WO2006074061A2 - Procede pour l'identification de risque de faible densite minerale osseuse et traitements correspondants - Google Patents

Procede pour l'identification de risque de faible densite minerale osseuse et traitements correspondants Download PDF

Info

Publication number
WO2006074061A2
WO2006074061A2 PCT/US2005/047411 US2005047411W WO2006074061A2 WO 2006074061 A2 WO2006074061 A2 WO 2006074061A2 US 2005047411 W US2005047411 W US 2005047411W WO 2006074061 A2 WO2006074061 A2 WO 2006074061A2
Authority
WO
WIPO (PCT)
Prior art keywords
polymorphic
nucleic acid
subject
nucleotide sequence
osteoporosis
Prior art date
Application number
PCT/US2005/047411
Other languages
English (en)
Other versions
WO2006074061A3 (fr
Inventor
Steven Mah
Carolyn R. Hoyal-Wrightson
Stefan Kammerer
Matthew R. Nelson
Andreas Braun
Rikard H. Reneland
Original Assignee
Sequenom, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sequenom, Inc. filed Critical Sequenom, Inc.
Priority to US10/578,392 priority Critical patent/US20070292849A1/en
Publication of WO2006074061A2 publication Critical patent/WO2006074061A2/fr
Publication of WO2006074061A3 publication Critical patent/WO2006074061A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6827Hybridisation assays for detection of mutation or polymorphism

Definitions

  • the invention relates to genetic methods for identifying susceptibility to low bone mineral density (BMD) and/or bone damage generally associated with human diseases, and in particular to osteoporosis, and treatments that specifically target the disease.
  • BMD bone mineral density
  • Osteoporosis is a common disease characterized by low bone mineral density (BMD), . deterioration of bone micro-architecture and increased risk of bone damage, such as fracture.
  • BMD bone mineral density
  • Common types of osteoporosis include postmenopausal and senile osteoporosis, which generally occur later in life, e.g., 70+ years.
  • Osteoporosis is a major public health problem which affects quality of life and increases costs to health care providers. It is estimated that 44 million Americans and 100 million people worldwide are at risk for osteoporosis. In the United States today, 10 million individuals are estimated to already have the disease and almost 34 million more, or 55% of the people 50 years of age and older, have low bone mass, which puts them at increased risk of developing osteoporosis and related fractures. Of the 10 million Americans estimated to have osteoporosis, eight million are women and 2 million are men. These numbers are growing as the elderly population increases. It is estimated that by the middle of the next century the number of osteoporosis sufferers will double in the West, but may increase six-fold in Asia and South America.
  • Fracture is the most serious endpoint of osteoporosis, particularly fracture of the hip which affects up to 1.7 million people worldwide each year.
  • One in two women and one in four men over age 50 will have an osteoporosis-related fracture in their lifetime. It is estimated that by the year 2050, the number of hip fractures worldwide will increase to over 6 million, as life expectancy and age of the population increase (See Spangler et al. "The Genetic Component of Osteoporosis Mini-review"; http:www.csa.com.osteointro.html).
  • Peak bone mass is mainly genetically determined, though dietary factors and physical activity can have positive effects. Peak bone mass occurs when skeletal growth ceases, after which time bone loss starts. In contrast to the positive balance that occurs during growth, in osteoporosis, the resorbed cavity is not completely refilled by bone and BMD decreases. Based on studies of family histories, twin studies, and racial factors, some attribute 50-60% of total bone variation (e.g., Bone Mineral Density to genetic effects and suggest there may be a predisposition for osteoporosis.
  • Osteoporosis can be considered a complex genetic trait with variants of several genes underlying the genetic determination of the variability of the phenotype.
  • Low BMD is an important risk factor for fractures, the clinically most relevant feature of osteoporosis.
  • Segregation analysis in families has shown that BMD is under polygenic control.
  • biochemical markers of bone turnover have shown to have strong genetic components.
  • VDR vitamin D receptor
  • BMD bone mineral density
  • the methods comprise detecting the presence or absence of one or more of the polymorphic variations described herein in a human nucleic acid sample.
  • two or more polymorphic variations are detected and in some embodiments, 3 or more, or 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 or more polymorphic variants are detected.
  • nucleic acids that include one or more polymorphic variations associated with occurrence of low BMD, as well as polypeptides encoded by these nucleic acids.
  • identifying candidate therapeutic molecules for osteoporosis and other low BMD-related disorders as well as methods for treating osteoporosis in a subject by identifying a subject at risk of low BMD and treating the subject with a suitable prophylactic, treatment or therapeutic molecule.
  • compositions comprising a cell from a subject suffering from low BMD or at risk of low BMD, and a CJSTP 1 PROL4, GRID2, PDE4D or GPX3/TNIP1 /TNIPl nucleic acid, with a RNAi, siRNA, antisense DNA or RNA, or ribozyme nucleic acid designed from a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence, or a nucleic acid that hybridizes to such a nucleotide sequence under stringent conditions.
  • the RNAi, siRNA, antisense DNA or RNA, or ribozyme nucleic acid is designed from a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence that includes one or more low BMD associated polymorphic variations, and in some instances, specifically interacts with such a nucleotide sequence.
  • arrays of nucleic acids bound to a solid surface in which one or more nucleic acid molecules of the array have a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence, or a fragment or substantially identical nucleic acid thereof, or a complementary nucleic acid of the foregoing.
  • compositions comprising a cell from a subject having low BMD or at risk of low BMD and/or a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 polypeptide, with an antibody that specifically binds to the polypeptide.
  • the antibody specifically binds to an epitope in the polypeptide that includes a non-synonymous amino acid modification associated with low BMD (e.g., results in an amino acid substitution in the encoded polypeptide associated with low BMD).
  • the antibody specifically binds to an epitope comprising an arginine corresponding to position 120 in a PROL4 polypeptide (SEQ ID NO: 12).
  • Figures 1-4 show the position of each SNP in the chromosome on the x-axis, while the y- axis provides the negative logarithm of the p-value comparing the estimated allele frequency in the cases to that of the control group. Also shown in the figures are exons and introns of the genes in approximate chromosomal positions. More specifically, Figure 1 shows proximal SNPs in a CETP region in genomic DNA. Figure 2 shows proximal SNPs in a PROL4 region in genomic DNA. Figure 3 shows proximal SNPs in a GRID2 region in genomic DNA. Figure 4 shows proximal SNPs in a PDE4D region in genomic DNA. Figure 5 shows proximal SNPs in a GPX3/TNIP1 region in genomic DNA. Detailed Description
  • polymorphic variants described in a CETP, PROL4, GRID2, PDE4D and GPX3/TNIP1 loci in human genomic DNA are associated with occurrence of low BMD in subjects.
  • detecting genetic determinants in and around this locus associated with an increased risk of low BMD occurrence can lead to early identification of a risk of low BMD, or its associated disorders such as osteoporosis, and early application of preventative and treatment measures.
  • Associating the polymorphic variants with low BMD also has provided new targets for diagnosing low BMD, for prognosing osteoporosis, and methods for screening molecules useful in osteoporosis treatments and osteoporosis preventatives.
  • CETP Cholestery ester transfer protein transfers cholesteryl esters between lipoproteins.
  • the transfer of insoluble cholesteryl esters among lipoprotein particles (HDL to LDL) by CETP is a step in normal cholesterol homeostasis.
  • the lipoprotein phenotype of CETP deficiency which is characterized by increased levels of HDL and decreased levels of low density lipoprotein (LDL), appears to have antiatherogenic potential. Bone mineral density is reduced by atherogenic diets (increased LDL/HDL ratios), possibly as a result of a shift in the balance of bone marrow stromal cells away from osteoblasts and towards adipocytes, which could result in reduced bone synthesis during remodeling.
  • Lipoproteins may directly regulate bone density via the LRP5, the low-density lipoprotein receptor related protein 5.
  • LRP5 was cloned from an osteoblast cDNA library (Dong, et al.: Biochem Biophys Res Commun. 1998 Oct 29;251(3):784-90) and mouse knockouts show a reduced BMD phenotype (Kato et. al: JCeIl Biol. 2002 Apr 15;157(2):303-14).
  • a G171V mutation in LRP5 resulted in increased numbers of osteoblasts, increased AP activity, and increased trabecular number and thickness in mice (Babij et al.: J Bone Miner Res. 2003 Jun; 18(6): 960-74).
