeptide coding sequence in a host cell or organism.
  • As used herein, the terms “reporter,” “reporter system”, “reporter gene,” or “reporter gene product” shall mean an operative genetic system in which a nucleic acid comprises a gene that encodes a product that when expressed produces a reporter signal that is a readily measurable, e.g., by biological assay, immunoassay, radio immunoassay, or by colorimetric, fluorogenic, chemiluminescent or other methods. The nucleic acid may be either RNA or DNA, linear or circular, single or double stranded, antisense or sense polarity, and is operatively linked to the necessary control elements for the expression of the reporter gene product. The required control elements will vary according to the nature of the reporter system and whether the reporter gene is in the form of DNA or RNA, but may include, but not be limited to, such elements as promoters, enhancers, translational control sequences, poly A addition signals, transcriptional termination signals and the like.
  • The introduced nucleic acid may or may not be integrated (covalently linked) into nucleic acid of the recipient cell or organism. In bacterial, yeast, plant and mammalian cells, for example, the introduced nucleic acid may be maintained as an episomal element or independent replicon such as a plasmid. Alternatively, the introduced nucleic acid may become integrated into the nucleic acid of the recipient cell or organism and be stably maintained in that cell or organism and further passed on or inherited to progeny cells or organisms of the recipient cell or organism. Finally, the introduced nucleic acid may exist in the recipient cell or host organism only transiently.
  • The term “selectable marker gene@ refers to a gene that when expressed confers a selectable phenotype, such as antibiotic resistance, on a transformed cell.
  • The term “operably linked” means that the regulatory sequences necessary for expression of the coding sequence are placed in the DNA molecule in the appropriate positions relative to the coding sequence so as to effect expression of the coding sequence. This same definition is sometimes applied to the arrangement of transcription units and other transcription control elements (e.g. enhancers) in an expression vector.
  • The terms Arecombinant organism,” or Atransgenic organism@ refer to organisms which have a new combination of genes or nucleic acid molecules. A new combination of genes or nucleic acid molecules can be introduced into an organism using a wide array of nucleic acid manipulation techniques available to those skilled in the art. The term “organism” relates to any living being comprised of a least one cell. An organism can be as simple as one eukaryotic cell or as complex as a mammal. Therefore, the phrase Aa recombinant organism@ encompasses a recombinant cell, as well as eukaryotic and prokaryotic organism.
  • The term “isolated protein” or “isolated and purified protein” is sometimes used herein. This term refers primarily to a protein produced by expression of an isolated nucleic acid molecule of the invention. Alternatively, this term may refer to a protein that has been sufficiently separated from other proteins with which it would naturally be associated, so as to exist in “substantially pure” form. “Isolated” is not meant to exclude artificial or synthetic mixtures with other compounds or materials, or the presence of impurities that do not interfere with the fundamental activity, and that may be present, for example, due to incomplete purification, addition of stabilizers, or compounding into, for example, immunogenic preparations or pharmaceutically acceptable preparations.
  • A “specific binding pair” comprises a specific binding member (sbm) and a binding partner (bp) which have a particular specificity for each other and which in normal conditions bind to each other in preference to other molecules. Examples of specific binding pairs are antigens and antibodies, ligands and receptors and complementary nucleotide sequences. The skilled person is aware of many other examples. Further, the term Aspecific binding pair@ is also applicable where either or both of the specific binding member and the binding partner comprise a part of a large molecule. In embodiments in which the specific binding pair comprises nucleic acid sequences, they will be of a length to hybridize to each other under conditions of the assay, preferably greater than 10 nucleotides long, more preferably greater than 15 or 20 nucleotides long. “Sample” or “patient sample” or “biological sample” generally refers to a sample which may be tested for a particular molecule, preferably a T1D specific marker molecule, such as a marker shown in Tables 1-4. Samples may include but are not limited to cells, body fluids, including blood, serum, plasma, urine, saliva, tears, pleural fluid and the like.
  • The terms “agent” and “test compound” are used interchangeably herein and denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials such as bacteria, plants, fungi, or animal (particularly mammalian) cells or tissues. Biological macromolecules include siRNA, shRNA, antisense oligonucleotides, small molecules, antibodies, peptides, peptide/DNA complexes, and any nucleic acid based molecule, for example an oligo, which exhibits the capacity to modulate the activity of the SNP containing nucleic acids described herein or their encoded proteins. Agents are evaluated for potential biological activity by inclusion in screening assays described herein below.
  • The term “modulate” as used herein refers increasing or decreasing. For example, the term modulate refers to the ability of a compound or test agent to interfere with signaling or activity of a gene or protein of the present invention. Therefore, modulating the signaling mediated by the target genes disclosed herein (e.g., UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7) means that an agent or compound inhibits or enhances the activity of the proteins encoded by the gene. This includes altering the activity of natural killer cells, and preventing autoimmune beta cell destruction.
  • Methods of Using T1D-Associated SNPs for T1D Detection Assays
  • T1D SNP containing nucleic acids, including but not limited to those listed in Tables 1-5, may be used for a variety of purposes in accordance with the present invention. T1D-associated SNP containing DNA, RNA, or fragments thereof may be used as probes to detect the presence of and/or expression of T1D specific markers. Methods in which T1D specific marker nucleic acids may be utilized as probes for such assays include, but are not limited to: (1) in situ hybridization; (2) Southern hybridization (3) northern hybridization; and (4) assorted amplification reactions such as polymerase chain reactions (PCR).
  • Further, assays for detecting T1D-associated SNPs may be conducted on any type of biological sample, including but not limited to body fluids (including blood, urine, serum, gastric lavage), any type of cell (such as white blood cells, mononuclear cells) or body tissue.
  • From the foregoing discussion, it can be seen that T1D associated SNP containing nucleic acids, vectors expressing the same, T1D SNP containing marker proteins and anti-T1D specific marker antibodies of the invention can be used to detect T1D associated SNPs in body tissue, cells, or fluid, and alter T1D SNP containing marker protein expression for purposes of assessing the genetic and protein interactions involved in T1D.
  • In most embodiments for screening for T1D-associated SNPs, the T1D-associated SNP containing nucleic acid in the sample will initially be amplified, e.g. using PCR, to increase the amount of the template as compared to other sequences present in the sample. This allows the target sequences to be detected with a high degree of sensitivity if they are present in the sample. This initial step may be avoided by using highly sensitive array techniques that are becoming increasingly important in the art.
  • Alternatively, new detection technologies can overcome this limitation and enable analysis of small samples containing as little as 1 μg of total RNA. Using Resonance Light Scattering (RLS) technology, as opposed to traditional fluorescence techniques, multiple reads can detect low quantities of mRNAs using biotin labeled hybridized targets and anti-biotin antibodies. Another alternative to PCR amplification involves planar wave guide technology (PWG) to increase signal-to-noise ratios and reduce background interference. Both techniques are commercially available from Qiagen Inc. (USA).
  • Thus, any of the aforementioned techniques may be used to detect or quantify T1D-associated SNP marker expression and accordingly, detect patient susceptibility for developing T1D.
  • Kits and Articles of Manufacture
  • Any of the aforementioned products can be incorporated into a kit which may contain an T1D-associated SNP specific marker polynucleotide or one or more such markers immobilized on a Gene Chip, an oligonucleotide, a polypeptide, a peptide, an antibody, a label, marker, or reporter, a pharmaceutically acceptable carrier, a physiologically acceptable carrier, instructions for use, a container, a vessel for administration, an assay substrate, or any combination thereof.
  • Methods of Using T1D-Associated SNPs for Development of Therapeutic Agents
  • Since the SNPs identified herein have been associated with the etiology of T1D, methods for identifying agents that modulate the activity of the genes and their encoded products containing such SNPs should result in the generation of efficacious therapeutic agents for the treatment of a variety of disorders associated with this condition.
  • Chromosomes 21, 6, 15, 9 and 1 contain regions which provide suitable targets for the rational design of therapeutic agents which modulate the activity of proteins encoded by these sequences. Small nucleic acid molecules or peptides corresponding to these regions may be used to advantage in the design of therapeutic agents which effectively modulate the activity of the encoded proteins.
  • Molecular modeling should facilitate the identification of specific organic molecules with capacity to bind to the active site of the proteins encoded by the SNP containing nucleic acids based on conformation or key amino acid residues required for function. A combinatorial chemistry approach will be used to identify molecules with greatest activity and then iterations of these molecules will be developed for further cycles of screening. In certain embodiments, candidate drugs can be screened from large libraries of synthetic or natural compounds. One example is an FDA approved library of compounds that can be used by humans. In addition, compound libraries are commercially available from a number of companies including but not limited to Maybridge Chemical Co. (Trevillet, Cornwall, UK), Comgenex (Princeton, N.J.), Microsource (New Milford, Conn.), Aldrich (Milwaukee, Wis.), AKos Consulting and Solutions GmbH (Basel, Switzerland), Ambinter (Paris, France), Asinex (Moscow, Russia), Aurora (Graz, Austria), BioFocus DPI, Switzerland, Bionet (Camelford, UK), ChemBridge, (San Diego, Calif.), ChemDiv, (San Diego, Calif.), Chemical Block Lt, (Moscow, Russia), ChemStar (Moscow, Russia), Exclusive Chemistry, Ltd (Obninsk, Russia), Enamine (Kiev, Ukraine), Evotec (Hamburg, Germany), Indofine (Hillsborough, NJ), Interbioscreen (Moscow, Russia), Interchim (Montlucon, France), Life Chemicals, Inc. (Orange, Conn.), Microchemistry Ltd. (Moscow, Russia), Otava, (Toronto, ON), PharmEx Ltd. (Moscow, Russia), Princeton Biomolecular (Monmouth Junction, N.J.), Scientific Exchange (Center Ossipee, N.H.), Specs (Delft, Netherlands), TimTec (Newark, Del.), Toronto Research Corp. (North York ON), UkrOrgSynthesis (Kiev, Ukraine), Vitas-M, (Moscow, Russia), Zelinsky Institute, (Moscow, Russia), and Bicoll (Shanghai, China).
  • Libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are commercially available or can be readily prepared by methods well known in the art. It is proposed that compounds isolated from natural sources, such as animals, bacteria, fungi, plant sources, including leaves and bark, and marine samples may be assayed as candidates for the presence of potentially useful pharmaceutical agents. It will be understood that the pharmaceutical agents to be screened could also be derived or synthesized from chemical compositions or man-made compounds. Several commercial libraries can be used in the screens.
  • The polypeptides or fragments employed in drug screening assays may either be free in solution, affixed to a solid support or within a cell. One method of drug screening utilizes eukaryotic or prokaryotic host cells which are stably transformed with recombinant polynucleotides expressing the polypeptide or fragment, preferably in competitive binding assays. Alternatively, primary cells may be isolated from donors expressing the minor or major SNP alleles associated with the T1D described herein. Such cells, either in viable or fixed form, can be used for standard binding assays. One may determine, for example, formation of complexes between the polypeptide or fragment and the agent being tested, or examine the degree to which the formation of a complex between the polypeptide or fragment and a known substrate is interfered with by the agent being tested.
  • Another technique for drug screening provides high throughput screening for compounds having suitable binding affinity for the encoded polypeptides and is described in detail in Geysen, PCT published application WO 84/03564, published on Sep. 13, 1984. Briefly stated, large numbers of different, small peptide test compounds, such as those described above, are synthesized on a solid substrate, such as plastic pins or some other surface. The peptide test compounds are reacted with the target polypeptide and washed. Bound polypeptide is then detected by methods well known in the art.
  • A further technique for drug screening involves the use of host eukaryotic cell lines or cells (such as described above) which have a nonfunctional or altered T1D associated gene. These host cell lines or cells are defective at the polypeptide level. The host cell lines or cells are grown in the presence of drug compound. The rate of cellular metabolism of the host cells is measured to determine if the compound is capable of regulating cellular metabolism in the defective cells. Host cells contemplated for use in the present invention include but are not limited to bacterial cells, fungal cells, insect cells, mammalian cells, and plant cells. The T1D-associated SNP encoding DNA molecules may be introduced singly into such host cells or in combination to assess the phenotype of cells conferred by such expression. Alternatively, donor cells expressing the alleles described herein may be employed. Methods for introducing DNA molecules are also well known to those of ordinary skill in the art. Such methods are set forth in Ausubel et al. eds., Current Protocols in Molecular Biology, John Wiley & Sons, NY, N.Y. 1995, the disclosure of which is incorporated by reference herein.
  • Cells and cell lines suitable for studying the effects of the SNP encoding nucleic acids on glucose metabolism and methods of use thereof for drug discovery are provided. Such cells and cell lines will either already express the SNP or be transfected with the SNP encoding nucleic acids described herein and the effects on glucagon secretion, insulin secretion and/or beta cell apoptosis can be determined. Such cells and cell lines will also be contacted with the siRNA molecules provided herein to assess the effects thereof on glucagon secretion, insulin secretion and/or beta cell apoptosis. The siRNA molecules will be tested alone and in combination of 2, 3, 4, and 5 siRNAs to identify the most efficacious combination for down regulating at least one target gene, e.g., UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7. Cells suitable for these purposes include, without limitation, INS cells (ATCC CRL 11605), PC12 cells (ATCC CRL 1721), MIN6 cells, alpha-TC6 cells and INS-1 832/13 cells (Fernandez et al., J. of Proteome Res. (2007). 7:400-411). Pancreatic islet cells can be isolated and cultured as described in Joseph, J. et al., (J. Biol. Chem. (2004) 279:51049). Diao et al. (J. Biol. Chem. (2005) 280:33487-33496), provide methodology for assessing the effects of the SNP encoding nucleic acids and/or the siRNAs provided herein on glucagon secretion and insulin secretion. Park, J. et al. (J. of Bioch. and Mol. Biol. (2007) 40:1058-68) provide methodology for assessing the effect of these nucleic acid molecules on glucosamine induced beta cell apoptosis in pancreatic islet cells.
  • A wide variety of expression vectors are available that can be modified to express the novel DNA or RNA sequences of this invention. The specific vectors exemplified herein are merely illustrative, and are not intended to limit the scope of the invention. Expression methods are described by Sambrook et al. Molecular Cloning: A Laboratory Manual or Current Protocols in Molecular Biology 16.3-17.44 (1989). Expression methods in Saccharomyces are also described in Current Protocols in Molecular Biology (1989).
  • Suitable vectors for use in practicing the invention include prokaryotic vectors such as the pNH vectors (Stratagene Inc., 11099 N. Torrey Pines Rd., La Jolla, Calif. 92037), pET vectors (Novogen Inc., 565 Science Dr., Madison, Wis. 53711) and the pGEX vectors (Pharmacia LKB Biotechnology Inc., Piscataway, N.J. 08854). Examples of eukaryotic vectors useful in practicing the present invention include the vectors pRc/CMV, pRc/RSV, and pREP (Invitrogen, 11588 Sorrento Valley Rd., San Diego, Calif. 92121); pcDNA3.1/V5&His (Invitrogen); baculovirus vectors such as pVL1392, pVL1393, or pAC360 (Invitrogen); and yeast vectors such as YRP17, YIPS, and YEP24 (New England Biolabs, Beverly, Mass.), as well as pRS403 and pRS413 Stratagene Inc.); Picchia vectors such as pHIL-D1 (Phillips Petroleum Co., Bartlesville, Okla. 74004); retroviral vectors such as PLNCX and pLPCX (Clontech); and adenoviral and adeno-associated viral vectors.
  • Promoters for use in expression vectors of this invention include promoters that are operable in prokaryotic or eukaryotic cells. Promoters that are operable in prokaryotic cells include lactose (lac) control elements, bacteriophage lambda (pL) control elements, arabinose control elements, tryptophan (trp) control elements, bacteriophage T7 control elements, and hybrids thereof. Promoters that are operable in eukaryotic cells include Epstein Barr virus promoters, adenovirus promoters, SV40 promoters, Rous Sarcoma Virus promoters, cytomegalovirus (CMV) promoters, baculovirus promoters such as AcMNPV polyhedrin promoter, Picchia promoters such as the alcohol oxidase promoter, and Saccharomyces promoters such as the gal4 inducible promoter and the PGK constitutive promoter, as well as neuronal-specific platelet-derived growth factor promoter (PDGF), and the Thy-1 promoter.
  • In addition, a vector of this invention may contain any one of a number of various markers facilitating the selection of a transformed host cell. Such markers include genes associated with temperature sensitivity, drug resistance, or enzymes associated with phenotypic characteristics of the host organisms.
  • Host cells expressing the T1D-associated SNPs of the present invention or functional fragments thereof provide a system in which to screen potential compounds or agents for the ability to modulate the development of T1D. Thus, in one embodiment, the nucleic acid molecules of the invention may be used to create recombinant cell lines for use in assays to identify agents which modulate aspects of the diabetic phenotype. Also provided herein are methods to screen for compounds capable of modulating the function of proteins encoded by the SNP containing nucleic acids described below.
  • Another approach entails the use of phage display libraries engineered to express fragment of the polypeptides encoded by the SNP containing nucleic acids on the phage surface. Such libraries are then contacted with a combinatorial chemical library under conditions wherein binding affinity between the expressed peptide and the components of the chemical library may be detected. U.S. Pat. Nos. 6,057,098 and 5,965,456 provide methods and apparatus for performing such assays.
  • The goal of rational drug design is to produce structural analogs of biologically active polypeptides of interest or of small molecules with which they interact (e.g., agonists, antagonists, inhibitors) in order to fashion drugs which are, for example, more active or stable forms of the polypeptide, or which, e.g., enhance or interfere with the function of a polypeptide in vivo. See, e.g., Hodgson, (1991) Bio/Technology 9:19-21. In one approach, discussed above, the three-dimensional structure of a protein of interest or, for example, of the protein-substrate complex, is solved by x-ray crystallography, by nuclear magnetic resonance, by computer modeling or most typically, by a combination of approaches. Less often, useful information regarding the structure of a polypeptide may be gained by modeling based on the structure of homologous proteins. An example of rational drug design is the development of HIV protease inhibitors (Erickson et al., (1990) Science 249:527-533). In addition, peptides may be analyzed by an alanine scan (Wells, (1991) Meth. Enzym. 202:390-411). In this technique, an amino acid residue is replaced by Ala, and its effect on the peptide's activity is determined. Each of the amino acid residues of the peptide is analyzed in this manner to determine the important regions of the peptide.