  • the human Gl 71V mutation was found to have the same phenotype, presumably by increasing WNT signaling in osteoblasts (Mao et. al.: MoI Cell. 2001 Apr;7(4):801-9). Overall, there is evidence that lipoprotein profiles play a role in osteoblast differentiation.
  • a method for treating osteoporosis or low bone mineral density by modulating a CETP function in a subject suffering from osteoporosis or low bone mineral density.
  • CETP inhibitors and methods of making them are described in US 6,586,613 (Substituted tetrahydronaphthaline and analogous compounds); US 6,562,976 (4- phenyltetrahydrochinoline utilized as an inhibitor of the cholesterol ester transfer protein); US 6,387,929 (4-heteroaryl-tetrahydroquinolines and their use as inhibitors of the cholesterin-ester transfer protein); US 6,291,477 (Tetrahydroquinolines, processes for their preparation, pharmaceutical compositions containing them, and their use to prevent or treat hyperlipoproteinaemia); US 6,218,431 (Substituted biphenyls); US 6,207,671 (Cycloalkano-pyridines); US 6,127,383 (2-aryl-substituted pyridines); US 6,121,330 (5-Hydroxyalkyl substituted phenyls and their use in medicaments for the treatment of arteriosclerosis and hyperlipoproteinaemia); US 6,
  • PROL4 also known as Lacrimal proline rich protein (LPRP) is a member of the proline- rich secreted protein family and contains a conserved acidic N-terminal region. It has 45.5% amino acid homology to a salivary Parotid acidic protein (PRHl). PRHl and related proline-rich (salivary) proteins act as potent inhibitors of hydroxyapatite crystal growth and bind calcium with a strength that suggests that they are important in maintaining the concentration of ionic calcium in saliva. The N- terminal region of salivary proline-rich proteins, which is also the region of strongest homology to PROL4, mediates this binding.
  • PRHl salivary Parotid acidic protein
  • PRHl and related proline-rich (salivary) proteins act as potent inhibitors of hydroxyapatite crystal growth and bind calcium with a strength that suggests that they are important in maintaining the concentration of ionic calcium in saliva.
  • the N- terminal region of salivary proline-rich proteins which is also the region
  • PROL4 plays a role in maintaining calcium levels during bone remodeling, and that a loss of function of PROL4 facilitates bone loss.
  • a method of treating low BMD or osteoporosis by increasing expression of PROL4 or otherwise increasing levels of active PROL4 in a patient suffering from low BMD or osteoporosis is expected that PROL4 plays a role in maintaining calcium levels during bone remodeling, and that a loss of function of PROL4 facilitates bone loss.
  • a method of treating low BMD or osteoporosis by increasing expression of PROL4 or otherwise increasing levels of active PROL4 in a patient suffering from low BMD or osteoporosis.
  • Human glutamate receptor delta-2 (GRID2) is a member of the family of ionotropic glutamate receptors that are excitatory neurotransmitter receptors in mammalian brain.
  • Glutamate receptors have been detected in osteoblasts and osteoclasts and may regulate bone resorption.
  • Provided is a method of treating low bone mineral density or osteoporosis by modulating GRID2 function in a patient suffering from low BMD or osteoporosis.
  • PDE4D encodes cyclic AMP-dependent phosphodiesterase 4D.
  • Phosphodiesterases are a superfamily of enzymes involved in degradation of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) (Manganiello VC, et al.: Arch Biochem Biophys 1995, 322(1):1-13; and Beavo JA: Physiol Rev 1995, 75(4):725-748).
  • cAMP and cGMP are important second messengers participating in the response of various cells to hormones. In osteoblasts, cAMP produced in response to parathyroid hormone or prostaglandins regulates osteoblastic differentiation (Farndale RW, et al.
  • cAMP Intracellular levels of cAMP are regulated by G protein-coupled adenylyl cyclase (Casperson GF, Bourne HR: Annu Rev Pharmacol Toxicol 1987, 27:371-384), and degradation is mediated by the phosphodiesterases.
  • G protein-coupled adenylyl cyclase (Casperson GF, Bourne HR: Annu Rev Pharmacol Toxicol 1987, 27:371-384), and degradation is mediated by the phosphodiesterases.
  • the phosphodiesterase superfamily consists of seven families, PDEl -7, distinguished by substrate specificity, chromatographic behavior during purification, and affinity for biochemical activators and inhibitors. Of these, the PDE4 family is specific for cAMP and is selectively inhibited by rolipram.
  • PDE4 genes 4A, 4B, 4C, and 4D, have been cloned from rat and humans, all of which are predicted to have multiple protein products due to alternate spicing of RNAs.
  • PDE4 inhibitors have been shown to increase bone formation in normal mice (Kinoshita T, et al. Bone 2000, 27(6): 811-817) and to ameliorate loss of bone mass in animal models of osteopenia (Miyamoto K, et al. Biochem Pharmacol 1997, 54(5):613-617; and Waki Y, et al. Jpn J Pharmacol 1999, 79(4):477-483).
  • PDE4A and PDE4D are expressed in two common mouse osteoblastic cell lines, ST2 and MC3T3-E1, that represent different stages in the osteoblast differentiation pathway (Wakabayashi S, et al.. J Bone Miner Res 2002, 17(2):249-256).
  • PDE4 inhibition with rolipram increased BMP2-induced alkaline phosphatase activity, a marker of early osteoblast differentiation in ST2 cells.
  • rolipram increased the expression of alkalic phosphatase, osteopontin, collagen type I and osteocalcin in the same osteoblast precursor cells (Wakabayashi S, et al. J Bone Miner Res 2002, 17(2):249-256).
  • a method for treating osteoporosis or low bone mineral density by modulating a PDE4 function in a human suffering from osteoporosis or low bone mineral density may increase bone formation and treat an osteoporotic condition.
  • PDE4D inhibitors and methods of making them are described in the following US patents: 6,218,400 (Treatment method using a cGMP-Specific PDE inhibitor), 5,891,896 (Tri-substituted phenyl derivatives useful as PDE FV inhibitors), 5,849,770 (Tri-substituted phenyl derivatives useful as PDE FV inhibitors), 5,712,298 (Fluoroalkoxy-substituted benzamides and their use as cyclic nucleotide phosphodiesterase inhibitors), 5,491,147 (Tri-substituted phenyl derivatives and their use in pharmaceutical compositions and methods of treatment).
  • SNP rs869975 is contained within the GPX3 gene, and the TNIP3 gene may not be ruled out due to linkage disequilibrium; therefore the GPX3/TNIP3 region is considered associated with low BMD.
  • the GPX3 gene encodes the Plasma glutathione peroxidase 3 precursor. Glutathione peroxidase catalyzes the reduction of hydrogen peroxide, organic hydroperoxide, and lipid peroxides by reduced glutathione and functions in the protection of cells against oxidative damage. This enzyme, found mainly in the cytosol of mammalian cells, is unusual in its content of a selenocysteine residue in its active site that is encoded by a TGA opal codon.
  • Osteoblasts produce glutathione peroxidase, possibly as a defense against hydrogen peroxide produced by osteoclasts during bone remodeling and thus may contribute to lower BMD.
  • Provided is a method for treating osteoporosis or low bone mineral density by modulating a GPX function in a human suffering from osteoporosis or low bone mineral density.
  • NAFl was identified by a yeast two-hybrid screen as an interacting protein to the HTV protein, Nef. Subsequently, it was found to be an A20-binding protein that is critical for the A20-mediated negative feedback regulation of NF-kappa B activation in response to tumor necrosis factor (TNF).
  • TNF tumor necrosis factor
  • RA rheumatoid arthritis
  • FEBS Lett. 2003 Sep l l;551(l-3):8-12 tested TNF-alpha- modulated gene expression in cultured primary human synoviocytes in vitro.
  • TNIPl Genes upregulated included TNIPl and implicate TNIPl as a potential modulator of TNF-alpha bioactivity in RA.
  • the differentiation and functions of osteoclasts are stimulated and regulated by osteoblast/stromal cell derived factors, such as receptor activator of NFKB ligand (RANKL).
  • RNKL receptor activator of NFKB ligand
  • the present invention is applicable to any disease in which low BMD and/or bone fracture is a factor, and is therefore particularly concerned with diseases such as osteoporosis.