  • It is also possible to isolate a target-specific antibody, selected by a functional assay, and then to solve its crystal structure. In principle, this approach yields a pharmacophore upon which subsequent drug design can be based.
  • One can bypass protein crystallography altogether by generating anti-idiotypic antibodies (anti-ids) to a functional, pharmacologically active antibody. As a mirror image of a mirror image, the binding site of the anti-ids would be expected to be an analog of the original molecule. The anti-id could then be used to identify and isolate peptides from banks of chemically or biologically produced banks of peptides. Selected peptides would then act as the pharmacophore.
  • Thus, one may design drugs which have, e.g., improved polypeptide activity or stability or which act as inhibitors, agonists, antagonists, etc. of polypeptide activity. By virtue of the availability of SNP containing nucleic acid sequences described herein, sufficient amounts of the encoded polypeptide may be made available to perform such analytical studies as x-ray crystallography. In addition, the knowledge of the protein sequence provided herein will guide those employing computer modeling techniques in place of, or in addition to x-ray crystallography.
  • In another embodiment, the availability of T1D-associated SNP containing nucleic acids enables the production of strains of laboratory mice carrying the T1D-associated SNPs of the invention. Transgenic mice expressing the T1D-associated SNP of the invention provide a model system in which to examine the role of the protein encoded by the SNP containing nucleic acid in the development and progression towards T1D. Methods of introducing transgenes in laboratory mice are known to those of skill in the art. Three common methods include: (1) integration of retroviral vectors encoding the foreign gene of interest into an early embryo; (2) injection of DNA into the pronucleus of a newly fertilized egg; and (3) the incorporation of genetically manipulated embryonic stem cells into an early embryo. Production of the transgenic mice described above will facilitate the molecular elucidation of the role that a target protein plays in various cellular metabolic processes, including: aberrant lipid deposition, altered cellular metabolism and glucose regulation. Such mice provide an in vivo screening tool to study putative therapeutic drugs in a whole animal model and are encompassed by the present invention.
  • The term “animal” is used herein to include all vertebrate animals, except humans. It also includes an individual animal in all stages of development, including embryonic and fetal stages. A “transgenic animal” is any animal containing one or more cells bearing genetic information altered or received, directly or indirectly, by deliberate genetic manipulation at the subcellular level, such as by targeted recombination or microinjection or infection with recombinant virus. The term “transgenic animal” is not meant to encompass classical cross-breeding or in vitro fertilization, but rather is meant to encompass animals in which one or more cells are altered by or receive a recombinant DNA molecule. This molecule may be specifically targeted to a defined genetic locus, be randomly integrated within a chromosome, or it may be extra-chromosomally replicating DNA. The term “germ cell line transgenic animal” refers to a transgenic animal in which the genetic alteration or genetic information was introduced into a germ line cell, thereby conferring the ability to transfer the genetic information to offspring. If such offspring, in fact, possess some or all of that alteration or genetic information, then they, too, are transgenic animals.
  • The alteration of genetic information may be foreign to the species of animal to which the recipient belongs, or foreign only to the particular individual recipient, or may be genetic information already possessed by the recipient. In the last case, the altered or introduced gene may be expressed differently than the native gene. Such altered or foreign genetic information would encompass the introduction of T1D-associated SNP containing nucleotide sequences.
  • The DNA used for altering a target gene may be obtained by a wide variety of techniques that include, but are not limited to, isolation from genomic sources, preparation of cDNAs from isolated mRNA templates, direct synthesis, or a combination thereof.
  • A preferred type of target cell for transgene introduction is the embryonal stem cell (ES). ES cells may be obtained from pre-implantation embryos cultured in vitro (Evans et al., (1981) Nature 292:154-156; Bradley et al., (1984) Nature 309:255-258; Gossler et al., (1986) Proc. Natl. Acad. Sci. 83:9065-9069). Transgenes can be efficiently introduced into the ES cells by standard techniques such as DNA transfection or by retrovirus-mediated transduction. The resultant transformed ES cells can thereafter be combined with blastocysts from a non-human animal. The introduced ES cells thereafter colonize the embryo and contribute to the germ line of the resulting chimeric animal.
  • One approach to the problem of determining the contributions of individual genes and their expression products is to use isolated T1D-associated SNP genes as insertional cassettes to selectively inactivate a wild-type gene in totipotent ES cells (such as those described above) and then generate transgenic mice. The use of gene-targeted ES cells in the generation of gene-targeted transgenic mice was described, and is reviewed elsewhere (Frohman et al., (1989) Cell 56:145-147; Bradley et al., (1992) Bio/Technology 10:534-539).
  • Techniques are available to inactivate or alter any genetic region to a mutation desired by using targeted homologous recombination to insert specific changes into chromosomal alleles. However, in comparison with homologous extra-chromosomal recombination, which occurs at a frequency approaching 100%, homologous plasmid-chromosome recombination was originally reported to only be detected at frequencies between 10−6 and 10−3. Non-homologous plasmid-chromosome interactions are more frequent occurring at levels 105-fold to 102 fold greater than comparable homologous insertion.
  • To overcome this low proportion of targeted recombination in murine ES cells, various strategies have been developed to detect or select rare homologous recombinants. One approach for detecting homologous alteration events uses the polymerase chain reaction (PCR) to screen pools of transformant cells for homologous insertion, followed by screening of individual clones. Alternatively, a positive genetic selection approach has been developed in which a marker gene is constructed which will only be active if homologous insertion occurs, allowing these recombinants to be selected directly. One of the most powerful approaches developed for selecting homologous recombinants is the positive-negative selection (PNS) method developed for genes for which no direct selection of the alteration exists. The PNS method is more efficient for targeting genes which are not expressed at high levels because the marker gene has its own promoter. Non-homologous recombinants are selected against by using the Herpes Simplex virus thymidine kinase (HSV-TK) gene and selecting against its nonhomologous insertion with effective herpes drugs such as gancyclovir (GANC) or (1-(2-deoxy-2-fluoro-B-D arabinofluranosyl)-5-iodou-racil, (FIAU). By this counter selection, the number of homologous recombinants in the surviving transformants can be increased. Utilizing T1D-associated SNP containing nucleic acid as a targeted insertional cassette provides means to detect a successful insertion as visualized, for example, by acquisition of immunoreactivity to an antibody immunologically specific for the polypeptide encoded by T1D-associated SNP nucleic acid and, therefore, facilitates screening/selection of ES cells with the desired genotype.
  • As used herein, a knock-in animal is one in which the endogenous murine gene, for example, has been replaced with human T1D-associated SNP containing gene of the invention. Such knock-in animals provide an ideal model system for studying the development of T1D.
  • As used herein, the expression of a T1D-associated SNP containing nucleic acid, fragment thereof, or a T1D-associated SNP fusion protein can be targeted in a “tissue specific manner” or “cell type specific manner” using a vector in which nucleic acid sequences encoding all or a portion of T1D-associated SNP are operably linked to regulatory sequences (e.g., promoters and/or enhancers) that direct expression of the encoded protein in a particular tissue or cell type. Such regulatory elements may be used to advantage for both in vitro and in vivo applications. Promoters for directing tissue specific expression of proteins are well known in the art and described herein.
  • The nucleic acid sequence encoding the T1D-associated SNP of the invention may be operably linked to a variety of different promoter sequences for expression in transgenic animals. Such promoters include, but are not limited to a prion gene promoter such as hamster and mouse Prion promoter (MoPrP), described in U.S. Pat. No. 5,877,399 and in Borchelt et al., Genet. Anal. 13(6) (1996) pages 159-163; a rat neuronal specific enolase promoter, described in U.S. Pat. Nos. 5,612,486, and 5,387,742; a platelet-derived growth factor B gene promoter, described in U.S. Pat. No. 5,811,633; a brain specific dystrophin promoter, described in U.S. Pat. No. 5,849,999; a Thy-1 promoter; a PGK promoter; a CMV promoter; a neuronal-specific platelet-derived growth factor B gene promoter; and Glial fibrillar acidic protein (GFAP) promoter for the expression of transgenes in glial cells.
  • Methods of use for the transgenic mice of the invention are also provided herein. Transgenic mice into which a nucleic acid containing the T1D-associated SNP or its encoded protein have been introduced are useful, for example, to develop screening methods to screen therapeutic agents to identify those capable of modulating the development of T1D.
  • Pharmaceuticals and Peptide Therapies
  • The elucidation of the role played by the T1D associated SNPs described herein in cellular metabolism facilitates the development of pharmaceutical compositions useful for treatment and diagnosis of T1D. These compositions may comprise, in addition to one of the above substances, a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • Whether it is a polypeptide, antibody, peptide, nucleic acid molecule, small molecule or other pharmaceutically useful compound according to the present invention that is to be given to an individual, administration is preferably in a “prophylactically effective amount” or a “therapeutically effective amount” (as the case may be, although prophylaxis may be considered therapy), this being sufficient to show benefit to the individual.
  • As it is presently understood, RNA interference involves a multi-step process. Double stranded RNAs are cleaved by the endonuclease Dicer to generate nucleotide fragments (siRNA). The siRNA duplex is resolved into 2 single stranded RNAs, one strand being incorporated into a protein-containing complex where it functions as guide RNA to direct cleavage of the target RNA (Schwarz et al, Mol. Cell. 10:537 548 (2002), Zamore et al, Cell 101:25 33 (2000)), thus silencing a specific genetic message (see also Zeng et al, Proc. Natl. Acad. Sci. 100:9779 (2003)).
  • The invention includes a method of treating T1D in a mammal. An exemplary method entails administering to the mammal a pharmaceutically effective amount of an siRNA molecule directed to a gene target selected from the group consisting of UBASH3A (GenBank No.: NM018961; SEQ ID NO: 1), GLIS3, (GenBank No.: NM001042413; SEQ ID NO: 2), RASGRP1 (GenBank No.: NM005739; SEQ ID NO: 3), BACH2 (GenBank No.: NM021813; SEQ ID NO: 4) and EDG7 (GenBank Acc. No.: AY322547; SEQ ID NO: 5). The siRNA inhibits the expression of the aforementioned genes. Preferably, the mammal is a human. The term “patient” as used herein refers to a human.
  • Specific siRNA preparations directed at inhibiting the expression of UBASH3A, GLIS3, RASGRP1, BACH2 and EDG7, as well as delivery methods are provided as a novel therapy to treat T1D. See Tables 6-10. The siRNA can be delivered to a patient in vivo either systemically or locally with carriers, as discussed below. The compositions of the invention may be used alone or in combination with other agents or genes encoding proteins to augment the efficacy of the compositions.
  • A “membrane permeant peptide sequence” refers to a peptide sequence which is able to facilitate penetration and entry of the siRNA inhibitor across the cell membrane. Exemplary peptides include with out limitation, the signal sequence from Karposi fibroblast growth factor exemplified herein, the HIV tat peptide (Vives et al., J Biol. Chem., 272:16010-16017, 1997), Nontoxic membrane translocation peptide from protamine (Park et al., FASEB J. 19(11):1555-7, 2005), CHARIOT® delivery reagent (Active Motif; U.S. Pat. No. 6,841,535) and the antimicrobial peptide Buforin 2.
  • In one embodiment of the invention siRNAs are delivered for therapeutic benefit. There are several ways to administer the siRNA of the invention to in vivo to treat T1D including, but not limited to, naked siRNA delivery, siRNA conjugation and delivery, liposome carrier-mediated delivery, polymer carrier delivery, nanoparticle compositions, plasmid-based methods, and the use of viruses.
  • siRNA composition of the invention can comprise a delivery vehicle, including liposomes, for administration to a subject, carriers and diluents and their salts, and/or can be present in pharmaceutically acceptable formulations. This can be necessary to allow the siRNA to cross the cell membrane and escape degradation. Methods for the delivery of nucleic acid molecules are described in Akhtar et al., 1992, Trends Cell Bio., 2, 139; Delivery Strategies for Antisense Oligonucleotide Therapeutics, ed. Akhtar, 1995, Maurer et al., 1999, Mol. Membr. Biol., 16, 129-140; Hofland and Huang, 1999, Handb. Exp. Pharmacol., 137, 165-192; and Lee et al., 2000, ACS Symp. Ser., 752, 184-192; Beigelman et al., U.S. Pat. No. 6,395,713 and Sullivan et al., PCT WO 94/02595 further describe the general methods for delivery of nucleic acid molecules. These protocols can be utilized for the delivery of virtually any nucleic acid molecule.
  • The frequency of administration of the siRNA to a patient will also vary depending on several factors including, but not limited to, the type and severity of the T1D to be treated, the route of administration, the age and overall health of the individual, the nature of the siRNA, and the like. It is contemplated that the frequency of administration of the siRNA to the patient may vary from about once every few months to about once a month, to about once a week, to about once per day, to about several times daily.
  • Pharmaceutical compositions that are useful in the methods of the invention may be administered systemically in parenteral, oral solid and liquid formulations, ophthalmic, suppository, aerosol, topical or other similar formulations. In addition to the appropriate siRNA, these pharmaceutical compositions may contain pharmaceutically-acceptable carriers and other ingredients known to enhance and facilitate drug administration. Thus such compositions may optionally contain other components, such as adjuvants, e.g., aqueous suspensions of aluminum and magnesium hydroxides, and/or other pharmaceutically acceptable carriers, such as saline. Other possible formulations, such as nanoparticles, liposomes, resealed erythrocytes, and immunologically based systems may also be used to administer the appropriate siRNA to a patient according to the methods of the invention. The use of nanoparticles to deliver siRNAs, as well as cell membrane permeable peptide carriers that can be used are described in Crombez et al., Biochemical Society Transactions v35:p44 (2007).
  • Methods of the invention directed to treating T1D involve the administration of at least one UBASH3A, GLIS3, RASGRP1, BACH2 and EDG7 siRNA in a pharmaceutical composition. The siRNA is administered to an individual as a pharmaceutical composition comprising the siRNA and a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers are well known in the art and include aqueous solutions such as physiologically buffered saline, other solvents or vehicles such as glycols, glycerol, oils such as olive oil or injectable organic esters.
  • A pharmaceutically acceptable carrier can contain physiologically acceptable compounds that act, for example, to stabilize the siRNA or increase the absorption of the agent. Such physiologically acceptable compounds include, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients. One skilled in the art would know that the choice of a pharmaceutically acceptable carrier, including a physiologically acceptable compound, depends, for example, on the route of administration of the siRNA.
  • One skilled in the art appreciates that a pharmaceutical composition comprising siRNA can be administered to a subject by various routes including, for example, orally or parenterally, such as intravenously (i.v.), intramuscularly, subcutaneously, intraorbitally, intranasally, intracapsularly, intraperitoneally (i.p.), intracisternally, intratracheally (i.t.), or intra-articularly or by passive or facilitated absorption. The same routes of administration can be used other pharmaceutically useful compounds, for example, small molecules, nucleic acid molecules, peptides, antibodies and polypeptides as discussed hereinabove.
  • A pharmaceutical composition comprising siRNA inhibitor also can be incorporated, if desired, into liposomes, microspheres, microbubbles, or other polymer matrices (Gregoriadis, Liposome Technology, Vols. Ito III, 2nd ed., CRC Press, Boca Raton Fla. (1993)). Liposomes, for example, which consist of phospholipids or other lipids, are nontoxic, physiologically acceptable and metabolizable carriers that are relatively simple to make and administer.
  • The pharmaceutical preparation comprises a siRNA targeting the SNP containing sequences described herein or an expression vector encoding for the siRNA. Such pharmaceutical preparations can be administered to a patient for treating T1D.
  • Expression vectors for the expression of siRNA molecules preferably employ a strong promoter which may be constitutive or regulated. Such promoters are well known in the art and include, but are not limited to, RNA polymerase II promoters, the T7 RNA polymerase promoter, and the RNA polymerase III promoters U6 and H1 (see, e.g., Myslinski et al. (2001) Nucl. Acids Res., 29:2502 09).
  • A formulated siRNA composition can be a composition comprising one or more siRNA molecules or a vector encoding one or more siRNA molecules independently or in combination with a cationic lipid, a neutral lipid, and/or a polyethyleneglycol-diacylglycerol (PEG-DAG) or PEG-cholesterol (PEG-Chol) conjugate. Non-limiting examples of expression vectors are described in Paul et al., 2002, Nature Biotechnology, 19, 505; Miyagishi and Taira, 2002, Nature Biotechnology, 19, 497; Lee et al., 2002, Nature Biotechnology, 19, 500-505.
  • A lipid nanoparticle composition is a composition comprising one or more biologically active molecules independently or in combination with a cationic lipid, a neutral lipid, and/or a polyethyleneglycol-diacylglycerol (i.e., polyethyleneglycol diacylglycerol (PEG-DAG), PEG-cholesterol, or PEG-DMB) conjugate. In one embodiment, the biologically active molecule is encapsulated in the lipid nanoparticle as a result of the process of providing and aqueous solution comprising a biologically active molecule of the invention (i.e., siRNA), providing an organic solution comprising lipid nanoparticle, mixing the two solutions, incubating the solutions, dilution, ultrafiltration, resulting in concentrations suitable to produce nanoparticle compositions.
  • Nucleic acid molecules can be administered to cells by incorporation into other vehicles, such as biodegradable polymers, hydrogels, cyclodextrins. (see for example Gonzalez et al., 1999, Bioconjugate Chem., 10, 1068-1074; Wang et al., International PCT publication Nos. WO 03/47518 and WO 03/46185), poly(lactic-co-glycolic)acid (PLGA) and PLCA microspheres (see for example U.S. Pat. No. 6,447,796 and US Patent Application Publication No. US 2002130430), biodegradable nanocapsules, and bioadhesive microspheres, or by proteinaceous vectors (O′Hare and Normand, International PCT Publication No. WO 00/53722)
  • Cationic lipids and polymers are two classes of non-viral siRNA delivery which can form complexes with negatively charged siRNA. The self-assembly PEG-ylated polycation polyethylenimine (PEI) has also been used to condense and protect siRNAs (Schiffelers et al., 2004, Nuc. Acids Res. 32: 141-110). The siRNA complex can be condensed into a nanoparticle to allow efficient uptake of the siRNA through endocytosis. Also, the nucleic acid-condensing property of protamine has been combined with specific antibodies to deliver siRNAs and can be used in the invention (Song et al., 2005, Nat Biotech. 23:709-717).
  • In order to treat an individual having T1D, to alleviate a sign or symptom of the disease, siRNA should be administered in an effective dose. The total treatment dose can be administered to a subject as a single dose or can be administered using a fractionated treatment protocol, in which multiple doses are administered over a more prolonged period of time, for example, over the period of a day to allow administration of a daily dosage or over a longer period of time to administer a dose over a desired period of time. One skilled in the art would know that the amount of siRNA required to obtain an effective dose in a subject depends on many factors, including the age, weight and general health of the subject, as well as the route of administration and the number of treatments to be administered. In view of these factors, the skilled artisan would adjust the particular dose so as to obtain an effective dose for treating an individual having T1D.
  • The effective dose of siRNA will depend on the mode of administration, and the weight of the individual being treated. The dosages described herein are generally those for an average adult but can be adjusted for the treatment of children. The dose will generally range from about 0.001 mg to about 1000 mg.
  • The concentration of siRNA in a particular formulation will depend on the mode and frequency of administration. A given daily dosage can be administered in a single dose or in multiple doses so long as the siRNA concentration in the formulation results in the desired daily dosage. One skilled in the art can adjust the amount of siRNA in the formulation to allow administration of a single dose or in multiple doses that provide the desired concentration of siRNA over a given period of time.
  • In an individual suffering from T1D, in particular a more severe form of the disease, administration of siRNA can be particularly useful when administered in combination, for example, with a conventional agent for treating such a disease. The skilled artisan would administer siRNA, alone or in combination and would monitor the effectiveness of such treatment using routine methods such as pancreatic beta cell function determination, radiologic, immunologic or, where indicated, histopathologic methods. Other conventional agents for the treatment of diabetes include insulin administration, glucagon administration or agents that alter levels of either of these two molecules. Glucophage®, Avandia®, Actos®, Januvia® and Glucovance® are examples of such agents.
  • Administration of the pharmaceutical preparation is preferably in an “effective amount” this being sufficient to show benefit to the individual. This amount prevents, alleviates, abates, or otherwise reduces the severity of T1D symptoms in a patient.
  • The pharmaceutical preparation is formulated in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form, as used herein, refers to a physically discrete unit of the pharmaceutical preparation appropriate for the patient undergoing treatment. Each dosage should contain a quantity of active ingredient calculated to produce the desired effect in association with the selected pharmaceutical carrier. Procedures for determining the appropriate dosage unit are well known to those skilled in the art.
  • Dosage units may be proportionately increased or decreased based on the weight of the patient. Appropriate concentrations for alleviation of a particular pathological condition may be determined by dosage concentration curve calculations, as known in the art.
  • The methods set forth below are provided to facilitate the practice of the present invention.
  • Signal Distillation
  • We selected SNPs with at least a nominally significant combined P-value (major histocompatibility complex region excluded) from our genome wide genotype data generated on Caucasians from 563 T1D probands and 1,146 controls plus 483 complete T1D family trios, using the Illumina HumanHap550 BeadChip. We then genotyped these SNPs using the Illumina GoldenGate platform in an independent cohort of 939 nuclear T1D families from Montreal and the type 1 diabetes genetics consortium (T1DGC). Subsequently we looked across all three cohorts plus the publicly available Wellcome Trust Case Control Consortium (WTCCC) dataset on the world wide web at wtccc.org.uk17 for T1D to identify SNPs in loci that were both not previously described and nominally significant across all cohorts. We selected five loci for further investigation, which we queried in T1D probands from the DCCT/EDIC study using an independent matched control dataset from Philadelphia which were genotyped on the Illumina 1M and HumanHap550K BeadChips, respectively.
  • Subjects
  • 1. Type 1 Diabetes Cohort from Canada:
  • The Canadian cohort consisted of 1,120 nuclear family trios (one affected child and two parents) and 267 independent T1D cases, collected in pediatric diabetes clinics in Montreal, Toronto, Ottawa and Winnipeg. The median age at onset is 8 with lower and upper quartiles at 4.6 years and 11 years. All patients were diagnosed under the age of 18, were treated with insulin since diagnosis and none has stopped treatment for any reason since. Disease diagnosis was based on these clinical criteria, rather than any laboratory tests. Ethnic backgrounds were of mixed European descent, with the largest single subset (409 families) being French Canadian. The Research Ethics Board of the Montreal Children's Hospital and other participating centers approved the study, and written informed consent was obtained from all subjects.
  • 2. Type 1 Diabetes Genetics Consortium Cohort:
  • The Type 1 Diabetes Genetics Consortium cohort consisted of 549 families (2350 individuals) with at least two children diagnosed with diabetes and both parents available as of the July 2005 data freeze. Criteria were age at diagnosis below 35 years and uninterrupted treatment with insulin within six months of diagnosis. For siblings of probands diagnosed under the age of 35, the age-at-diagnosis limit was extended to 45 if they were lean and had positive antibodies and/or low C-peptide levels at diagnosis. The median age is 8 with quartiles at 4 years and 13 years. The samples were collected in Europe, North America and Australia and most subjects were of European ancestry. Autoantibody results are available but were not used to substantiate the diagnosis, except as noted above.
  • 3. Type 1 Diabetes Cohort from Philadelphia:
  • The T1D cohort consisted of 103 children recruited at the Children's Hospital of Philadelphia (CHOP), since September, 2006. All patients were diagnosed under the age of 18. Of those, 49 T1D patients (32 female, 17 male) were Caucasians by self-report (average age of onset 7.07 years; range 9 months-14 years) and were included in the analysis. All were treated with insulin since diagnosis and none has stopped treatment for any reason since. The Research Ethics Board of CHOP approved the study and written informed consent was obtained from all subjects.
  • 4. The Diabetes Control and Complications Trial/Epidemiology of Diabetes
  • Complications and Interventions (DCCT/EDIC) Type 1 diabetes cohort: The DCCT was a multi-center randomized clinical trial to determine the effect of intensive insulin treatment with respect to reduced development and progression of retinopathy and nephropathy complications in patients with type 1 diabetes19, 20. A total of 1,441 subjects with type 1 diabetes were recruited from 29 centers across North America into the DCCT between 1983 and 1989; they were between 13 and 39 years of age and 53% were male. They were recruited into two cohorts: the primary prevention cohort consisted of 726 subjects with no retinopathy, an albumin excretion rate <28 μg per minute, and diabetes duration of 1 to 5 years and were used to determine if intensive therapy prevented the development of diabetic retinopathy in patients with no retinopathy; the secondary intervention cohort consisted of 715 subjects who had non-proliferative retinopathy, a urinary albumin excretion rate <140 μg per minute, and diabetes duration of 1 to 15 years were studied to determine whether intensive therapy would affect the progression of early retinopathy19. Approval for the DCCT/EDIC Genetics study was provided by the Research Ethics Board of the Hospital for Sick Children, Toronto.
  • The Illumina 1M assay was genotyped on all available probands. To detect and remove outliers due to population stratification from the majority self-reported white probands Eigenstrat21 was used to select probands by sequential analysis. After exclusions of outliers, there were 1303 DCCT/EDIC probands, (695 male, 608 females) with mean age of diagnosis of T1D of 21 years (SD=8, range 0-38).
  • 5. Control Subjects from Philadelphia:
  • The control group included 1,146 children with self reported Caucasian status, mean age 9.42 years; 53.05% male and 46.95% female, who did not have diabetes or a first-degree relative with T1D. The control group used to match with the 1,100 DCCT/EDIC T1D probands included 2,024 children with self reported Caucasian ethnicity, mean age 8.82 years; 50.83% male and 49.17% female, who did not have diabetes or a first-degree relative with T1D. These individual were recruited by CHOP's clinicians and nursing staff within the CHOP's Health Care Network, including four primary care clinics and several group practices and outpatient practices that included well child visits. Of these 2024 individuals, 1673 were selected using population stratification analysis from eigenstrat, similar to that described above for DCCT/EDIC probands (868 males, 801 females, 4 with ambiguous gender). The Research Ethics Board of CHOP approved the study, and written informed consent was obtained from all subjects.
  • Genotyping
  • Genotypes for this study were obtained using the Infinium and GoldenGate platforms from Illumina. We performed high throughput genome-wide SNP genotyping, using the Illumina Infinium™ II HumanHap550 BeadChip technology1,2 (Illumina, San Diego), at the Center for Applied Genomics at CHOP. We used 750 ng of genomic DNA to genotype each sample, according to the manufacturer's guidelines. DCCT/EDIC samples were genotyped on the Illumina 1M chip at Illumina (San Diego, Calif.).
  • Statistics
  • All statistical tests for association were carried out using the software package plink22. The single marker analysis for the genome-wide data was carried out using a χ2 test on allele count differences between 563 cases and 1,146 controls. Odds ratios and the corresponding 95% confidence intervals were calculated for the association analysis. The transmission disequilibrium test was used to calculate P-values on differences between transmitted and untransmitted allele counts in the T1D trios and nuclear families. Counts of untransmitted and transmitted alleles from heterozygous parents to affected offspring were determined using the standard transmission disequilibrium test implemented in the Haploview software package4. The P-values from the case-control and family-based analyses in our three discovery cohorts were combined using Fisher's method5 to quantify the overall evidence for association.
  • The following examples are provided to illustrate certain embodiments of the invention. They are not intended to limit the invention in any way.
  • EXAMPLE I Gentic Loci Associated with TID
  • Type 1 diabetes (T1D) is a multifactorial disease with a strong genetic component that results from autoimmune destruction of the pancreatic β-cells. The major T1D susceptibility locus, mapping to the HLA class II genes at 6p211 and encoding highly polymorphic antigen-presenting proteins, accounts for almost 50% of the genetic risk for T1D2. Several other loci with more modest effects account for another 10-20% of the risk. These include: (1) the insulin (INS) VNTR3, modulating thymic expression of and tolerance to insulin, a major T1D autoantigen4,5; (2) the Arg620Trp single-nucleotide polymorphism (SNP) at PTPN22, which affects the function of a negative regulator of TCR signaling6; (3) Non-coding SNPs at IL2RA7-9, which encodes the α chain of the IL2 receptor complex (CD25), an important modulator of immunity; (4) variants in the CTLA4 locus10 whose protein product transmits inhibitory signals to attenuate T-cell activation. It is worth noting that all of these T1D-associated genes are expressed in cells with immune function and all except INS have been associated with other autoimmune disorders.
  • The recent development of high throughput single nucleotide polymorphism (SNP) genotyping array technologies has enabled us11 and others12,13 to perform genome-wide association (GWA) studies in search of the remaining T1D loci. The first successful use in T1D involved screening of 12,000 nonsynonymous SNPs, which found T1D association with rs1990760, involving an Ala946Thr substitution on the IFIH1 gene (Interferon-Induced with Helicase C domain 1)14. We recently reported the outcome of our GWA for T1D where we examined a large pediatric T1D cohort of European decent followed by a successful TDT replication attempt in an independent cohort15. In addition to confirming the previously identified loci, we observed highly significant association with KIAA0350, the gene product of which has been recently renamed C-type lectin domain family 16 member A (CLEC16A); subsequent follow-up of our data also revealed a locus on 12q1316. The Wellcome Trust Case Control Consortium17 also demonstrated association to the same regions of 16p13 and 12q13 which they subsequently followed up and replicated18, along with other loci on 12q24 and 18p11.
  • We carried out a follow-up strategy to uncover additional novel T1D risk loci. Here, we describe two loci that were significantly associated with T1D during this process, both of which reside in genes that are biologically relevant to autoimmunity. These genes encode ubiquitin-associated and SH3 domain-containing protein A (UBASH3A) and BTB and CNC homology 2 (BACH2), respectively, both of which are known to be involved in T cell signaling.
  • From a combination of our genotyping data generated on T1D probands and controls plus T1D family trios of the same ancestry, we selected 982 SNPs that fulfilled the two criteria of not residing in the major histocompatibility complex and being at least nominally significantly associated with T1D. We then took those SNPs forward with additional genotyping in an independent cohort of nuclear T1D families from Montreal and the T1DGC. As shown in Table 1, thirty three single point associations were at least nominally significant across all four cohorts utilized for the discovery stage of this process. However, the bulk of them had been previously reported and were therefore not novel i.e. they resided at the well-established PTPN226, 12q1316,18, KIAA035015,18, IL2RA7-9, CTLA410 and IFIH114 loci. However, six SNPs residing at five loci fulfilled our criteria for further replication efforts.
  • TABLE 1
    Cohort datasets leveraged in selection of candidate loci for further replication efforts.
    Case-Control Cohort WTCCC
    T1D families - Aff Ctrl Aff Ctrl
    Montreal and T1DGC allele allele T1D family trios Combined allele allele
    Chr SNP on Trans:untrans OR TDT P-value freq freq P OR Trans:untrans OR TDT P-value P freq freq P OR Gene
    1 rs2358994 114230984 398:287 1.387 2.22 × 10−5 0.232 0.175 7.11 × 10−5 1.426 181:127 1.425 0.0021 1.25 × 10−9 0.246 0.179 4.05 × 10−16 1.504 PTPN22
    1 rs12566340 114221851 492:379 1.298 1.29 × 10−4 0.288 0.237 0.0015 1.299 212:164 1.293 0.013 5.53 × 10−7 0.287 0.226 1.09 × 10−11 1.377 PTPN22
    1 rs7529353 114221985 474:354 1.339 3.04 × 10−5 0.294 0.242 9.75 × 10−4 1.309 218:165 1.321 0.0068 5.47 × 10−8 0.287 0.227 2.69 × 10−11 1.368 PTPN22
    1 rs1230661 113987113 456:331 1.378 8.36 × 10−6 0.267 0.216 8.75 × 10−4 1.324 209:161 1.298 0.013 2.68 × 10−8 0.276 0.217 3.34 × 10−11 1.371 PTPN22
    1 rs1217407 114195271 505:395 1.278 2.46 × 10−4 0.298 0.244 7.42 × 10−4 1.316 225:168 1.339 0.004 1.79 × 10−7 0.298 0.240 1.70 × 10−10 1.344 PTPN22
    1 rs4839335 114035394 482:354 1.362 9.56 × 10−6 0.3 0.25 0.002 1.285 224:172 1.302 0.009 4.66 × 10−8 0.299 0.241 2.81 × 10−10 1.339 PTPN22
    12 rs10876864 54687352 631:528 1.195 0.0025 0.458 0.388 8.39 × 10−5 1.336 265:188 1.41 2.97 × 10−4 1.86 × 10−8 0.475 0.414 2.04 × 10−9 1.283 12q13
    12 rs1701704 54698754 549:425 1.292 7.09 × 10−5 0.379 0.303 9.89 × 10−6 1.402 245:180 1.361 0.0016 4.61 × 10−10 0.397 0.339 5.91 × 10−9 1.282 12q13
    16 rs2041670 11082153 384:444 0.865 0.037 0.265 0.345 2.01 × 10−6 0.682 172:233 0.738 0.0024 5.00 × 10−8 0.264 0.315 7.05 × 10−8 0.781 KIAA0350
    16 rs725613 11077184 397:465 0.854 0.021 0.3 0.39 3.24 × 10−7 0.672 178:248 0.718 6.95 × 10−4 1.70 × 10−9 0.292 0.340 3.90 × 10−7 0.797 KIAA0350
    12 rs11171710 54654345 464:574 0.808 6.40 × 10−4 0.405 0.462 0.0016 0.792 197:244 0.807 0.025 4.32 × 10−6 0.404 0.452 2.40 × 10−6 0.821 12q13
    10 rs7072793 6146272 574:507 1.132 0.042 0.486 0.41 2.96 × 10−5 1.358 268:200 1.34 0.0017 4.54 × 10−7 0.455 0.409 6.24 × 10−6 1.207 IL2RA
    10 rs7073236 6146558 566:464 1.22 0.0015 0.487 0.414 5.67 × 10−5 1.343 264:196 1.347 0.0015 3.62 × 10−8 0.455 0.409 7.31 × 10−6 1.205 IL2RA
    10 rs3118470 6141719 504:431 1.169 0.017 0.365 0.306 4.62 × 10−4 1.308 240:181 1.326 0.004 5.29 × 10−6 0.361 0.319 1.32 × 10−5 1.208 IL2RA
    16 rs1035089 10955851 517:451 1.146 0.034 0.48 0.42 8.25 × 10−4 1.277 265:212 1.25 0.015 5.25 × 10−5 0.482 0.439 2.59 × 10−5 1.190 KIAA0350
    2 rs231726 204449111 502:427 1.176 0.014 0.358 0.321 0.028 1.184 228:177 1.288 0.011 4.00 × 10−4 0.372 0.332 5.10 × 10−5 1.191 CTLA4
    2 rs1990760 162832297 422:518 0.815 0.0017 0.398 0.434 0.048 0.864 203:251 0.809 0.024 2.00 × 10−4 0.350 0.389 8.73 × 10−5 0.845 IFIH1
    10 rs706779 6138830 424:506 0.838 0.0072 0.425 0.492 2.60 × 10−4 0.764 185:257 0.72 6.16 × 10−4 2.68 × 10−7 0.419 0.458 0.00012 0.852 IL2RA
    2 rs926169 204430997 521:459 1.135 0.048 0.426 0.38 0.0094 1.212 236:191 1.236 0.029 0.001 0.440 0.402 0.00022 1.167 CTLA4
    2 rs1024161 204429997 534:462 1.156 0.023 0.439 0.397 0.02 1.191 222:173 1.283 0.014 5.00 × 10−4 0.441 0.403 0.00024 1.166 CTLA4
    16 rs13330041 10996309 291:345 0.844 0.032 0.172 0.246 1.01 × 10−5 0.637 145:183 0.792 0.036 2.72 × 10−7 0.175 0.204 0.00028 0.825 KIAA0350
    16 rs17673553 11149407 319:387 0.824 0.01 0.202 0.279 1.30 × 10−6 0.655 146:203 0.719 0.0023 9.89 × 10−9 0.217 0.249 0.00031 0.838 KIAA0350
    2 rs2111485 162818782 417:500 0.834 0.0061 0.393 0.433 0.027 0.849 210:254 0.827 0.041 6.00 × 10−4 0.359 0.395 0.00034 0.858 IFIH1
    1 rs12029644 114338303 222:172 1.291 0.012 0.136 0.105 0.008 1.343 120:80  1.5 0.0047 5.42 × 10−5 0.130 0.109 0.0018 1.218 PTPN22
    21 rs9976767 42709459 571:504 1.13 0.041 0.474 0.437 0.038 1.164 260:203 1.281 0.008 0.001 0.493 0.461 0.002 1.135 UBASH3A*
    9 rs10758593 4282083 539:462 1.17 0.015 0.492 0.426 2.97 × 10 −4 1.303 254:209 1.215 0.037 2.25 × 10 −5 0.440 0.410 0.004 1.129 GLIS3*
    9 rs10758594 4285583 535:456 1.17 0.012 0.513 0.451 6.66 × 10 −4 1.282 253:209 1.211 0.041 4.17 × 10 −5 0.456 0.427 0.004 1.127 GLIS3*
    1 rs7520320 114336816 211:169 1.249 0.031 0.136 0.107 0.013 1.315 124:82  1.512 0.0034 1.00 × 10−4 0.134 0.115 0.0042 1.195 PTPN22
    16 rs12931878 10949695 362:446 0.812 0.0031 0.16 0.225 1.01 × 10−5 0.657 128:162 0.79 0.046 3.30 × 10−7 0.158 0.178 0.0088 0.865 KIAA0350
    15 rs8035957 36625556 423:342 1.24 0.0034 0.304 0.263 0.011 1.225 204:162 1.259 0.028 1.00 × 10 −4 0.292 0.268 0.01 1.126 RASGRP1*
    6 rs3757247 91014184 545:482 1.13 0.049 0.504 0.455 0.0075 1.216 253:209 1.211 0.041 0.001 0.511 0.489 0.033 1.092 BACH2*
    1 rs1983853 85083780 202:254 0.8 0.015 0.121 0.151 0.021 0.779 105:136 0.772 0.046 0.001 0.122 0.137 0.036 0.878 EDG7*
    11 rs1004446 2126719 378:514 0.735 5.27 × 10−6 0.254 0.354 4.38 × 10−9 0.622 160:228 0.7018 5.56 × 10−4 1.02 × 10−14 0.443 0.464 0.047 0.921 INS
    The six SNPs indicated in bold represent novel associations deemed appropriate for further investigation.
    Minor allele frequencies, P-values and odds ratios (OR) are shown.
    Combined P-values for the three discovery cohorts are also shown, together with the gene in which the markers resides or which they are nearest to.
    P-values are two-sided in each instance.
    Aff allele freq, allele frequency in affected individuals;
    Chr, chromosome;
    Ctrl allele freq, allele frequency in unaffected individuals;
    Trans:untrans, ratio of transmitted to untransmitted allele.
    *gene not previously implicated in T1D.
  • Turning to the DCCT/EDIC cohort, signals in the genes encoding ubiquitin-associated and SH3 domain-containing protein A (UBASH3A) and BTB and CNC homology 2 (BACH2) replicated in this fifth independent cohort (Table 2) and the P-values were significant after correcting for the six tests carried out. Clearly the risks are relatively modest compared to previously described associations, and it is only when we had this sample size at our disposal could we detect and establish these signals as true positives through an independent replication; however, Table 3 shows that rs9976767 is in fact significant at the genome-wide level when all five cohorts utilized were combined i.e. P=2.33×10−8.
  • TABLE 2
    Chr SNP Position Gene Aff allele freq Ctrl allele freq OR [95% CI] P
    21 rs9976767 42709459 UBASH3A 0.474 0.436 1.165 [1.051-1.292] 0.0036
    9 rs10758593 4282083 GLIS3 0.429 0.426 1.013 [0.913-1.124] 0.81
    9 rs10758594 4285583 GLIS3 0.434 0.443 0.963 [0.869-1.068] 0.48
    15 rs8035957 36625556 RASGRP1 0.270 0.261 1.047 [0.932-1.176] 0.44
    6 rs3757247 91014184 BACH2 0.497 0.463 1.144 [1.033-1.268] 0.010
    1 rs1983853 85083780 EDG7 0.132 0.153 0.842 [0.726-0.976] 0.022
    Replication results for the six SNPs of interest selected from the discovery process in the DCCT/EDIC T1D probands and CHOP controls.
    The two SNPs that successfully replicated are indicated in bold.
    Minor allele frequencies, P-values and odds ratios (OR) are shown, together with the gene in which the markers resides or which they are nearest to.
    P-values are two-sided in each instance.
    Aff allele freq, allele frequency in affected individuals;
    Chr, chromosome;
    CI, confidence interval;
    Ctrl allele freq, allele frequency in unaffected individuals.