  • Low BMD is defined by the World Health Organization as 2.5 standard deviations below the age-matched mean of bone mineral density for a given population.
  • Bone damage may be defined as any form of structural damage such as fractures or chips of the bone, and degradation or deterioration of the bone other than normal wear and tear resulting from low bone mineral density or another cause. Such low BMD and/or bone damage is associated with osteoporosis.
  • Osteoporosis or porous bone, is a disease characterized by low bone mass and structural deterioration of bone tissue, leading to bone fragility and an increased susceptibility to fractures, especially of the hip, spine and wrist.
  • primary and secondary there are two types of osteoporosis: primary and secondary.
  • primary osteoporosis includes postmenopausal osteoporosis, age-associated osteoporosis (affecting a majority of individuals over the age of 70 to 80), and idiopathic osteoporosis affecting middle-aged and younger men and women.
  • Secondary osteoporosis is the result of an identifiable disease process or agent.
  • Bone remodeling occurs throughout life, renewing the skeleton and maintaining the strength of bone.
  • This remodeling involves the erosion and filling of discrete sites on the surface of bones, by an organized group of cells called “basic multicellular units” or “BMUs".
  • BMUs primarily consist of "osteoclasts", “osteoblasts”, and their cellular precursors.
  • osteoclasts primarily consist of "osteoclasts", “osteoblasts”, and their cellular precursors.
  • osteoclasts primarily consist of "osteoclasts”, "osteoblasts”, and their cellular precursors.
  • This cavity is then filled with bone by an osteoblast.
  • Preferred methods for the treatment of osteoporosis include an initial diagnostic step to determine the presence of the disorder.
  • Initial diagnostic steps include determination of bone mass and rate of bone remodeling.
  • the rate of bone remodeling can be determined by the measurement of biochemical markers. See, for example, Hui et al, "The Contribution of Bone Loss to Postmenopausal Osteoporosis” Osteoporosis Int. 30 (1990). Diagnosis of those at risk of developing osteoporosis also allows more effective preventive measures. Part of diagnosis includes specialized tests called bone density tests that measure bone density in various sites of the body.
  • Such methods include the measurement of the radiodensity of skeletal radiographs, quantitative computerized tomography, single energy photon absorptiometry, and dual-energy photon absorptiometry. Diagnostic techniques among those useful herein are described in W. A. Peck et al, Physician's Resource Manual on Osteoporosis (1987), published by the National Osteoporosis Foundation. A bone density test can detect the presence of low BMD before a fracture occurs, predict your chances of fracturing in the future, determine rate of bone loss, and monitor response to treatment.
  • individuals having low BMD can be selected for genetic studies. Also, individuals having a family history of low BMD or diagnosed with osteoporosis often are selected for genetic studies. Other selection criteria can include: a tissue or fluid sample derived from an individual characterized as Caucasian; sample derived from an individual of Caucasian paternal and maternal descent; case samples derived from individuals diagnosed with osteoporosis; control samples derived from individuals with normal or high BMD levels and no family history of osteoporosis; and sufficient genomic DNA for all allelotyping and genotyping reactions performed during the study.
  • Phenotype information may included pre- or post-menopausal, familial predisposition, country or origin of mother and father, diagnosis with osteoporosis (date of primary diagnosis, age of individual as of primary diagnosis, osteoporosis-related fracture), biochemical measurements of markers of bone resorption (bone-specific alkaline phosphatase, urinary C-telopeptide of type I collagen, serum osteocalcin), current medication status (thyroid medication, hormone replacement therapy, steroid usage, bisphosphonates and cytotoxic agents for rheumatic diseases). Samples that meet the inclusion criteria and do not meet the exclusion criteria may be added to appropriate pools based on gender and disease status.
  • polymorphic site refers to a region in a nucleic acid at which two or more alternative nucleotide sequences are observed in a significant number of nucleic acid samples from a population of individuals.
  • a polymorphic site may be a nucleotide sequence of two or more nucleotides, an inserted nucleotide or nucleotide sequence, a deleted nucleotide or nucleotide sequence, or a microsatellite, for example.
  • a polymorphic site that is two or more nucleotides in length may be 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more, 20 or more, 30 or more, 50 or more, 75 or more, 100 or more, 500 or more, or about 1000 nucleotides in length, where all or some of the nucleotide sequences differ within the region.
  • a polymorphic site is often one nucleotide in length, which is referred to herein as a "single nucleotide polymorphism" or a "SNP.”
  • each nucleotide sequence is referred to as a "polymorphic variant" or "nucleic acid variant.”
  • polymorphic variants represented in a minority of samples from a population is sometimes referred to as a "minor allele” and the polymorphic variant that is more prevalently represented is sometimes referred to as a "major allele.”
  • minor allele the polymorphic variant represented in a minority of samples from a population
  • major allele the polymorphic variant that is more prevalently represented
  • Individuals who are homozygous with respect to one allele are sometimes predisposed to a different phenotype as
  • allelotyped and/or genotyped In genetic analysis that associate polymorphic variants with low BMD, samples from individuals having low BMD and individuals not having low BMD often are allelotyped and/or genotyped.
  • allelotype refers to a process for determining the allele frequency for a polymorphic variant in pooled DNA samples from cases and controls. By pooling DNA from each group, an allele frequency for each SNP in each group is calculated. These allele frequencies are then compared to one another.
  • genotyped refers to a process for determining a genotype of one or more individuals, where a "genotype" is a representation of one or more polymorphic variants in a population.
  • a genotype or polymorphic variant may be expressed in terms of a "haplotype," which as used herein refers to two or more polymorphic variants occurring within genomic DNA in a group of individuals within a population.
  • haplotype refers to two or more polymorphic variants occurring within genomic DNA in a group of individuals within a population.
  • two SNPs may exist within a gene where each SNP position includes a cytosine variation and an adenine variation.
  • Certain individuals in a population may carry one allele (heterozygous) or two alleles (homozygous) having the gene with a cytosine at each SNP position.
  • the two cytosines corresponding to each SNP in the gene travel together on one or both alleles in these individuals, the individuals can be characterized as having a cytosine/cytosine haplotype with respect to the two SNPs in the gene.
  • phenotype refers to a trait which can be compared between individuals, such as presence or absence of a condition, a visually observable difference in appearance between individuals, metabolic variations, physiological variations, variations in the function of biological molecules, and the like.
  • An example of a phenotype is occurrence of low BMD or clinically diagnosed osteoporosis.
  • a polymorphic variant is statistically significant and often biologically relevant if it is represented in 5% or more of a population, sometimes 10% or more, 15% or more, or 20% or more of a population, and often 25% or more, 30% or more, 35% or more, 40% or more, 45% or more, or 50% or more of a population.
  • a polymorphic variant may be detected on either or both strands of a double-stranded nucleic acid.
  • a polymorphic variant may be located within an intron or exon of a gene or within a portion of a regulatory region such as a promoter, a 5' untranslated region (UTR), a 3' UTR, and in DNA ⁇ e.g., genomic DNA (gDNA) and complementary DNA (cDNA)), RNA ⁇ e.g., mRNA, tRNA, and rRNA), or a polypeptide.
  • Polymorphic variations may or may not result in detectable differences in gene expression, polypeptide structure, or polypeptide function.
  • Polymorphic variants at the following positions were associated with low BMD: rsl 66017, rsl 93694, rs7205804, rsl 801706, rs7205459 and rs821465.
  • a thymine at position 14328, a thymine at position 14996, a guanine at position 37336, a guanine at position 50109, a thymine at position 57618, and a guanine at position 68805 were associated with low BMD.
  • Polymorphic variants at the following positions were associated with an increased risk of low BMD: rs2588350, rs619381, rs620878, rs759055, rs4262797, rs612808, rs3906863, rs7957888, rs763839, rs2418105, rs666841, rs3851578, rs7299578, rs621112, rslO47699, rsl548804, rs2232956, rsl520227 and rs2215716.
  • a cytosine at position 2424, a cytosine at position 3625, a guanine at position 7097, an adenine at position 15688, a guanine at position 22861, a cytosine at position 24138, a cytosine at position 32459, an adenine at position 35151, a guanine at position 36930, an adenine at position 37490, a cytosine at position 38432, an adenine at position 38688, a guanine at position 42665, an adenine at position 43038, a cytosine at position 49075, an adenine at position 50773, an adenine at position 52107, a cytosine at position 75246, and a guanine at position 93715 were associated with risk of low BMD.