    The co-ordinates for the linkage disequilibrium (LD) block that harbor the signals provided in Table 2 are set forth below. The present invention encompasses any SNP with these blocks that associated with an increased risk of T1D.
  • TABLE 3
    GENE CHR B36 Start B36 End
    UBASH3A 21 42689693 42725106
    GLIS3A 9 4267839 4290501
    RASGRP1 15 36601669 36728371
    BACH2 6 90944672 91078212
    EDG7 1 85002281 85127151

    See the world wide web at _//genome.ucsc.edu/cgi-bin/hgGateway for the details relating to build 36 of the human genome which was assembled in March of 2006.
  • TABLE 4
    Meta analysis of the five cohorts. Minor allele frequencies,
    P-values and odds ratios (OR) are shown, together with
    the relevant allele for each of the six SNPs.
    SNP Allele Gene OR [95% CI] P
    rs9976767 C UBASH3A 1.155 (1.098, 1.215) 2.33 × 10−8
    rs10758593 A GLIS3 1.131 (1.074, 1.190) 2.64 × 10−6
    rs10758594 A GLIS3 1.114 (1.058, 1.172) 3.51 × 10−5
    rs8035957 C RASGRP1 1.144 (1.080, 1.211) 3.92 × 10−6
    rs3757247 A BACH2 1.134 (1.078, 1.193) 1.25 × 10−6
    rs1983853 A EDG7 0.833 (0.773, 0.898) 1.87 × 10−6
  • UBASH3A is the only gene in this region of linkage disequilibrium. Mice lacking Sts2 (the mouse homologue for UBASH3A) have been shown to be normal in all respects, including T-cell function23. Mice lacking both Sts1 and Sts2 do have increased splenocyte numbers and are hyperresponsive to T-cell receptor stimulation. It has been suggested that STS1 and STS2 are critical regulators of the signaling pathways that regulate T-cell activation23.
  • BACH2 is also the only gene at this locus. The gene product is a member of the small Maf family which are basic region leucine zipper proteins that function either as transcriptional activators or repressors, depending on the proteins they heterodimerize with. Muto et al24 found that Bach2 −/− mice had relatively high levels of serum IgM but low levels of IgA and IgG subclasses. The Bach2 −/− mice have also been reported to present with deficient T cell-independent and T cell-dependent IgG responses, leading the authors to conclude that BACH2 was a regulator of the antibody response24. Using IPA software, we generated a BACH-2 related protein-protein interaction network of differentially expressed genes (DEG) for T1D. Grey represents genes that are up regulated in TID group versus controls. White represents gens that are down regulated in T1D. Solid lines show direct protein-protein interactions whereas dotted lines show indirect interactions. Different shapes represent different molecule types. See FIG. 2.
  • It should also be noted that rs1983853 yielded nominally significant association with T1D in all the cohorts but did not survive correction for multiple testing in the final replication attempt in the Toronto dataset. This SNP resides in endothelial differentiation gene 7 (EDG7; formerly LPA3), which has been implicated in mechanisms of embryo implantation25. Using IPA software, we generated an EDG7-LPAR3-related protein-protein interaction network of differentially expressed genes (DEG) for T1D. Grey represents genes that are up regulated in TID group versus controls. White represents gens that are down regulated in T1D. Solid lines show direct protein-protein interactions whereas dotted lines show indirect interactions. Different shapes represent different molecule types. See FIG. 3.
  • REFERENCES
  • 1. Todd, J. A., Bell, J. I. & McDevitt, H. O. HLA-DQ beta gene contributes to susceptibility and resistance to insulin-dependent diabetes mellitus. Nature 329, 599-604 (1987).
  • 2. Risch, N. Assessing the role of HLA-linked and unlinked determinants of disease. Am J Hum Genet 40, 1-14 (1987).
  • 3. Barratt, B. J. et al. Remapping the insulin gene/IDDM2 locus in type 1 diabetes. Diabetes 53, 1884-9 (2004).
  • 4. Pugliese, A. et al. The insulin gene is transcribed in the human thymus and transcription levels correlated with allelic variation at the INS VNTR-IDDM2 susceptibility locus for type 1 diabetes. Nat Genet 15, 293-7 (1997).
  • 5. Vafiadis, P. et al. Insulin expression in human thymus is modulated by INS VNTR alleles at the IDDM2 locus. Nat Genet 15, 289-92 (1997).
  • 6. Bottini, N., Vang, T., Cucca, F. & Mustelin, T. Role of PTPN22 in type 1 diabetes and other autoimmune diseases. Seminars in Immunology 18, 207-213 (2006).
  • 7. Vella, A. et al. Localization of a type 1 diabetes locus in the IL2RA/CD25 region by use of tag single-nucleotide polymorphisms. Am J Hum Genet 76, 773-9 (2005).
  • 8. Qu, H. Q., Montpetit, A., Ge, B., Hudson, T. J. & Polychronakos, C. Toward further mapping of the association between the IL2RA locus and type 1 diabetes. Diabetes 56, 1174-6 (2007).
  • 9. Lowe, C. E. et al. Large-scale genetic fine mapping and genotype-phenotype associations implicate polymorphism in the IL2RA region in type 1 diabetes. Nat Genet 39, 1074-1082 (2007).
  • 10. Ueda, H. et al. Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature 423, 506-11 (2003).
  • 11. Hakonarson, H. et al. A genome-wide association study identifies KIAA0350 as a type 1 diabetes gene. Nature 448, 591-4 (2007).
  • 12. Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661-678 (2007).
  • 13. Todd, J. A. et al. Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet 39, 857-864 (2007).
  • 14. Smyth, D. J. et al. A genome-wide association study of nonsynonymous SNPs identifies a type 1 diabetes locus in the interferon-induced helicase (IFIH1) region. Nat Genet 38, 617-619 (2006).
  • 15. Hakonarson, H. et al. A genome-wide association study identifies KIAA0350 as a type 1 diabetes gene. Nature 448, 591-594 (2007).
  • 16. Hakonarson, H. et al. A novel susceptibility locus for type 1 diabetes on Chr12q13 identified by a genome-wide association study. Diabetes 57, 1143-6 (2008).
  • 17. Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661-78 (2007).
  • 18. Todd, J. A. et al. Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet 39, 857-64 (2007).
  • 19. The Diabetes Control and Complications Trial Research Group. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med 329, 977-86 (1993).
  • 20. The Diabetes Control and Complications Trial (DCCT). The Diabetes Control and Complications Trial (DCCT). Design and methodologic considerations for the feasibility phase. The DCCT Research Group. Diabetes 35, 530-45 (1986).
  • 21. Price, A. L. et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet 38, 904-9 (2006).
  • 22. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 81, 559-75 (2007).
  • 23. Carpino, N. et al. Regulation of ZAP-70 activation and TCR signaling by two related proteins, Sts-1 and Sts-2. Immunity 20, 37-46 (2004).
  • 24. Muto, A. et al. Identification of Bach2 as a B-cell-specific partner for small maf proteins that negatively regulate the immunoglobulin heavy chain gene 3′ enhancer. Embo J 17, 5734-43 (1998).
  • 25. Ye, X. et al. LPA3-mediated lysophosphatidic acid signaling in embryo implantation and spacing. Nature 435, 104-8 (2005).
  • EXAMPLE II The RASGRP1 Locus and T1D
  • As described above, we had previously identified a SNP, rs8035957, in the RASGRP1 locus that was associated with T1D. The two genome-wide association studies published by us and by the Wellcome Trust Case-Control Consortium (WTCCC) revealed a number of novel loci.
  • In additional studies, we analyzed data from two sources: 1) The previously published second stage of our study, with a total sample size of the two stages consisting of 1,046 Canadian case-parent trios and 538 multiplex families with 929 affected offspring from the Type 1 Diabetes Genetics Consortium (T1DGC); 2) The RR2 project of the T1DGC, which genotyped 4,417 individuals from 1,062 non-overlapping families, including 2,059 affected individuals (mostly sibling pairs) for the 1,536 markers with the highest statistical significance for type 1 diabetes in the WTCCC results.
  • One locus, mapping to an LD block at chr15q14, reached statistical significance by combining results from two markers (rs17574546 and rs7171171) in perfect linkage disequilibrium (LD) with each other (r2=1). We obtained a joint p value of 1.3 ×10−6, which exceeds by an order of magnitude the conservative threshold of 3.26×10−5 obtained by correcting for the 1,536 SNPs tested in our study. Meta-analysis with the original WTCCC genome-wide data produced a p value of 5.83×10−9.
  • These studies confirm that the results presented in Example 1 identifying a novel type 1 diabetes locus involving the RASGRP1 gene. This gene is known to play a crucial role in thymocyte differentiation and TCR signaling by activating the Ras signaling pathway.
  • The following materials and methods are similar to those described in Example I and are provided to facilitate the practice of Example 2.
  • 1. The T1DGC RR2 study genotyped 4,417 individuals from 1,062 type 1 diabetes families, including 2,059 affected siblings and both their parents for the 1,536 markers with the highest statistical significance for type 1 diabetes in the WTCCC results. Genotyping was performed at the Sanger Institute on the Illumina Golden Gate platform. Most subjects were of European ancestry, with a median age at onset of 10 years (lower and upper quartiles at 6 years and 15.5 years).
  • 2. In our study, we genotyped 1,046 type 1 diabetes type 1 diabetes case-parent trios, collected in pediatric diabetes clinics in Montreal, Toronto, Ottawa and Winnipeg. The median age at onset is 8.4 years with lower and upper quartiles at 5.0 years and 11.8 years. Ethnic backgrounds were of mixed European descent, with the largest single subset (40%) being French Canadian. The Research Ethics Board of the Montreal Children's Hospital and other participating centers approved the study, and written informed consent was obtained from all subjects. In addition, we genotyped 549 families with at least one child with type 1 diabetes and both parents (946 total affected). The median age at onset is 8 with quartiles at 4 years and 13 years. The samples were collected in Europe, North America and Australia and most subjects were of European ancestry. Genotyping data from 11 overlapping families that were also included in the RR2 study were removed for analysis. As we previously described4, we used the Illumina Golden Gate array to genotype 982 markers with p<0.05 in both the TDT and case-control phase of our original GWAS. In addition, 15 single-nucleotide polymorphisms (SNP) with p<0.1 in each of our two GWA cohorts and p<0.01 in WTCCC were genotyped using mass spectrometry on the Sequenom iPlex platform.
  • 3. Statistics
  • Type 1 diabetes association was tested by the Family Based Association Test (FBAT) software available on the world wide web at biostat.harvard.edu/˜fbat/fbat.htm8. Considering most of the T1DGC families have multiple siblings, the option of the empirical variance was used in the FBAT statistics to permit a robust but unbiased test of genetic association. As 1,536 SNPs were tested in the RR2 study, we used a conserved significance threshold corrected for multiple comparisons at 3.26×10−5.
  • Results
  • Recently, two independent studies validated the type 1 diabetes association of UBASH3A and BACH22, 3. Further research confirms that the RASGRP1 locus is also an important type 1 diabetes locus. Overlap in the markers selected in the two projects was determined either by identity of SNPs or, in cases of physical proximity (<1 Mb), by LD (r2 value>0.8). After excluding known type 1 diabetes loci, there was only one locus nominally significant (P<0.05) in both projects. It involves a locus evaluated in the RR2 cohort by SNP rs17574546 (P =3.41×10−3) and in our set by rs7171171 (P=8.40×10−5, Table 5).
  • TABLE 5
    Association analysis between the RASGRP1
    variations and type 1 diabetes
    Hardy- Informative
    Minor allele Weinberg family
    Cohort (Frequency) p number* Z (P value)
    The T1DGC RR2 cohort
    rs17574546 C (0.225) 0.931 302 2.93 (P =
    3.41 × 10−3)
    The Canadian cohort and extra T1DGC samples