  • Polymorphic variants at the following positions were associated with an increased risk of low BMD: rsl433661, rs7679839, rsl368716, rsl905707, rsl905708, rsl994253, rsl485019, rsl905734, rsl485012, rs7670552, rs7691929, rsl948018, rs 1948017, rs 1485024, rs7694568, rs4693320, rs6848749, rs6532406, rs6532407 and rs6819866.
  • a thymine at position 206 a guanine at position 8612, an adenine at position 9285, a thymine at position 11866, a guanine at position 11958, a cytosine at position 28773, a thymine at position 29876, a cytosine at position 35588, a guanine at position 37663, a thymine at position 39375, a cytosine at position 43705, an adenine at position 48962, a cytosine at position 49110, an adenine at position 65050, a cytosine at position 78331, a thymine at position 85405, a guanine at position 86441, an adenine at position 86967, a cytosine at position 87121, and an adenine at position 90969 were associated with risk of low BMD.
  • Polymorphic variants atthe following positions were associated with an increased risk oflow BMD: rs7714708, rsl498602, rs4699934, rsl006431, rsl353747, rsl498608, rsl498609, rs2968010, rs2936202 and rsl391649.
  • an adenine at position 1599, a cytosine at position 39626, athymine at position 40356, athymine at position 43555, athymine at position 44066, athymine at position 49652, a cytosine at position 51103, an adenine at position 57173, a guanine at position 63980, and an adenine at position 82591 were associated with risk oflow BMD.
  • Polymorphic variants at the following positions were associated with an increased risk of low BMD: rsl478398, rsl 160114, rsl 160113, rsl 160112, rs4958858, rs4958431, rs6898463, rs4958859, rs4958860, rs4608909, rs707144, rs2042235, rs3763013, rs2042236, rs8177404, rs8177426, rs8177427, rs8177429, rs3792795, rs4958873, rs8177437, rs869975, rs8177447, rsl 1548, rs2277940, rs8177834, rs2233311, rs2233302, rs7727034, rs7727250, rs3805434, rs7734456,
  • regions with significant association have been identified in loci associated with low BMD. Any polymorphic variants associated with low BMD in a region of significant association can be utilized for embodiments described herein.
  • the following table reports such regions, where "begin” and "end” designate the boundaries of the region according to chromosome positions within the genomic sequence provided in SEQ ID Nos:l-5. The locus, the chromosome on which the locus resides and an incident polymorphism in the locus also are noted.
  • methods for identifying a polymorphic variation associated with low BMD that is proximal to an incident polymorphic variation associated with low BMD which comprises identifying a polymorphic variant proximal to the incident polymorphic variant associated with low BMD, where the incident polymorphic variant is in a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence.
  • the nucleotide sequence often comprises a polynucleotide sequence selected from the group consisting of (a) a polynucleotide sequence of SEQ ID NO's:l-5; (b) a polynucleotide sequence that encodes a polypeptide having an amino acid sequence encoded by a polynucleotide sequence of SEQ ID NO's: 1- 5; and (c) a polynucleotide sequence that encodes a polypeptide having an amino acid sequence that is 90% or more identical to an amino acid sequence encoded by a nucleotide sequence of SEQ ID NO's: 1-5 or a polynucleotide sequence 90% or more identical to the polynucleotide sequence of SEQ ID NO's: 1-5.
  • the presence or absence of an association of the proximal polymorphic variant with low BMD then is determined using a known association method, such as a method described in the Examples hereafter.
  • the incident polymorphic variant is a polymorphic variant associated with low BMD described herein.
  • the proximal polymorphic variant identified sometimes is a publicly disclosed polymorphic variant, which for example, sometimes is published in a publicly available database.
  • the polymorphic variant identified is not publicly disclosed and is discovered using a known method, including, but not limited to, sequencing a region surrounding the incident polymorphic variant in a group of nucleic samples.
  • a known association method such as a method described in the Examples hereafter.
  • the incident polymorphic variant is a polymorphic variant associated with low BMD described herein.
  • the proximal polymorphic variant identified sometimes is a publicly disclosed polymorphic variant, which for example, sometimes is published in a publicly available database.
  • the polymorphic variant identified is not publicly disclosed and is discovered using a known method,
  • the proximal polymorphic variant often is identified in a region surrounding the incident polymorphic variant.
  • this surrounding region is about 50 kb flanking the first polymorphic variant (e.g. about 50 kb 5' of the first polymorphic variant and about 50 kb 3' of the first polymorphic variant), and the region sometimes is composed of shorter flanking sequences, such as flanking sequences of about 40 kb, about 30 kb, about 25 kb, about 20 kb, about 15 kb, about 10 kb, about 7 kb, about 5 kb, or about 2 kb 5' and 3' of the incident polymorphic variant.
  • the region is composed of longer flanking sequences, such as flanking sequences of about 55 kb, about 60 kb, about 65 kb, about 70 kb, about 75 kb, about 80 kb, about 85 kb, about 90 kb, about 95 kb, or about 100 kb 5' and 3' of the incident polymorphic variant.
  • polymorphic variants associated with low BMD are identified iteratively. For example, a first proximal polymorphic variant is associated with low BMD using the methods described above and then another polymorphic variant proximal to the first proximal polymorphic variant is identified (e.g., publicly disclosed or discovered) and the presence or absence of an association of one or more other polymorphic variants proximal to the first proximal polymorphic variant with low BMD is determined.
  • the methods described herein are useful for identifying or discovering additional polymorphic variants that may be used to further characterize a gene, region or loci associated with a condition, a disease (e.g., osteoporosis), or a disorder.
  • allelotyping or genotyping data from the additional polymorphic variants may be used to identify a functional mutation or a region of linkage disequilibrium.
  • polymorphic variants identified or discovered within a region comprising the first polymorphic variant associated with low BMD are genotyped using the genetic methods and sample selection techniques described herein, and it can be determined whether those polymorphic variants are in linkage disequilibrium with the first polymorphic variant.
  • the size of the region in linkage disequilibrium with the first polymorphic variant also can be assessed using these genotyping methods.
  • methods for determining whether a polymorphic variant is in linkage disequilibrium with a first polymorphic variant associated with low BMD and such information can be used in prognosis/diagnosis methods described herein.
  • a nucleic acid variant may be represented on one or both strands in a double-stranded nucleic acid or on one chromosomal complement (heterozygous) or both chromosomal complements (homozygous)).
  • nucleic acid includes DNA molecules (e.g., a complementary DNA (cDNA) and genomic DNA (gDNA)) and RNA molecules (e.g., mRNA, rRNA, siRNA and tRNA) and analogs of DNA or RNA, for example, by use of nucleotide analogs.
  • the nucleic acid molecule can be single-stranded and it is often double-stranded.
  • isolated or purified nucleic acid refers to nucleic acids that are separated from other nucleic acids present in the natural source of the nucleic acid.
  • isolated includes nucleic acids which are separated from the chromosome with which the genomic DNA is naturally associated.
  • An "isolated” nucleic acid is often free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5' and/or 3' ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived.
  • the isolated nucleic acid molecule can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb or 0.1 kb of 5' and/or 3' nucleotide sequences which flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived.
  • an "isolated" nucleic acid molecule such as a cDNA molecule, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free ofchemical precursors or other chemicals when chemically synthesized.
  • the term "gene” refers to a nucleotide sequence that encodes a polypeptide.
  • the nucleic acid often comprises a part of or all of a nucleotide sequence in SEQ ID NO's: 1-5, or a substantially identical sequence thereof.
  • a nucleotide sequence sometimes is a 5' and/or 3' sequence flanking a polymorphic variant described above that is 5-1000 nucleotides in length, or in some embodiments 5-500, 5-100, 5-75, 5-50, 5-45, 5-40, 5-35, 5-30, 5-25 or 5-20 nucleotides in length.
  • Other embodiments are directed to methods of identifying a polymorphic variation at one or more positions in a nucleic acid (e.g., genotyping at one or more positions in the nucleic acid), such as at a position corresponding to rs 1801706 in the CETP gene, rs 1047699 in the PROL4 gene, rs 1948017 in the GRID2 gene, rsl498608 in the PDE4D gene, or rs869975 in the GPX3 gene.
  • nucleic acid fragments are also included herein. These fragments often are a nucleotide sequence identical to a nucleotide sequence of SEQ ID NO's:l-10, a nucleotide sequence substantially identical to a nucleotide sequence of SEQ ID NO' s: 1-10, or a nucleotide sequence that is complementary to the foregoing.
  • the nucleic acid fragment may be identical, substantially identical or homologous to a nucleotide sequence in an exon or an intron in a nucleotide sequence of SEQ ID NO's:l-5, and may encode a domain or part of a domain of a polypeptide.
  • the fragment will comprises one or more of the polymorphic variations described herein as being associated with low BMD.
  • the nucleic acid fragment is often 50, 100, or 200 or fewer base pairs in length, and is sometimes about 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 2000, 3000, 4000, 5000, 10000, 15000, or 20000 base pairs in length.
  • a nucleic acid fragment that is complementary to a nucleotide sequence identical or substantially identical to a nucleotide sequence in SEQ ID NO's: 1 -5 and hybridizes to such a nucleotide sequence under stringent conditions is often referred to as a "probe.”
  • Nucleic acid fragments often include one or more polymorphic sites, or sometimes have an end that is adjacent to a polymorphic site as described hereafter.
  • CETP nucleic acid fragments sometimes encode the mature protein from positions 182 to 1609 of the mRNA sequence (SEQ ID NO: 6), for example.
  • oligonucleotide refers to a nucleic acid comprising about 8 to about 50 covalently linked nucleotides, often comprising from about 8 to about 35 nucleotides, and more often from about 10 to about 25 nucleotides.
  • the backbone and nucleotides within an oligonucleotide may be the same as those of naturally occurring nucleic acids, or analogs or derivatives of naturally occurring nucleic acids, provided that oligonucleotides having such analogs or derivatives retain the ability to hybridize specifically to a nucleic acid comprising a targeted polymorphism.
  • Oligonucleotides described herein may be used as hybridization probes or as components of prognostic or diagnostic assays, for example, as described herein.
  • Oligonucleotides are typically synthesized using standard methods and equipment, such as the ABITM3900 High Throughput DNA Synthesizer and the EXPEDITETM 8909 Nucleic Acid Synthesizer, both of which are available from Applied Biosystems (Foster City, CA). Analogs and derivatives are exemplified in U.S. Pat. Nos.
  • Oligonucleotides may also be linked to a second moiety.
  • the second moiety may be an additional nucleotide sequence such as a tail sequence (e.g., a polyadenosine tail), an adapter sequence (e.g., phage M13 universal tail sequence), and others.
  • the second moiety may be a non- nucleotide moiety such as a moiety which facilitates linkage to a solid support or a label to facilitate detection of the oligonucleotide.
  • Such labels include, without limitation, a radioactive label, a fluorescent label, a chemiluminescent label, a paramagnetic label, and the like.
  • the second moiety may be attached to any position of the oligonucleotide, provided the oligonucleotide can hybridize to the nucleic acid comprising the polymorphism.
  • Nucleic acid coding sequences may be used for diagnostic purposes for detection and control of polypeptide expression.
  • oligonucleotide sequences such as antisense RNA, small-interfering RNA (siRNA) and DNA molecules and rib ⁇ 2ymes that function to inhibit translation of a polypeptide. Antisense techniques and RNA interference techniques are known in the art and are described herein.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • hammerhead motif ribozyme molecules may be engineered that specifically and efficiently catalyze endonucleolytic cleavage of RNA sequences corresponding to or complementary to CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequences.
  • ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites which include the following sequences, GUA, GUU and GUC. Once identified, short RNA sequences of between fifteen (15) and twenty (20) ribonucleotides corresponding to the region of the target gene containing the cleavage site may be evaluated for predicted structural features such as secondary structure that may render the oligonucleotide sequence unsuitable. The suitability of candidate targets may also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using ribonuclease protection assays.
  • Antisense RNA and DNA molecules, siRNA and ribozymes may be prepared by any method known in the art for the synthesis of RNA molecules. These include techniques for chemically synthesizing oligodeoxyribonucleotides well known in the art such as solid phase phosphoramidite chemical synthesis. Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors which incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • DNA encoding a polypeptide also may have a number of uses for the diagnosis of diseases, including low BMD, resulting from aberrant expression of a target gene described herein.
  • the nucleic acid sequence may be used in hybridization assays of biopsies or autopsies to diagnose abnormalities of expression or function (e.g., Southern or Northern blot analysis, in situ hybridization assays).
  • the expression of a polypeptide during embryonic development may also be determined using nucleic acid encoding the polypeptide.
  • production of functionally impaired polypeptide is the cause of various disease states, such as osteoporosis.
  • In situ hybridizations using polypeptide as a probe may be employed to predict problems related to low BMD.
  • administration of human active polypeptide, recombinantly produced as described herein may be used to treat disease states related to functionally impaired polypeptide.
  • gene therapy approaches may be employed to remedy deficiencies of functional polypeptide or to replace or compete with dysfunctional polypeptide.
  • nucleic acid vectors often expression vectors, which contain a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence or a substantially identical sequence thereof.
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked and can include a plasmid, cosmid, or viral vector.
  • the vector can be capable of autonomous replication or it can integrate into a host DNA.
  • Viral vectors may include replication defective retroviruses, adenoviruses and adeno-associated viruses for example.
  • a vector can include a CETP, PROL4, GPJD2, PDE4D or GPX3/TNIP1 nucleotide sequence in a form suitable for expression of an encoded target polypeptide or target nucleic acid in a host cell.
  • a "target polypeptide” is a polypeptide encoded by a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence or a substantially identical nucleotide sequence thereof.
  • the recombinant expression vector typically includes one or more regulatory sequences operatively linked to the nucleic acid sequence to be expressed.
  • the term “regulatory sequence” includes promoters, enhancers and other expression control elements (e.g., polyadenylation signals).
  • Regulatory sequences include those that direct constitutive expression of a nucleotide sequence, as well as tissue-specific regulatory and/or inducible sequences.
  • the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of polypeptide desired, and the like.
  • Expression vectors can be introduced into host cells to produce target polypeptides, including fusion polypeptides.
  • Recombinant expression vectors can be designed for expression of target polypeptides in prokaryotic or eukaryotic cells.
  • target polypeptides can be expressed in E. coli, insect cells (e.g., using baculovirus expression vectors), yeast cells, or mammalian cells. Suitable host cells are discussed further in Goeddel, Gene Expression Technology: Methods in En ⁇ ymology 185, Academic Press, San Diego, CA (1990).
  • the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
  • Fusion vectors add a number of amino acids to a polypeptide encoded therein, usually to the amino terminus of the recombinant polypeptide.
  • Such fusion vectors typically serve three purposes: 1) to increase expression of recombinant polypeptide; 2) to increase the solubility of the recombinant polypeptide; and 3) to aid in the purification of the recombinant polypeptide by acting as a ligand in affinity purification.
  • a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant polypeptide to enable separation of the recombinant polypeptide from the fusion moiety subsequent to purification of the fusion polypeptide.
  • enzymes, and their cognate recognition sequences include Factor Xa, thrombin and enterokinase.
  • Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith & Johnson, Gene 67: 31-40 (1988)), pMAL (New England Biolabs, Beverly, MA) and pRIT5 (Pharmacia, Piscataway, NJ) which fuse glutathione S-transferase (GST), maltose E binding polypeptide, or polypeptide A, respectively, to the target recombinant polypeptide.
  • GST glutathione S-transferase
  • fusion polypeptides can be used in screening assays and to generate antibodies specific for target polypeptides.
  • fusion polypeptide expressed in a retroviral expression vector is used to infect bone marrow cells that are subsequently transplanted into irradiated recipients. The pathology of the subject recipient is then examined after sufficient time has passed ⁇ e.g., six (6) weeks).
  • the expression vector's control functions are often provided by viral regulatory elements.
  • viral regulatory elements For example, commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40.
  • Recombinant mammalian expression vectors are often capable of directing expression of the nucleic acid in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid).
  • tissue-specific promoters include an albumin promoter (liver-specific; Pinkert et al., Genes Dev. 1: 268-277 (1987)), lymphoid-specific promoters (Calame & Eaton, Adv. Immunol.