    rs7171171 G (0.207) 0.873 665 3.93 (P =
    8.40 × 10−5)
    Combined — (0.209)  1.000 967 4.84 (P =
    analysis 1.30 × 10−6)
    *Number of families informative for (with a non-zero contribution to) the FBAT analysis;
    No redundant sample with the T1DGC RR2 cohort.
  • The genotype calling rate of rs17574546 in the RR2 samples is 99.8%, and for rs7171171 in our own samples is 99.9%. No Mendelian error was found in either. As these SNPs are in perfect LD (r2=1) we performed a direct combined analysis which showed P=1.30×10−6. This exceeds by more than an order of magnitude the corrected significance level. The OR (95% CI) estimated on the combined family dataset is 1.22 (1.12, 1.33), while the OR (95% CI) in the WTCCC case-control set is 1.21 (1.09, 1.33) (P=2.67×10−4). The meta-analysis of these two results gives an OR (95% CI)=1.21 (1.14, 1.30) and P=5.83×10−9, a significance level accepted for genome-wide studies. Based on these results, we can conclude that the RASGRP1 locus is associated with type 1 diabetes. It is interesting to note that rs17574546 and rs7171171 both have D′=0.902, and r2=0.553, with rs8035957 described in Example 1.
  • This novel type 1 diabetes association signal maps to a LD block at Chr15q14, ˜13 kb upstream of the transcription start site of the RASGRP1 gene, and has no LD with any known type 1 diabetes locus. See FIG. 1. As type 1 diabetes is caused by the autoimmune destruction of pancreatic β-cells, it is interesting that the RASGRP1 gene has an important immune function. RASGRP1 (NCBI GeneID: 10125) encodes calcium and DAG-regulated RAS guanyl releasing protein 1 (RasGRP1)9. RasGRP1 plays crucial roles in thymocyte differentiation and TCR signaling by activating the Ras signaling pathway. RasGRP1-null mutant mice have approximately normal numbers of immature thymocytes but a marked deficiency of mature, single-positive (CD4+CD8 and CD4 CD8+) thymocytes10. Transgenic expression of RasGRP1 induces the maturation of double-negative thymocytes and enhances the production of CD4CD8+ thymocytes11. In addition, RasGRP1 has dramatic effect on the development and function of CD4+CD25+ regulatory T-cells (Treg). In the absence of RasGRP1, the development of CD4+CD25+ Treg in the thymus is severely impaired, whereas the peripheral expansion and function of CD4+CD25+ Treg are greatly increased12. CD4+CD25+ Treg plays a critical role in maintaining immune homeostasis and inhibiting autoimmune reaction of type 1 diabetes and other autoimmune diseases13. As the transfer of CD4+CD25+ CD4+CD25+ Treg cells can prevent type 1 diabetes in the recipient NOD mice14 , knowledge of the role of genes involved in the generation of this subset in type 1 diabetes may play an important role in the development of preventive interventions.
  • REFERENCES
  • 1. Hakonarson H, Grant S F, Bradfield J P, Marchand L, Kim C E, Glessner J T, Grabs R, Casalunovo T, Taback S P, Frackelton E C, Lawson M L, Robinson L J, Skraban R, Lu Y, Chiavacci R M, Stanley C A, Kirsch S E, Rappaport E F, Orange J S, Monos D S, Devoto M, Qu H Q, Polychronakos C. A genome-wide association study identifies KIAA0350 as a type 1 diabetes gene. Nature 2007; 448:591-4.
  • 2. Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 2007; 447:661-678.
  • 3. Todd J A, Walker N M, Cooper J D, Smyth D J, Downes K, Plagnol V, Bailey R, Nejentsev S, Field S F, Payne F, Lowe C E, Szeszko J S, Hafler J P, Zeitels L, Yang J H M, Vella A, Nutland S, Stevens H E, Schuilenburg H, Coleman G, Maisuria M, Meadows W, Smink L J, Healy B, Burren O S, Lam A A C, Ovington N R, Allen J, Adlem E, Leung H-T, Wallace C, Howson J M M, Guja C, Ionescu-Tirgoviste C, Simmonds M J, Heward J M, Gough S C L, Dunger D B, Wicker L S, Clayton D G. Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet 2007; 39:857-864.
  • 4. Grant S F, Qu H Q, Bradfield J P, Marchand L, Kim C E, Glessner J T, Grabs R, Taback S P, Frackelton E C, Eckert A W, Annaiah K, Lawson M L, Otieno F G, Santa E, Shaner J L, Smith R M, Skraban R, Imielinski M, Chiavacci R M, Grundmeier R W, Stanley C A, Kirsch S E, Waggott D, Paterson A D, Monos D S, Polychronakos C, Hakonarson H. Follow-up analysis of genome-wide association data identifies novel loci for type 1 diabetes. Diabetes 2009; 58:290-5.
  • 5. Cooper J D, Smyth D J, Smiles A M, Plagnol V, Walker N M, Allen J E, Downes K, Barrett J C, Healy B C, Mychaleckyj J C, Warram J H, Todd J A. Meta-analysis of genome-wide association study data identifies additional type 1 diabetes risk loci. Nat Genet 2008; 40:1399-401.
  • 6. Concannon P, Onengut-Gumuscu S, Todd J A, Smyth D J, Pociot F, Bergholdt R, Akolkar B, Erlich H A, Hilner J E, Julier C, Morahan G, Nerup J, Nierras C R, Chen W M, Rich S S. A human type 1 diabetes susceptibility locus maps to chromosome 21q22.3. Diabetes 2008; 57:2858-61.
  • 7. Rich S S, Concannon P, Erlich H, Julier C, Morahan G, Nerup J, Pociot F, Todd J A. The Type 1 Diabetes Genetics Consortium. Ann NY Acad Sci 2006; 1079:1-8.
  • 8. Horvath S, Xu X, Laird N M. The family based association test method: strategies for studying general genotype-phenotype associations. Eur J Hum Genet 2001; 9:301-6.
  • 9. Ebinu J O, Bottorff D A, Chan E Y, Stang S L, Dunn R J, Stone J C. RasGRP, a Ras guanyl nucleotide-releasing protein with calcium- and diacylglycerol-binding motifs. Science 1998; 280:1082-6.
  • 10. Dower N A, Stang S L, Bottorff D A, Ebinu J O, Dickie P, Ostergaard H L, Stone J C. RasGRP is essential for mouse thymocyte differentiation and TCR signaling. Nature Immunology 2000; 1:317-321.
  • 11. Norment A M, Bogatzki L Y, Klinger M, Ojala E W, Bevan M J, Kay R J. Transgenic expression of RasGRP 1 induces the maturation of double-negative thymocytes and enhances the production of CD8 single-positive thymocytes. J Immunol 2003; 170:1141-9.
  • 12. Chen X, Priatel J J, Chow M T, Teh H-S. Preferential Development of CD4 and CD8 T Regulatory Cells in RasGRP1-Deficient Mice. J Immunol 2008; 180:5973-5982.
  • 13. Shevach E M. Certified professionals: CD4(+)CD25(+) suppressor T cells. J Exp Med 2001; 193:F41-6.
  • 14. Salomon B, Lenschow D J, Rhee L, Ashourian N, Singh B, Sharpe A, Bluestone J A. B7/CD28 Costimulation Is Essential for the Homeostasis of the CD4+CD25+ Immunoregulatory T Cells that Control Autoimmune Diabetes. Immunity 2000; 12:431.
  • EXAMPLE III Diagnostic Methods for T1D and Screening Assays to Identify Theraeutic Agents Useful for the Treatment of T1D
  • The information herein above can be applied clinically to patients for diagnosing an increased susceptibility for developing T1D, and therapeutic intervention. A preferred embodiment of the invention comprises clinical application of the information described herein to a patient. Diagnostic compositions, including microarrays, and methods can be designed to identify the genetic alterations described herein in nucleic acids from a patient to assess susceptibility for developing T1D. This can occur after a patient arrives in the clinic; the patient has blood drawn, and using the diagnostic methods described herein, a clinician can detect a SNP in the regions of chromosome 21, 15, 6, 9 and 1 described herein. The typical age range for a patient to be screened is between 9 and 12 years of age. The information obtained from the patient sample, which can optionally be amplified prior to assessment, will be used to diagnose a patient with an increased or decreased susceptibility for developing T1D. Kits for performing the diagnostic method of the invention are also provided herein. Such kits comprise a microarray comprising at least one of the SNPs provided herein in and the necessary reagents for assessing the patient samples as described above.
  • The identity of T1D-involved genes and the patient results will indicate which variants are present, and will identify those that possess an altered risk for developing T1D. The information provided herein allows for therapeutic intervention at earlier times in disease progression that previously possible. Also as described herein above, UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7 provide novel targets for the development of new therapeutic agents efficacious for the treatment of T1D. In particular, it would be desirable to block expression of these genes in those patients that are more prone to develop the disease. In this regard, the therapeutic siRNAs described herein can be used to block expression of the gene product based on the patient signal, thereby inhibiting the pancreatic 3-cell destruction that occurs in T1D.
  • Candidate siRNA compositions for use in the invention are provided in Tables 6-10. The sequences in Tables 6-10 include several siRNAs (i.e., sense sequences for a target region). Those of skill in the art can determine the sequence of an antisense siRNA strand based on the disclosure of the sense strand, and will appreciate the difference between any “U” and “T” designations in the sequences which correspond to RNA and DNA molecules, respectively. Also, methods of using known inhibitors of UBASH3A, GLIS3, RASGRP1, BACH2 and EDG7 to treat T1D are also provided. See the tables below. In addition, shRNA constructs can be designed based on the sense sequence provided in Tables 4-8, and may be effective to inhibit UBASH3A, GLIS3, RASGRP1, BACH2 and EDG7. The shRNA constructs utilizing the sense strand from Tables 6-10 for the respective targets would include a hairpin loop 3′ to the sense sequence (e.g., suitable hairpins include, but are not limited to: TCAAGAG, TTCAAGAGA, GAAGCTTG, and TTCG) followed by the corresponding antisense sequence from the sense strand provided in Tables 6-10. Table 10B provides known small molecule antagonists of EDG7 which should have utility for the treatment of T1D.
  • TABLE 6
    Candidate UBASH3 siRNA molecules (sense)
    GCATTTAACTGGAGGAACTtt SEQ ID NO: 6
    CAAGAGTTCTGGAGAGAGAtt SEQ ID NO: 7
    GAACAGAGCTCATGAGGTCtt SEQ ID NO: 8
    AATCAAGATACGAGTGGAAtt SEQ ID NO: 9
    GGATCGAGCCAGTGAGTCTtt SEQ ID NO: 10
    CGGCGAGCATGGTGCAAATtt SEQ ID NO: 11
    GGAACTGGATCTCAGGCAAtt SEQ ID NO: 12
    GTGGATGAGCTGACGCTAAtt SEQ ID NO: 13
    GAAAATGGGAGTTGGTGAAtt SEQ ID NO: 14
    ACGCCAAGGTCTCCAACAAtt SEQ ID NO: 15
    GGACATGGCCCTAACCTGAtt SEQ ID NO: 16
    GGGAGAGAGTGGATCAGATtt SEQ ID NO: 17
    CCAAACTCATCCTGGAAGAtt SEQ ID NO: 18
    GAGTCTGACACGTGGGTGAtt SEQ ID NO: 19
    GGAGAGAGAGCAAGCGCCAtt SEQ ID NO: 20
    GAAGAGAGCTGGAGACAGGtt SEQ ID NO: 21
    GGGAATTCGCCATGACCTTtt SEQ ID NO: 22
    GGCCCTAACCTGAGGCTGAtt SEQ ID NO: 23
    CGTGAAGCCTTGCACCAAAtt SEQ ID NO: 24
    GGAAAATGGGAGTTGGTGAtt SEQ ID NO: 25
    GGGCGAACGCAGCATTTAAtt SEQ ID NO: 26
    AGTTCTGGAGAGAGAGCAAtt SEQ ID NO: 27
    GTGAAGACCAGAAGGTGGAtt SEQ ID NO: 28
    AGGCTGAGCAATTTAACTAtt SEQ ID NO: 29
    CAGCAGATGCAGCGGGGAAtt SEQ ID NO: 30
    GGACAGTGGTATCAGAATCtt SEQ ID NO: 31
    AGACGCAGCTCTACGCCAAtt SEQ ID NO: 32
    AGGCATGGCTGCAGCAATGtt SEQ ID NO: 33
    TGGAAGAACTCAAACTGGAtt SEQ ID NO: 34
    CTGAAGAGAGCTGGAGACAtt SEQ ID NO: 35
    AACCTGAGGCTGAGCAATTtt SEQ ID NO: 36
    GCACCAAACAGCTGCATCTtt SEQ ID NO: 37
    GCACTCTACTCCCGAGACAtt SEQ ID NO: 38
    GGGTGAAGCACAGGATGTAtt SEQ ID NO: 39
    CCACAAACGGCAAGGAGTCtt SEQ ID NO: 40
    CAAACGGCAAGGAGTCTTAtt SEQ ID NO: 41
    GGTGAAGCCAGCAGCAGATtt SEQ ID NO: 42
    CAAAATGGGAAGCTGGCAAtt SEQ ID NO: 43
    GCCTGGAAGAGCTGAAAGAtt SEQ ID NO: 44
    AAGAGCTGAAAGAGGCAAAtt SEQ ID NO: 45
    CGGTGAAGACCCTGACCCAtt SEQ ID NO: 46
    GAGCCCTATTCCAGTACAAtt SEQ ID NO: 47
    GCAAGGAGTCTTAGCAGCTtt SEQ ID NO: 48
    CCATTATCATCGTGTGGCAtt SEQ ID NO: 49
    GGAAGAGCTGAAAGAGGCAtt SEQ ID NO: 50
    CAACATTGACACTGATTACtt SEQ ID NO: 51
    GAAAATAAAGAGGAAGGAAtt SEQ ID NO: 52
    CTTCAAGAGTTCTGGAGAGtt SEQ ID NO: 53
    CCGGAAAACTACACGGATCtt SEQ ID NO: 54
    GTGAAGCACAGGATGTACAtt SEQ ID NO: 55
  • TABLE 7
    Candidate GLIS3 siRNA molecules (sense)
    GCACAGAGCTCCATCCAGAtt SEQ ID NO: 56
    GCTATAAACTGCTGATCCAtt SEQ ID NO: 57
    TCACATACTTTAAAGCCAAtt SEQ ID NO: 58
    GGGCAGCACCGTAGACCTAtt SEQ ID NO: 59
    GGTCAGTGGTCATCACATTtt SEQ ID NO: 60