  • promoters of T cell receptors (Winoto & Baltimore, EMBO J. 8: 729-733 (1989)) promoters of immunoglobulins (Banerji et al., Cell 33: 729-740 (1983); Queen & Baltimore, Cell 33: 741-948 (1983)), neuron-specific promoters (e.g., the neurofilament promoter; Byrne & Ruddle, Proc. Natl. Acad.
  • pancreas-specific promoters Eslund et al, Science 230: 912- 916 (1985)
  • mammary gland-specific promoters e.g., milk whey promoter; U.S. Patent No. 4,873,316 and European Application Publication No. 264,166
  • Developmentally-regulated promoters are sometimes utilized, for example, the murine hox promoters (Kessel & Gruss, Science 249: 374-379 (1990)) and the ⁇ -fetopolypeptide promoter (Campes & Tilghman, Genes Dev. 3: 537-546 (1989)).
  • a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid may also be cloned into an expression vector in an antisense orientation.
  • Regulatory sequences e.g., viral promoters and/or enhancers
  • operatively linked to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid cloned in the antisense orientation can be chosen for directing constitutive, tissue specific or cell type specific expression of antisense RNA in a variety of cell types.
  • Antisense expression vectors can be in the form of a recombinant plasmid, phagemid or attenuated virus.
  • host cells that include a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence within a recombinant expression vector or a fragment of such a nucleotide sequence which facilitate homologous recombination into a specific site of the host cell genome.
  • host cell and “recombinant host cell” are used interchangeably herein. Such terms refer not only to the particular subject cell but rather also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • a host cell can be any prokaryotic or eukaryotic cell.
  • a target polypeptide can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells).
  • bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells).
  • mammalian cells such as Chinese hamster ovary cells (CHO) or COS cells.
  • Other suitable host cells are known to those skilled in the art.
  • Vectors can be introduced into host cells via conventional transformation or transfection techniques.
  • a host cell provided herein can be used to produce (i.e., express) a target polypeptide or a substantially identical polypeptide thereof. Accordingly, further provided are methods for producing a target polypeptide using host cells described herein. In one embodiment, the method includes culturing host cells into which a recombinant expression vector encoding a target polypeptide has been introduced in a suitable medium such that a target polypeptide is produced. In another embodiment, the method further includes isolating a target polypeptide from the medium or the host cell.
  • cells or purified preparations of cells which include a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 transgene, or which otherwise misexpress target polypeptide.
  • Cell preparations can consist of human or non-human cells, e.g., rodent cells, e.g., mouse or rat cells, rabbit cells, or pig cells.
  • the cell or cells include a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 transgene (e.g., a heterologous form of a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 gene, such as a human gene expressed in non-human cells).
  • the transgene can be misexpressed, e.g., overexpressed or underexpressed.
  • the cell or cells include a gene which misexpress an endogenous target polypeptide ⁇ e.g., expression of a gene is disrupted, also known as a knockout).
  • Such cells can serve as a model for studying disorders which are related to mutated or mis-expressed alleles or for use in drug screening.
  • human cells e.g., a hematopoietic stem cells transformed with a CETP, P ROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid.
  • cells or a purified preparation thereof e.g., human cells
  • an endogenous CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid is under the control of a regulatory sequence that does not normally control the expression of the endogenous gene corresponding to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence.
  • the expression characteristics of an endogenous gene within a cell e.g., a cell line or microorganism
  • an endogenous corresponding gene e.g., a gene which is "transcriptionally silent,” not normally expressed, or expressed only at very low levels
  • a regulatory element which is capable of promoting the expression of a normally expressed gene product in that cell.
  • Techniques such as targeted homologous recombinations, can be used to insert the heterologous DNA as described in, e.g., Chappel, US 5,272,071; WO 91/06667, published on May 16, 1991.
  • Non-human transgenic animals that express a heterologous target polypeptide (e.g., expressed from a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid or substantially identical sequence thereof) can be generated. Such animals are useful for studying the function and/or activity of a target polypeptide and for identifying and/or evaluating modulators of the activity of CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acids and encoded polypeptides.
  • a heterologous target polypeptide e.g., expressed from a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid or substantially identical sequence thereof.
  • a "transgenic animal” is a non-human animal such as a mammal (e.g., a non-human primate such as chimpanzee, baboon, or macaque; an ungulate such as an equine, bovine, or caprine; or a rodent such as a rat, a mouse, or an Israeli sand rat), a bird (e.g., a chicken or a turkey), an amphibian (e.g., a frog, salamander, or newt), or an insect (e.g., Drosophila melanogaster), in which one or more of the cells of the animal includes a transgene.
  • a mammal e.g., a non-human primate such as chimpanzee, baboon, or macaque
  • an ungulate such as an equine, bovine, or caprine
  • a rodent such as a rat, a mouse, or an Israeli sand
  • a transgene is exogenous DNA or a rearrangement (e.g., a deletion of endogenous chromosomal DNA) that is often integrated into or occurs in the genome of cells in a transgenic animal.
  • a transgene can direct expression of an encoded gene product in one or more cell types or tissues of the transgenic animal, and other transgenes can reduce expression ⁇ e.g., a knockout).
  • a transgenic animal can be one in which an endogenous nucleic acid homologous to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal (e.g., an embryonic cell of the animal) prior to development of the animal.
  • Intronic sequences and polyadenylation signals can also be included in the transgene to increase expression efficiency of the transgene.
  • One or more tissue-specific regulatory sequences can be operably linked to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence to direct expression of an encoded polypeptide to particular cells.
  • a transgenic founder animal can be identified based upon the presence of a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence in its genome and/or expression of encoded mRNA in tissues or cells of the animals.
  • a transgenic founder animal can then be used to breed additional animals carrying the transgene.
  • transgenic animals carrying a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence can further be bred to other transgenic animals carrying other transgenes.
  • Target polypeptides can be expressed in transgenic animals or plants by introducing, for example, a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid into the genome of an animal that encodes the target polypeptide.
  • the nucleic acid is placed under the control of a tissue specific promoter, e.g., a milk or egg specific promoter, and recovered from the milk or eggs produced by the animal. Also included is a population of cells from a transgenic animal.
  • isolated target polypeptides which are encoded by a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence (e.g., SEQ ID NO's:l-10) or a substantially identical nucleotide sequence thereof, such as the polypeptides having amino acid sequences in SEQ ID NO's:l 1-15.
  • polypeptide as used herein includes proteins and peptides.
  • An "isolated” or “purified” polypeptide or protein is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized.
  • the language "substantially free” means preparation of a target polypeptide having less than about 30%, 20%, 10% and more preferably 5% (by dry weight), of non-target polypeptide (also referred to herein as a "contaminating protein"), or of chemical precursors or non-target chemicals.
  • the target polypeptide or a biologically active portion thereof is recombinantly produced, it is also preferably substantially free of culture medium, specifically, where culture medium represents less than about 20%, sometimes less than about 10%, and often less than about 5% of the volume of the polypeptide preparation.
  • Isolated or purified target polypeptide preparations are sometimes 0.01 milligrams or more or 0.1 milligrams or more, and often 1.0 milligrams or more and 10 milligrams or more in dry weight.
  • the polypeptide fragment may be a domain or part of a domain of a target polypeptide.
  • the polypeptide fragment may be a full-length polypeptide or a mature polypeptide (i.e., the polypeptide minus the signal peptide).
  • a fragment sometimes is a CTEP mature protein that corresponds to amino acid positions 18-493 of SEQ ID NO: 11.
  • the polypeptide fragment may have increased, decreased or unexpected biological activity.
  • the polypeptide fragment is often 50 or fewer, 100 or fewer, or 200 or fewer amino acids in length, and is sometimes 300, 400, 500, 600, 700, or 900 or fewer amino acids in length.
  • Substantially identical target polypeptides may depart from the amino acid sequences of target polypeptides in different manners. For example, conservative amino acid modifications may be introduced at one or more positions in the amino acid sequences of target polypeptides.
  • a "conservative amino acid substitution” is one in which the amino acid is replaced by another amino acid having a similar structure and/or chemical function. Families of amino acid residues having similar structures and functions are well known.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • non-essential amino acids may be replaced.
  • a "non-essential" amino acid is one that can be altered without abolishing or substantially altering the biological function of a target polypeptide, whereas altering an "essential” amino acid abolishes or substantially alters the biological function of a target polypeptide.
  • Amino acids that are conserved among target polypeptides are typically essential amino acids.
  • target polypeptides may exist as chimeric or fusion polypeptides.
  • a target "chimeric polypeptide” or target “fusion polypeptide” includes a target polypeptide linked to a non-target polypeptide.
  • a "non-target polypeptide” refers to a polypeptide having an amino acid sequence corresponding to a polypeptide which is not substantially identical to the target polypeptide, which includes, for example, a polypeptide that is different from the target polypeptide and derived from the same or a different organism.
  • the target polypeptide in the fusion polypeptide can correspond to an entire or nearly entire target polypeptide or a fragment thereof.
  • the non-target polypeptide can be fused to the N-terminus or C-terminus of the target polypeptide.
  • Fusion polypeptides can include a moiety having high affinity for a ligand.
  • the fusion polypeptide can be a GST-target fusion polypeptide in which the target sequences are fused to the C-terminus of the GST sequences, or a polyhistidine-target fusion polypeptide in which the target polypeptide is fused at the N- or C-terminus to a string of histidine residues.
  • Such fusion polypeptides can facilitate purification of recombinant target polypeptide.
  • Fusion polypeptides are commercially available that already encode a fusion moiety (e.g., a GST polypeptide), and a nucleotide sequence in SEQ ID NO's:l-10, or a substantially identical nucleotide sequence thereof, can be cloned into an expression vector such that the fusion moiety is linked in-frame to the target polypeptide.
  • the fusion polypeptide can be a target polypeptide containing a heterologous signal sequence at its N- terminus.
  • expression, secretion, cellular internalization, and cellular localization of a target polypeptide can be increased through use of a heterologous signal sequence.
  • Fusion polypeptides can also include all or a part of a serum polypeptide (e.g., an IgG constant region or human serum albumin).
  • Target polypeptides can be incorporated into pharmaceutical compositions and administered to a subject in vivo. Administration of these target polypeptides can be used to affect the bioavailability of a substrate of the target polypeptide and may effectively increase target polypeptide biological activity in a cell.
  • Target fusion polypeptides may be useful therapeutically for the treatment of disorders caused by, for example, (i) aberrant modification or mutation of a gene encoding a target polypeptide; (ii) mis-regulation of the gene encoding the target polypeptide; and (iii) aberrant post- translational modification of a target polypeptide.
  • target polypeptides can be used as immunogens to produce anti-target antibodies in a subject, to purify target polypeptide ligands or binding partners, and in screening assays to identify molecules which inhibit or enhance the interaction of a target polypeptide with a substrate.
  • polypeptides can be chemically synthesized using techniques known in the art (See, e.g., Creighton, 1983 Proteins. New York, N.Y.: W. H. Freeman and Company; and Hunkapiller et al., (1984) Nature July 12 -18;310(5973):105-ll).
  • a relative short fragment can be synthesized by use of a peptide synthesizer.
  • non-classical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the fragment sequence.
  • Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids, 2,4-diaminobutyric acid, a-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, g- Abu, e-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoroamino acids, designer amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na-methyl amino acids, and amino acid analogs in general. Furthermore, the amino, acid can
  • Polypeptides and polypeptide fragments sometimes are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications may be carried out by known techniques, including but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH4; acetylation, formylation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; and the like.
  • Additional post-translational modifications include, for example, N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O- linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of prokaryotic host cell expression.
  • the polypeptide fragments may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the polypeptide.
  • chemically modified derivatives of polypeptides that can provide additional advantages such as increased solubility, stability and circulating time of the polypeptide, or decreased immunogenicity (see e.g., U.S. Pat. No: 4,179,337.
  • the chemical moieties for derivitization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like.
  • the polypeptides may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three or more attached chemical moieties.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the preferred molecular weight is between about 1 kDa and about 100 kDa (the term "about” indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing.
  • Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a therapeutic protein or analog).
  • polymers should be attached to the polypeptide with consideration of effects on functional or antigenic domains of the polypeptide.
  • attachment methods available to those skilled in the art (e.g., EP 0 401 384 (coupling PEG to G-CSF) and Malik et al. (1992) Exp Hematol. September;20(8):1028-35 (pegylation of GM-CSF using tresyl chloride)).
  • polyethylene glycol may be covalently bound through amino acid residues via a reactive group, such as a free amino or carboxyl group.
  • Reactive groups are those to which an activated polyethylene glycol molecule may be bound.
  • the amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residues; those having a free carboxyl group may include as'partic acid residues, glutamic acid residues and the C-terminal amino acid residue.
  • Sulfhydryl groups may also be used as a reactive group for attaching the polyethylene glycol molecules.
  • the attachment sometimes is at an amino group, such as attachment at the N- terminus or lysine group.
  • Proteins can be chemically modified at the N-terminus.
  • polyethylene glycol as an illustration of such a composition, one may select from a variety of polyethylene glycol molecules (by molecular weight, branching, and the like), the proportion of polyethylene glycol molecules to protein (polypeptide) molecules in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-terminally pegylated protein.
  • the method of obtaining the N- terminally pegylated preparation i.e., separating this moiety from other monopegylated moieties if necessary
  • Selective proteins chemically modified at the N-terminus may be accomplished by reductive alkylation, which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N- terminus with a carbonyl group containing polymer is achieved.
  • nucleotide sequences and polypeptide sequences that are substantially identical to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence and the target polypeptide sequences encoded by those nucleotide sequences, respectively, are included herein.
  • the term "substantially identical” as used herein refers to two or more nucleic acids or polypeptides sharing one or more identical nucleotide sequences or polypeptide sequences, respectively.
  • nucleotide sequences or polypeptide sequences that are 55% or more, 60% or more, 65% or more, 70% or more, 75% or more, 80% or more, 85% or more, 90% or more, 95% or more (each often within a 1%, 2%, 3% or 4% variability) identical to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence or the encoded target polypeptide amino acid sequences.
  • One test for determining whether two nucleic acids are substantially identical is to determine the percent of identical nucleotide sequences or polypeptide sequences shared between the nucleic acids or polypeptides.
  • sequence identity is often performed as follows. Sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • the length of a reference sequence aligned for comparison purposes is sometimes 30% or more, 40% or more, 50% or more, often 60% or more, and more often 70% or more, 80% or more, 90% or more, or 100% of the length of the reference sequence.
  • the nucleotides or amino acids at corresponding nucleotide or polypeptide positions, respectively, are then compared among the two sequences.
  • the nucleotides or amino acids are deemed to be identical at that position.
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, introduced for optimal alignment of the two sequences.
  • Comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. Percent identity between two amino acid or nucleotide sequences can be determined using the algorithm of Meyers & Miller, CABIOS 4: 11-17 (1989), which has been incorporated into the ALIGN program (version 2.0), using a PAMl 20 weight residue table, a gap length penalty of 12 and a gap penalty of 4. Also, percent identity between two amino acid sequences can be determined using the Needleman & Wunsch, J. MoI. Biol.
  • Another manner for determining if two nucleic acids are substantially identical is to assess whether a polynucleotide homologous to one nucleic acid will hybridize to the other nucleic acid under stringent conditions.
  • stringent conditions refers to conditions for hybridization and washing. Stringent conditions are known to those skilled in the art and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N. Y., 6.3.1-6.3.6 (1989). Aqueous and non-aqueous methods are described in that reference and either can be used.
  • stringent hybridization conditions is hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45 0 C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 5O 0 C.
  • Another example of stringent hybridization conditions are hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45 0 C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 55 0 C.
  • a further example of stringent hybridization conditions is hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45 0 C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 6O 0 C.
  • stringent hybridization conditions are hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 65 0 C. More often, stringency conditions are 0.5M sodium phosphate, 7% SDS at 65 0 C, followed by one or more washes at 0.2X SSC, 1% SDS at 65 0 C.
  • SSC sodium chloride/sodium citrate
  • An example of a substantially identical nucleotide sequence to a nucleotide sequence in SEQ ID NO's:l-10 is one that has a different nucleotide sequence but still encodes the same polypeptide sequence encoded by the nucleotide sequence in SEQ ID NO's:l-10.
  • Another example is a nucleotide sequence that encodes a polypeptide having a polypeptide sequence that is more than 70% or more identical to, sometimes more than 75% or more, 80% or more, or 85% or more identical to, and often more than 90% or more and 95% or more identical to a polypeptide sequence encoded by a nucleotide sequence in SEQ ID NO's:l-10.