    ACGCAGGAGCTGAGAGGTTtt SEQ ID NO: 61
    CCTATCAGCCAGAAACAAAtt SEQ ID NO: 62
    TCAGAATGGCCTTGATCTAtt SEQ ID NO: 63
    GGAAAAGGCAGCTGCAACAtt SEQ ID NO: 64
    GGGCAATGAATGCAGCCAAtt SEQ ID NO: 65
    AGGAGTGGTCCCAGGGCTAtt SEQ ID NO: 66
    CCGAACGCCTGGAGGAGTTtt SEQ ID NO: 67
    GAGCAACAAGCAAGGAAAAtt SEQ ID NO: 68
    GGAGACAAATGCTCACCAAtt SEQ ID NO: 69
    CCAGATCAGTCCTAGCTTAtt SEQ ID NO: 70
    GAATATACCTCCTTCAGATtt SEQ ID NO: 71
    GTTTGAAGGTTGCGAGAAGtt SEQ ID NO: 72
    GGACGCATCTGGACACCAAtt SEQ ID NO: 73
    AGAGCAACAAGCAAGGAAAtt SEQ ID NO: 74
    AGCCAAAGCAGCAGGAGTTtt SEQ ID NO: 75
    GCTTTGGGCCTCAGTGCAAtt SEQ ID NO: 76
    TATTCAAGCCGAAGTGGAAtt SEQ ID NO: 77
    CTTCAATACTGCAAAGAACtt SEQ ID NO: 78
    CTAACAACCTCCATCTCAAtt SEQ ID NO: 79
    GCAACAATCTAGTGGTCACtt SEQ ID NO: 80
    CCTCAAGCATGAAGCAGGAtt SEQ ID NO: 81
    GGATGGCTCCTCAGAACAAtt SEQ ID NO: 82
    ACCTTGAGTCTGACGGAAAtt SEQ ID NO: 83
    GTACCAAACGCTACACAGAtt SEQ ID NO: 84
    CTGTCTACACCGAAGGCTAtt SEQ ID NO: 85
    TGTCTACACCGAAGGCTAAtt SEQ ID NO: 86
    GCATGAAGCAGGAGTGGTCtt SEQ ID NO: 87
    CCAAAGAGCAACAAGCAAGtt SEQ ID NO: 88
    TAGAGATGCTGCTGCTGAAtt SEQ ID NO: 89
    AGCAATTATTCAAGCCGAAtt SEQ ID NO: 90
    TCAGATACCAGGTCCCTTAtt SEQ ID NO: 91
    CCTAGCTTACAGAGGGCAAtt SEQ ID NO: 92
    TTGTCAAATTCCAGGATGTtt SEQ ID NO: 93
    ACCCAAGTTCCCTAAGAAAtt SEQ ID NO: 94
    CCTAAGAAAGCATGTGAAGtt SEQ ID NO: 95
    TCCTCCAAATCCTGGGAAAtt SEQ ID NO: 96
    CCTTATTTCGCGTGAGTCTtt SEQ ID NO: 97
    CAGTCGGCCTCAAGCATGAtt SEQ ID NO: 98
    ACACAGGCGAGAAGCCGTAtt SEQ ID NO: 99
    CACCAAACCTTATGCTTGTtt SEQ ID NO: 100
    ACCTTATGCTTGTCAAATTtt SEQ ID NO: 101
    CAGCAATTATTCAAGCCGAtt SEQ ID NO: 102
    CCACAGAGCCTTCTCGACTtt SEQ ID NO: 103
    CCAATGGGAAGCCGCGATTtt SEQ ID NO: 104
    GGAAAGGGGCTCTTGGCTTtt SEQ ID NO: 105
  • TABLE 8
    Candidate RASGRP1 siRNA molecules (sense)
    AAGCAAGACTAGAGGCAAAtt SEQ ID NO: 106
    GAAACTTACTCAAAGGATAtt SEQ ID NO: 107
    CCAGAAACTACGACAATTAtt SEQ ID NO: 108
    TGAAATATGCACAGAAGAAtt SEQ ID NO: 109
    CCACAGAGCTCCACCACTAtt SEQ ID NO: 110
    GGAAAGTGAACGTCCATAAtt SEQ ID NO: 111
    GCAAACACGTCCAGAGGATtt SEQ ID NO: 112
    GGATGAAATCTATGAGCTTtt SEQ ID NO: 113
    CCTAAAGATCCAACTGAAAtt SEQ ID NO: 114
    ACAAGGATATCGATGTAAAtt SEQ ID NO: 115
    GGATATCGTTCTCTGATTAtt SEQ ID NO: 116
    AAACAAGGATATCGATGTAtt SEQ ID NO: 117
    TGGTTGTGTTTGAGTGTAAtt SEQ ID NO: 118
    TGGTGAAAGCTAAGGGTGAtt SEQ ID NO: 119
    GCAAAGATCTGGTTGTGTTtt SEQ ID NO: 120
    TTGTCAAGTGGGAGAATAAtt SEQ ID NO: 121
    GCACAGAAGAAAATAGAATtt SEQ ID NO: 122
    TCAATAAGGTTCTCGGTGAtt SEQ ID NO: 123
    CGACCAGGATGGATACATTtt SEQ ID NO: 124
    CGGGATGAACTGTCACAAAtt SEQ ID NO: 125
    GCCCAGTCTTGGTCAGAAAtt SEQ ID NO: 126
    AGGAACTGGTGAAAGCTAAtt SEQ ID NO: 127
    GCTCCATGCACCTGAGGAAtt SEQ ID NO: 128
    GAATAAAGACTCCCTCATAtt SEQ ID NO: 129
    AGGTATTGGATAACAGAATtt SEQ ID NO: 130
    AAGCTAAGGGTGAGGAGTTtt SEQ ID NO: 131
    ACACTGAGGATGAAATCTAtt SEQ ID NO: 132
    TGACAACTGTGCTGGATTTtt SEQ ID NO: 133
    GGATATCGATGTAAAGACTtt SEQ ID NO: 134
    AATAAAGACTCCCTCATAAtt SEQ ID NO: 135
    GATGGAAACCTGTGTCGAAtt SEQ ID NO: 136
    GAGAGAGGCTCCGCGGAAAtt SEQ ID NO: 137
    GGGTACAACTGATGGTTCTtt SEQ ID NO: 138
    GGGATGAGATCACAGCCTAtt SEQ ID NO: 139
    GTAAGAAGCGAGCCAAGAAtt SEQ ID NO: 140
    ATAAAGACTCCCTCATAAAtt SEQ ID NO: 141
    GAAATATGCACAGAAGAAAtt SEQ ID NO: 142
    GGAAACCTGTGTCGAAGTAtt SEQ ID NO: 143
    GGTATTGGATAACAGAATTtt SEQ ID NO: 144
    AGCTAAGGGTGAGGAGTTAtt SEQ ID NO: 145
    TGACACAACTCAAATCAATtt SEQ ID NO: 146
    CAGAAGAGCTATCCGAGCAtt SEQ ID NO: 147
    CCTTCTGTGTGATGGACAAtt SEQ ID NO: 148
    CCTCACAACTTCCAAGAGAtt SEQ ID NO: 149
    GAGTGATCAAACAAGGATAtt SEQ ID NO: 150
    GATCAAACAAGGATATCGAtt SEQ ID NO: 151
    AGGAAGACAGCCCAGGATAtt SEQ ID NO: 152
    CCAAGAACTGGAACAGGAAtt SEQ ID NO: 153
    AAGAACTGGAACAGGAAATtt SEQ ID NO: 154
    CCCTAAAGATCCAACTGAAtt SEQ ID NO: 155
  • TABLE 9
    Candidate BACH2 siRNA molecules (sense)
    GAAGATAACTCTAGCAACAtt SEQ ID NO: 156
    GTGAAGAGAATGAGGAAGAtt SEQ ID NO: 157
    CAAATTGGTGTGTGAGAAAtt SEQ ID NO: 158
    CAGGAGAGGAGGAGGATGAtt SEQ ID NO: 159
    AGAATGAGGAAGAGAGCATtt SEQ ID NO: 160
    CAGAACAGTTAGAGTTTATtt SEQ ID NO: 161
    GGAAATGACTGATAAGTGTtt SEQ ID NO: 162
    CTTTGATCGTGGAGAGGAAtt SEQ ID NO: 163
    GAGAGGAGGAGGATGAAGAtt SEQ ID NO: 164
    GTACCAAGAATGTCTATAAtt SEQ ID NO: 165
    GAGATGAGCCTGACGCCAAtt SEQ ID NO: 167
    GAGAAACTGTTGTCAGAGAtt SEQ ID NO: 168
    AGAGAGGAATCAACTGAAAtt SEQ ID NO: 169
    CAGTGAAGAGAATGAGGAAtt SEQ ID NO: 170
    GGTGTGTGAGAAAGAGAAAtt SEQ ID NO: 171
    GAGGAGGAGACGATGGATTtt SEQ ID NO: 172
    GGGAAGATAACTCTAGCAAtt SEQ ID NO: 173
    AAAGGAAACTGGACTGTATtt SEQ ID NO: 174
    GCAAATTGGTGTGTGAGAAtt SEQ ID NO: 175
    AAGAGAAACTGTTGTCAGAtt SEQ ID NO: 176
    ATGAAGAGGAGGAGACGATtt SEQ ID NO: 177
    GGTTGGAGGCTCTCTGTAAtt SEQ ID NO: 178
    CAGCAACACCTCCGAGAATtt SEQ ID NO: 179
    AAACAGTGACCGTGGACTTtt SEQ ID NO: 180
    GAACAGCCCAGGAAAGATTtt SEQ ID NO: 181
    CCTCAGAACAGTTAGAGTTtt SEQ ID NO: 182
    GTGCTGAGTTCCTGCGCATtt SEQ ID NO: 183
    GCGAGAACTCTGCAGGAGAtt SEQ ID NO: 184
    CTGCAGGAGAGGAGGAGGAtt SEQ ID NO: 185
    CCGTAGCAGAGAAGGAAGAtt SEQ ID NO: 186
    GCCAGAAGGAGGTGTCCAAtt SEQ ID NO: 187
    GGGTGAGCAGTTTGGACAAtt SEQ ID NO: 188
    CCAGGAAAGATTATACCTAtt SEQ ID NO: 189
    CCTCAATGACCAGCGGAAAtt SEQ ID NO: 190
    CTGTTACTCAGCAGAGAAAtt SEQ ID NO: 191
    GCCAGGAAATGACTGATAAtt SEQ ID NO: 192
    AGAAGGAGGTGTCCAACTTtt SEQ ID NO: 193
    CCAAATTAAATGTGAGCAGtt SEQ ID NO: 194
    AGGAATCAACTGAAAGCATtt SEQ ID NO: 195
    CAGGAAGTTTGCCGAGACAtt SEQ ID NO: 196
    AGGAGGAGGATGAAGAGGAtt SEQ ID NO: 197
    AGGAGGATGAAGAGGAGGAtt SEQ ID NO: 198
    ACCAAGGAGAGCTCAGAAAtt SEQ ID NO: 199
    TCACAGGGAATTATGGACAtt SEQ ID NO: 200
    GTTGGAGGCTCTCTGTAAAtt SEQ ID NO: 201
    GAGACCAGGACCAGGACTTtt SEQ ID NO: 202
    CCACAGAACATCAGGAACCtt SEQ ID NO: 203
    TTAAATGTGAGCAGTCTTAtt SEQ ID NO: 204
    TCTCGGAAGCAGACAGTGAtt SEQ ID NO: 205
    CTTGAACCCAGGAGCCAAAtt SEQ ID NO: 206
  • TABLE 10A
    Candidate EDG7 siRNA molecules (sense)
    CCTACAAGGACGAGGACATtt SEQ ID NO: 207
    TCTACTACCTGTTGGCTAAtt SEQ ID NO: 208
    TCATCATGGTTGTGGTGTAtt SEQ ID NO: 209
    GTACATAGAGGATAGTATTtt SEQ ID NO: 210
    GTCGATGACTGGACAGGAAtt SEQ ID NO: 211
    GTGGAGAGGCACATGTCAAtt SEQ ID NO: 212
    GGATGCGGGTCCATAGCAAtt SEQ ID NO: 213
    CCATGAAGCTAATGAAGACtt SEQ ID NO: 214
    AATAGGAGCAACACTGATAtt SEQ ID NO: 215
    AGTACATAGAGGATAGTATtt SEQ ID NO: 216
    CAATAAAAGCACTTCCTAAtt SEQ ID NO: 217
    ATGACAAGCACATGGACTTtt SEQ ID NO: 218
    GTGTACGTCAAGAGGAAAAtt SEQ ID NO: 219
    AAGCTAATGAAGACGGTGAtt SEQ ID NO: 220
    GCACCATGAAGAAGATGATtt SEQ ID NO: 221
    AGAGGATAGTATTAGCCAAtt SEQ ID NO: 222
    CCGATTTCTTCGCTGGAATtt SEQ ID NO: 223
    ACACAGGCCCAGTTTCAAAtt SEQ ID NO: 224
    CCATTTACAGCAGGAGTTAtt SEQ ID NO: 225
    GGACACCCATGAAGCTAATtt SEQ ID NO: 226
    ACAAGGACGAGGACATGTAtt SEQ ID NO: 227
    TGTCCAACCTCATGGCCTTtt SEQ ID NO: 228
    CCTCAGCAGGAGTGACACAtt SEQ ID NO: 229
    GCCAGTACATAGAGGATAGtt SEQ ID NO: 230
    TAATTTAGCTGCTGCCGATtt SEQ ID NO: 231
    CCTATGTATTCCTGATGTTtt SEQ ID NO: 232
    TAACACAGGCCCAGTTTCAtt SEQ ID NO: 233
    CCCATGAAGCTAATGAAGAtt SEQ ID NO: 234
    CAGCCAGTACATAGAGGATtt SEQ ID NO: 235
    GAGGATAGTATTAGCCAAGtt SEQ ID NO: 236
    CTGGAATTGCCTATGTATTtt SEQ ID NO: 237
    TGGAGAGGCACATGTCAATtt SEQ ID NO: 238
    CATAGCAACCTGACCAAAAtt SEQ ID NO: 239
    AGGACACCCATGAAGCTAAtt SEQ ID NO: 240
    TGGCGTGCAGCATGTGAAAtt SEQ ID NO: 241
    ACACTGATACTGTCGATGAtt SEQ ID NO: 242
    GGATAGTATTAGCCAAGGTtt SEQ ID NO: 243
    CGATTTCTTCGCTGGAATTtt SEQ ID NO: 244
    CGATGACTGGACAGGAACAtt SEQ ID NO: 245
    CATACAAGTGGGTCCATCAtt SEQ ID NO: 246
    TAGTATTAGCCAAGGTGCAtt SEQ ID NO: 247
    TCATCGCGGCAGTGATCAAtt SEQ ID NO: 248
    TGAAGACGGTGATGACTGTtt SEQ ID NO: 249
    CTGGACAGGAACAAAGCTTtt SEQ ID NO: 250
    GGTCATCGCGGCAGTGATCtt SEQ ID NO: 251
    GGAGCAACACTGATACTGTtt SEQ ID NO: 252
    CTTCTGGACAGTAGCTTGAtt SEQ ID NO: 253
    GAGAGGCACATGTCAATCAtt SEQ ID NO: 254
    TGACAAGCACATGGACTTTtt SEQ ID NO: 255
    CCCATTTACAGCAGGAGTTtt SEQ ID NO: 256
  • TABLE 10B
    Antagonists for EDG7
    Active
    Ingre-
    Company Product dient Phase Description Indication MOA
    Amira AM966 PC AM966 is a LPA1 (lysophosphatidic acid receptor 1) Pulmonary Lysophosphatidic Acid
    Pharmaceuticals inhibitor. Lysophosphatidic acid receptor 1 (LPAR1; Fibrosis Type 1 Receptor (LPA1)
    (Private) EDG2; LPA1) is an EDG receptor that is part of the (Idiopathic Antagonist
    phosphatidic acid/lysophosphatidylcholine Pulmonary
    pathway. Lysophosphatidic acid (LPA) is an anionic Fibrosis)
    bioactive lipid that acts through EDG receptors to
    mediate cell proliferation, platelet aggregation,
    smooth muscle contraction, inhibition of
    neuroblastoma cell differentiation, chemotaxis and
    tumor cell invasion. LPA also elicits angiogenesis.
    AM966 is being developed for the treatment of
    idiopathic pulmonary fibrosis (IPF).
    Amira LPA1 PC LPA1 Receptor Antagonist acts by blocking the Cancer Lysophosphatidic Acid
    Pharmaceuticals Receptor LPA-induced migration of cancer cells and is being Type 1 Receptor (LPA1)
    (Private) Antagonist developed for the treatment of cancer. Antagonist
    Amira LPA1 PC LPA1 Receptor Antagonist is an anti-fibrotic and Kidney Disease Lysophosphatidic Acid
    Pharmaceuticals Receptor anti-inflammatory agent which is being developed (Kidney Fibrosis) Type 1 Receptor (LPA1)
    (Private) Antagonist for the treatment of kidney fibrosis. Antagonist
    Catena VPC51299 PC VPC51299 is a potent lysophosphatidic acid (LPA1) Solid Tumors Lysophosphatidic Acid
    Pharmaceuticals, receptor antagonist which targets LPA G-protein Type 1 Receptor (LPA1)
    Inc. (Private) coupled receptors (GPCRs) with high potency. Antagonist
    These GPCRs elicit a signaling cascade upon LPA
    binding the receptor leading to LPA's biological
    effects, such as proliferation, migration,
    angiogenesis. VPC51299 antagonizes (blocks) this
    signaling cascade. VPC51299 is being developed
    as oral formulation for the treatment of solid
    tumors.
    Debiopharm Debio0719 PC Debio 0719 is a highly potent, selective, small Bone Metastases Lysophosphatidic Acid
    Group (Private) molecule inhibitor of type 1 lysophosphatidic acid (Bone Damage Type 1 Receptor (LPA1)
    receptor (LPA1) and type 3 (LPA3). Debio 0719 is Induced by Antagonist;
    being developed for the treatment of osteolytic Metastatic Lysophosphatidic
    bone damage induced by breast tumour Breast Cancer) Acid Type 3 Receptor
    metastasis. (LPA3) Antagonist
    Kyowa Hakko Debio0719 PC Debio0719 is a highly potent, selective, small Bone Metastases Lysophosphatidic Acid
    Kirin Co., Ltd. molecule inhibitor of type 1 lysophosphatidic acid (Bone Damage Type 1 Receptor (LPA1)
    (formerly Kyowa receptor (LPA1) and type 3 (LPA3). Debio0719 is Induced by Antagonist;
    Hakko Kogyo being developed for the treatment of bone damage Metastatic Lysophosphatidic
    Co., Ltd.) (Stock induced by metastatic breast cancer. Note: This Breast Cancer) Acid Type 3 Receptor
    Code Number: product is added upon merger of Kyowa Hakko (LPA3) Antagonist
    4151 (TYO)) Kogyo Co., Ltd. with Kirin Pharma Company
    Limited
    Amira AM152 1 AM152 is a lysophosphatidic acid (LPA1) Pulmonary Lysophosphatidic Acid
    Pharmaceuticals antagonist that acts by blocking phosphatidic Fibrosis Type 1 Receptor (LPA1)
    (Private) acid/lysophosphatidylcholine pathway involved in (Idiopathic Antagonist
    fibrotic disease and is being developed for the Pulmonary
    treatment of idiopathic pulmonary fibrosis. Fibrosis)
  • While certain of the preferred embodiments of the present invention have been described and specifically exemplified above, it is not intended that the invention be limited to such embodiments. It will be apparent to one skilled in the art that various changes and modifications can be made therein without departing from the scope of the present invention, as set forth in the following claims.

Claims (26)

1. A method for detecting the presence or absence of at least one genetic alteration in a target nucleic acid isolated from a patient for assessing susceptibility for developing type 1 diabetes (T1D), said method comprising:
a) providing a target nucleic acid from a patient sample, said target nucleic acid having a predetermined sequence in the normal population;
b) assessing said target nucleic acid for the presence of a single nucleotide polymorphism which is indicative of an increased or decreased susceptibility of developing T1D, wherein genetic alteration is at least one single nucleotide polymorphism (SNP) set forth in Tables 1, 2, 4 or 5.
2. The method as claimed in claim 1, wherein said genetic alteration is selected from the group consisting of inversion, deletion, duplication, and insertion of at least one nucleotide in said sequence.
3. The method of claim 1, wherein said target nucleic acid is assessed for genetic alterations via a method selected from the group consisting of size analysis, hybridization of allele specific probes, allele-specific primer extension, oligomer ligation, DNA sequencing, single-stranded conformation polymorphism, and quantitative PCR.
4. The method as claimed in claim 1, wherein said genetic alteration is a single nucleotide polymorphism (SNP) set forth in Table 2.
5. The method as claimed in claim 1, wherein said genetic alteration is present in a linkage disequilibrium block provided in Table 3.
6. The method of claim 1, wherein said SNP is present in a nucleic acid selected from the group consisting of UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7 encoding nucleic acids.
7. The method of claim 6 wherein said SNP is selected from the group consisting of rs9976767 present on chromosome 21 at position 42709459 within the UBASH3A gene, rs3757247 present on chromosome 6 at position 91014184 in the BACH2 gene and rs7171171 present on chromosome 15 at position 36694333.
8. A method for determining the presence or absence of at least one specific nucleotide in a target nucleic acid for the diagnosis of T1D, said method comprising the steps of:
a) providing a detectable amount of a target nucleic acid polymer isolated from a chromosomal region known to be associated with diabetes,
b) hybridizing said detectable amount of the nucleic acid polymer with one or more oligonucleotide primers, wherein each primer has a nucleotide sequence that is complementary to a sequence in the target nucleic acid polymer,
c) exposing the hybridized nucleic acid polymer to a polymerization agent in a mixture containing at least one deoxynucleotide, said deoxynucleotide comprising a detectable label,
d) analyzing the polymerization mixture of step (c) for the presence or absence of the primer extension product containing the labeled deoxynucleotide, whereby the identity of the specific nucleotide at the defined site is determined; and
e) assessing said target nucleic acid for the presence of a genetic alteration at said at least one single nucleotide loci, the presence of the polymorphism being associated with an altered risk of developing T1D.
9. A kit for practicing the method of claim 8.
10. A nucleic acid comprising at least one SNP identified in Tables 1, 2, 4 and 5.
11. A nucleic acid comprising at least one linkage disequilibrium block provided in Table 3, said nucleic acid comprising a genetic alteration indicative of an altered risk of developing T1D.
12. A microarray comprising the at least one nucleic acid of claim 10 or claim 11, said nucleic acid optionally being between 10 and 50 nucleotides in length.
13. A method of management for T1D comprising:
a) identifying a patient who is at an increased risk of developing T1D by detecting at least one of the polymorphisms in Tables 1, 2, 4, or 5;
b) administering to said patient a therapeutic agent in a therapeutically effective amount to manage T1D.
14. The method of claim 10, wherein said therapeutic agent modulates signaling or glucose regulating function mediated via a gene product selected from the group consisting of UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7.
15. The method according to claim 14, wherein said therapeutic agent is selected from the group consisting of a small molecule, an antibody, a protein, an oligonucleotide, or an siRNA molecule.
16. The method of claim 14, wherein said agent is at least one siRNA provided provided in Tables 6-10 in a pharmaceutically acceptable carrier.
17. The method of claim 14, wherein said therapeutic agent is delivered to an inflammatory cell.
18. The method of claim 14, wherein said therapeutic agent modulates natural killer cell activity.
19. The method of claim 14, wherein said therapeutic agent modulates signaling in an insulin-producing beta cell.
20. A single nucleotide polymorphism associated with an altered risk of developing T1D selected from the group consisting rs9976767 present on chromosome 21 at position 42709459 within the UBASH3A gene, rs3757247 present on chromosome 6 at position 91014184 in the BACH2 gene and gene and rs7171171 present on chromosome 15 at position 36694333 within the RASGRP1 gene.
21. A method for identifying agents which modulate beta cell destruction leading to T1D comprising:
a) providing a test subject having cells expressing a SNP selected from the group consisting those set forth in Tables 1, 2 , 4 or 5;
b) providing a test subject having cells which express the cognate sequence lacking the SNPs of step a);
c) contacting the cells of steps a) and b) with an agent; and
d) determining whether said agent alters beta cell destruction of the cells of step a) relative to those of step b), thereby identifying agents which modulate autoimmune beta cell destruction.
22. An siRNA composition which is effective to down regulate expression of a gene target selected from the group consisting of UBASH3A, GLIS3, RASGRP1, BACH2, and EDG7, having a sequence provided in Tables 6-10 in a pharmaceutically acceptable carrier for delivery to a patient.
23. A method of treating T1D in a patient in need thereof, comprising administration of an effective amount of the composition of claim 22.
24. The method of claim 22 wherein said siRNA modulates a parameter selected from the group consisting of insulin secretion, glucagon secretion and glucosamine induced beta cell apoptosis and the degree of said modification is determined.
25. A method for identifying agents which modulate the diabetic phenotype:
a) providing cells expressing a single nucleotide polymorphism selected from the group consisting of those set forth in Table 1, 2, 4 and 5;
b) providing cells which express the cognate sequences which lack the polymorphisms of step a);
c) contacting the cells of steps a) and b) with a test agent and
d) analyzing whether said agent alters an diabetic associated parameter in cells contacted in step a) relative to those of step b), thereby identifying agents which modulate the diabetic phenotype.
26. The method of claim 25, wherein said parameter is selected from the group consisting of insulin secretion, glucagon secretion and glucosamine induced beta cell apoptosis.
US12/947,564 2008-05-16 2010-11-16 Genetic Alterations on Chromosomes 21q, 6q and 15q and Methods of Use Thereof for the Diagnosis and Treatment of Type I Diabetes Abandoned US20110236370A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/947,564 US20110236370A1 (en) 2008-05-16 2010-11-16 Genetic Alterations on Chromosomes 21q, 6q and 15q and Methods of Use Thereof for the Diagnosis and Treatment of Type I Diabetes
US14/628,147 US10066266B2 (en) 2008-05-16 2015-02-20 Genetic alterations on chromosomes 21Q, 6Q and 15Q and methods of use thereof for the diagnosis and treatment of type 1 diabetes

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US5404008P 2008-05-16 2008-05-16
PCT/US2009/044356 WO2009140679A2 (en) 2008-05-16 2009-05-18 Genetic alterations on chromosomes 21q, 6q and 15q and methods of use thereof for the diagnosis and treatment of type i diabetes
US12/947,564 US20110236370A1 (en) 2008-05-16 2010-11-16 Genetic Alterations on Chromosomes 21q, 6q and 15q and Methods of Use Thereof for the Diagnosis and Treatment of Type I Diabetes

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/044356 Continuation-In-Part WO2009140679A2 (en) 2008-05-16 2009-05-18 Genetic alterations on chromosomes 21q, 6q and 15q and methods of use thereof for the diagnosis and treatment of type i diabetes

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/628,147 Continuation US10066266B2 (en) 2008-05-16 2015-02-20 Genetic alterations on chromosomes 21Q, 6Q and 15Q and methods of use thereof for the diagnosis and treatment of type 1 diabetes

Publications (1)

Publication Number Publication Date
US20110236370A1 true US20110236370A1 (en) 2011-09-29

Family

ID=41319379

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/947,564 Abandoned US20110236370A1 (en) 2008-05-16 2010-11-16 Genetic Alterations on Chromosomes 21q, 6q and 15q and Methods of Use Thereof for the Diagnosis and Treatment of Type I Diabetes
US14/628,147 Active 2029-10-07 US10066266B2 (en) 2008-05-16 2015-02-20 Genetic alterations on chromosomes 21Q, 6Q and 15Q and methods of use thereof for the diagnosis and treatment of type 1 diabetes

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/628,147 Active 2029-10-07 US10066266B2 (en) 2008-05-16 2015-02-20 Genetic alterations on chromosomes 21Q, 6Q and 15Q and methods of use thereof for the diagnosis and treatment of type 1 diabetes

Country Status (6)

Country Link
US (2) US20110236370A1 (en)
EP (1) EP2291544B1 (en)
JP (1) JP6095889B2 (en)
AU (1) AU2009246134B2 (en)
CA (1) CA2724475C (en)
WO (1) WO2009140679A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013059469A1 (en) * 2011-10-18 2013-04-25 The Children's Hospital Of Philadelphia Genetic alterations and methods of use thereof for the diagnosis and treatment of type i diabetes
CN114427002A (en) * 2022-03-17 2022-05-03 江苏省人民医院(南京医科大学第一附属医院) Kit for evaluating type 1 diabetes mellitus risk based on 22 SNP susceptibility sites

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5899527B2 (en) * 2011-03-14 2016-04-06 国立研究開発法人理化学研究所 Method for examining drug eruption risk with antiepileptic drugs based on single nucleotide polymorphism of chromosome 13 short arm 21.33 region
CN109402117B (en) * 2018-11-07 2022-03-01 上海中医药大学附属曙光医院 Adeno-associated virus for silencing expression of mouse intestinal RASGRP1 and preparation method and application thereof
KR102169699B1 (en) * 2019-12-27 2020-10-23 주식회사 클리노믹스 Customized DNA chip for genetic test and manufacturing method thereof)

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
US7205106B1 (en) * 2001-07-20 2007-04-17 Roche Molecular Systems, Inc. Association of polymorphisms in IL4-related genes with autoimmune disease

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1202962A (en) 1982-10-19 1986-04-08 David P. Clifford Substituted n-phenyl-n'-benzoyl ureas and their use as insecticides and acaricides
NZ207394A (en) 1983-03-08 1987-03-06 Commw Serum Lab Commission Detecting or determining sequence of amino acids
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4965188A (en) 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
CA2085127C (en) 1990-06-15 2002-12-10 Barbara Cordell Transgenic non-human mammal displaying the amyloid-forming pathology of alzheimer's disease
WO1993014200A1 (en) 1992-01-07 1993-07-22 Tsi Corporation Transgenic animal models for alzheimer's disease
US5604102A (en) 1992-04-15 1997-02-18 Athena Neurosciences, Inc. Methods of screening for β-amyloid peptide production inhibitors
GB9212416D0 (en) 1992-06-11 1992-07-22 Medical Res Council Reversible binding substances
EP1251170A3 (en) 1992-07-17 2002-10-30 Ribozyme Pharmaceuticals, Inc. Method and reagent for treatment of NF-kappaB dependent animal diseases
US5877399A (en) 1994-01-27 1999-03-02 Johns Hopkins University Transgenic mice expressing APP-Swedish mutation develop progressive neurologic disease
US6447796B1 (en) 1994-05-16 2002-09-10 The United States Of America As Represented By The Secretary Of The Army Sustained release hydrophobic bioactive PLGA microspheres
US5849999A (en) 1996-10-16 1998-12-15 The Mclean Hospital Corporation Transgenic non-human mice expressing Flag-APP-C100 protein develop alzheimer's disease brain morphology and behavior
US7312196B2 (en) * 1997-01-08 2007-12-25 Amylin Pharmaceuticals, Inc. Formulations for amylin agonist peptides
US6057098A (en) 1997-04-04 2000-05-02 Biosite Diagnostics, Inc. Polyvalent display libraries
US6395713B1 (en) 1997-07-23 2002-05-28 Ribozyme Pharmaceuticals, Inc. Compositions for the delivery of negatively charged molecules
AU767195B2 (en) 1999-03-10 2003-11-06 Phogen Limited Delivery of substances to cells
EP1305333A4 (en) 2000-07-31 2006-04-12 Active Motif Peptide-mediated delivery of molecules into cells
US20020130430A1 (en) 2000-12-29 2002-09-19 Castor Trevor Percival Methods for making polymer microspheres/nanospheres and encapsulating therapeutic proteins and other products
GB0104440D0 (en) * 2001-02-22 2001-04-11 Oxford Biomedica Ltd Novel genes
US20030152951A1 (en) 2001-07-20 2003-08-14 Mirel Daniel B. IL-4 receptor sequence variation associated with type 1 diabetes
US7060498B1 (en) 2001-11-28 2006-06-13 Genta Salus Llc Polycationic water soluble copolymer and method for transferring polyanionic macromolecules across biological barriers
US7141540B2 (en) 2001-11-30 2006-11-28 Genta Salus Llc Cyclodextrin grafted biocompatible amphilphilic polymer and methods of preparation and use thereof
CA2441701C (en) * 2002-10-04 2010-07-13 F. Hoffmann-La Roche Ag Detection of il4 and il13 polymorphisms in determination of type 1 diabetes susceptibility
EP2305813A3 (en) * 2002-11-14 2012-03-28 Dharmacon, Inc. Fuctional and hyperfunctional sirna
AU2003290432A1 (en) * 2002-12-26 2004-07-22 Asahi Kasei Pharma Corporation T cell activating gene
WO2005116204A1 (en) * 2004-05-11 2005-12-08 Rnai Co., Ltd. Polynucleotide causing rna interfere and method of regulating gene expression with the use of the same
US20080199514A1 (en) * 2004-07-09 2008-08-21 The Burnham Institute Functional Variant of Lymphoid Tyrosine Phosphatase is Associated with Autoimmune Disorders
US7901885B2 (en) * 2006-05-09 2011-03-08 Dsm Ip Assets B.V. Genes and markers in type 2 diabetes and obesity
JP5493118B2 (en) * 2006-05-31 2014-05-14 国立大学法人名古屋大学 Genes associated with polycystic kidney disease and uses thereof

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
US7205106B1 (en) * 2001-07-20 2007-04-17 Roche Molecular Systems, Inc. Association of polymorphisms in IL4-related genes with autoimmune disease

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
dbSNP rs9976767, November 2003 *
Infinium II HumanHap 500 Beach Chip (Fact sheet, Copyright 2006). *
NEB catalog (1998/1999), pp. 121, 284 *
UCSC Genome Browser on Human (March 2006, NCBI36/hg18 Assembly). *
Wang et al. (BioTechniques, Vol. 39, No. 6, pages 885-892, 2005). *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013059469A1 (en) * 2011-10-18 2013-04-25 The Children's Hospital Of Philadelphia Genetic alterations and methods of use thereof for the diagnosis and treatment of type i diabetes
CN114427002A (en) * 2022-03-17 2022-05-03 江苏省人民医院(南京医科大学第一附属医院) Kit for evaluating type 1 diabetes mellitus risk based on 22 SNP susceptibility sites

Also Published As

Publication number Publication date
WO2009140679A2 (en) 2009-11-19
JP2011523552A (en) 2011-08-18
US10066266B2 (en) 2018-09-04
AU2009246134B2 (en) 2016-03-03
EP2291544A4 (en) 2012-09-12
US20150299793A1 (en) 2015-10-22
EP2291544B1 (en) 2017-10-25
AU2009246134A1 (en) 2009-11-19
CA2724475A1 (en) 2009-11-19
WO2009140679A3 (en) 2010-01-07
EP2291544A2 (en) 2011-03-09
JP6095889B2 (en) 2017-03-15
CA2724475C (en) 2019-05-07
WO2009140679A9 (en) 2012-01-05

Similar Documents

Publication Publication Date Title
US10266896B2 (en) Genetic alterations on chromosome 16 and methods of use thereof for the diagnosis and treatment of type 1 diabetes
US10066266B2 (en) Genetic alterations on chromosomes 21Q, 6Q and 15Q and methods of use thereof for the diagnosis and treatment of type 1 diabetes
US20090312394A1 (en) Protection against and treatment of age related macular degeneration
US20190153523A1 (en) Identification of novel loci in asthma and methods of use thereof for the diagnosis and treatment of asthma
US9926600B2 (en) Genetic alterations associated with type I diabetes and methods for use thereof for diagnosis and treatment
AU2009246086B2 (en) Asthma susceptibility loci located at chromosome 1q31 for use in diagnostic and therapeutic methods
CA3111819A1 (en) Compositions and methods for the diagnosis and treatment of lymphatic system disorders
US20220349008A1 (en) Novel genetic markers for postural orthostatic tachycardia syndrome (pots) and methods of use thereof for diagnosis and treatment of the same
US20090298764A1 (en) Gene and pathway and their use in methods and compositions for predicting onset or progression of autoimmune and/or autoinflammatory diseases
US20110151456A1 (en) Test method for type-2 diabetes using gene polymorphism
US20080194419A1 (en) Genetic Association of Polymorphisms in the Atf6-Alpha Gene with Insulin Resistance Phenotypes
US20130071408A1 (en) Methods for Diagnosis and Treatment of Non-Insulin Dependent Diabetes Mellitus
WO2013113762A1 (en) Methods and kits for predicting the risk of having a cutaneous melanoma in a subject
WO2010065953A1 (en) T cell receptor alpha genes which predispose the airway to bacterial colonization

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE CHILDREN'S HOSPITAL OF PHILADELPHIA, PENNSYLVA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MCGILL UNIVERSITY;REEL/FRAME:026589/0111

Effective date: 20110114

AS Assignment

Owner name: THE CHILDREN'S HOSPITAL OF PHILADELPHIA, PENNSYLVA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HAKONARSON, HAKON;GRANT, STRUAN;BRADFIELD, JONATHAN;REEL/FRAME:027583/0063

Effective date: 20120124

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:CHILDREN'S HOSPITAL OF PHILADELPHIA;REEL/FRAME:027866/0360

Effective date: 20120312

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR, MA

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:CHILDREN'S HOSPITAL OF PHILADELPHIA;REEL/FRAME:051328/0388

Effective date: 20191111