  • SEQ ID NO's:l-10 typically refers to one or more sequences in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, and/or 10. Many of the embodiments described herein are applicable to (a) a nucleotide sequence of SEQ ID NO's:l-10; (b) a nucleotide sequence which encodes a polypeptide consisting of an amino acid sequence encoded by a nucleotide sequence of SEQ ID NO's:l-10; (c) a nucleotide sequence which encodes a polypeptide that is 90% or more identical to an amino acid sequence encoded by a nucleotide sequence of SEQ ID NO's:l-10, or a nucleotide sequence about 90% or more identical to a nucleotide sequence of SEQ ID NO' s: 1-10; (d) a fragment of a nucleotide sequence of (a), (b), or (c); and/or a nucleotide sequence complementary to the nucleo
  • nucleotide sequences from subjects that differ by naturally occurring genetic variance which sometimes is referred to as background genetic variance
  • background genetic variance e.g., nucleotide sequences differing by natural genetic variance sometimes are 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to one another.
  • Gapped BLAST can be utilized as described in Altschul et at, Nucleic Acids Res. 25(17): 3389-3402 (1997).
  • default parameters of the respective programs e.g., XBLAST and NBLAST
  • default parameters of the respective programs e.g., XBLAST and NBLAST
  • a nucleic acid that is substantially identical to a nucleotide sequence in SEQ ID NO's:l-10 may include polymorphic sites at positions equivalent to those described herein when the sequences are aligned.
  • SNPs in a sequence substantially identical to a sequence in SEQ ID NO's:l-10 can be identified at nucleotide positions that match with or correspond to (i.e., align) nucleotides at SNP positions in each nucleotide sequence in SEQ ID NO's: 1-10.
  • insertion or deletion of a nucleotide sequence from a reference sequence can change the relative positions of other polymorphic sites in the nucleotide sequence.
  • Substantially identical nucleotide and polypeptide sequences include those that are naturally occurring, such as allelic variants (same locus), splice variants, homologs (different locus), and orthologs (different organism) or can be non-naturally occurring.
  • Non-naturally occurring variants can be generated by mutagenesis techniques, including those applied to polynucleotides, cells, or organisms.
  • the variants can contain nucleotide substitutions, deletions, inversions and insertions. Variation can occur in either or both the coding and non-coding regions. The variations can produce both conservative and non-conservative amino acid substitutions (as compared in the encoded product).
  • Orthologs, homologs, allelic variants, and splice variants can be identified using methods known in the art. These variants normally comprise a nucleotide sequence encoding a polypeptide that is 50% or more, about 55% or more, often about 70-75% or more or about 80-85% or more, and sometimes about 90-95% or more identical to the amino acid sequences of target polypeptides or a fragment thereof. Such nucleic acid molecules can readily be identified as being able to hybridize under stringent conditions to a nucleotide sequence in SEQ ID NO's:l-10 or a fragment of this sequence.
  • Nucleic acid molecules corresponding to orthologs, homologs, and allelic variants of a nucleotide sequence in SEQ ID NO's:l-10 can further be identified by mapping the sequence to the same chromosome or locus as the nucleotide sequence in SEQ ID NO's:l-10.
  • substantially identical nucleotide sequences may include codons that are altered with respect to the naturally occurring sequence for enhancing expression of a target polypeptide in a particular expression system.
  • the nucleic acid can be one in which one or more codons are altered, and often 10% or more or 20% or more of the codons are altered for optimized expression in bacteria (e.g., E. coli.), yeast (e.g., S. cervesiae), human (e.g., 293 cells), insect, or rodent (e.g., hamster) cells.
  • nucleotide sequence comprises a polynucleotide sequence selected from the group consisting of: (a) a nucleotide sequence of SEQ ID NO's:l-10; (b) a nucleotide sequence which encodes a polypeptide consisting of an amino acid sequence encoded by a nucleotide sequence of SEQ ID NO's:l-10; (c) a nucleotide sequence which encodes a polypeptide that is 90% or more identical to an amino acid sequence encoded by
  • polymorphic variants at the positions described herein are detected for determining a risk of osteoporosis, and polymorphic variants at positions in linkage disequilibrium with these positions are detected for determining a risk of osteoporosis.
  • SEQ ID NO's:l-10 refers to individual sequences in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and/or 12.
  • Results from prognostic tests may be combined with other test results to diagnose osteoporosis.
  • prognostic results may be gathered, an initial diagnostic test may be ordered based on a determined predisposition to low BMD, and the results of the analysis may be utilized to diagnose osteoporosis.
  • osteoporosis diagnostic methods can be developed from studies used to generate prognostic/diagnostic methods in which populations are stratified into subpopulations having different progressions of osteoporosis.
  • prognostic results may be gathered; a patient's risk factors for developing osteoporosis analyzed (e.g., age, race, family history, age of menopause); and an initial diagnostic test may be ordered based on a determined predisposition to low BMD.
  • a patient's risk factors for developing osteoporosis analyzed e.g., age, race, family history, age of menopause
  • an initial diagnostic test may be ordered based on a determined predisposition to low BMD.
  • Risk factors believed to be associated with low BMD include personal history of fracture after age 50; current low bone mass; history of fracture in a 1° relative; being female; being thin and/or having a small frame; low body weight; advanced age; a family history of osteoporosis; estrogen deficiency, especially as a result of menopause which is early or surgically induced; abnormal absence of menstrual periods (amenorrhea); anorexia nervosa; low lifetime calcium intake; use of certain medications, such as corticosteroids and anticonvulsants; low testosterone levels in men; an inactive lifestyle; current cigarette smoking; excessive use of alcohol; being Caucasian or Asian, although African Americans and Hispanic Americans are at significant risk as well. (See National Osteoporosis Foundation; http://www.nof.org/osteoporosis/stats.htm).
  • the results from predisposition analyses described herein may be combined with other test results indicative of osteoporosis, which were previously, concurrently, or subsequently gathered with respect to the predisposition testing.
  • the combination of the prognostic test results with other test results can be probative of osteoporosis, and the combination can be utilized as an osteoporosis diagnostic.
  • the results of any test indicative of osteoporosis known in the art may be combined with the methods described herein. Examples of such tests are bone density tests that measure bone density in various sites of the body.
  • Such methods include the measurement of the radiodensity of skeletal radiographs, quantitative computerized tomography, single energy photon absorptiometry, and dual-energy photon absorptiometry. Diagnostic techniques among those useful herein are described in W. A. Peck et al., Physician's Resource Manual on Osteoporosis (1987), published by the National Osteoporosis Foundation (incorporated by reference herein).
  • Risk of low BMD sometimes is expressed as a probability, such as an odds ratio, percentage, or risk factor.
  • the risk sometimes is expressed as a relative risk with respect to a population average risk of low BMD, and sometimes is expressed as a relative risk with respect to the lowest risk group.
  • Such relative risk assessments often are based upon penetrance values determined by statistical methods and are particularly useful to clinicians and insurance companies for assessing risk of osteoporosis (e.g., a clinician can target appropriate detection, prevention and therapeutic regimens to a patient after determining the patient's risk of osteoporosis, and an insurance company can fine tune actuarial tables based upon population genotype assessments of osteoporosis risk).
  • Risk of osteoporosis sometimes is expressed as an odds ratio, which is the odds of a particular person having a genotype has or will develop osteoporosis with respect to another genotype group (e.g., the most disease protective genotype or population average).
  • the risk often is based upon the presence or absence of one or more polymorphic variants described herein, and also may be based in part upon phenotypic traits of the individual being tested.
  • two or more polymorphic variations are detected in a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 locus.
  • 3 or more, or 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 or more polymorphic variants are detected in the sample.
  • the nucleic acid sample typically is isolated from a biological sample obtained from a subject.
  • nucleic acid can be isolated from blood, saliva, sputum, urine, cell scrapings, and biopsy tissue.
  • the nucleic acid sample can be isolated from a biological sample using standard techniques, such as the technique described in Example 2.
  • the term "subject” refers primarily to humans but also refers to other mammals such as dogs, cats, and ungulates (e.g., cattle, sheep, and swine).
  • Subjects also include avians (e.g., chickens and turkeys), reptiles, and fish (e.g., salmon), as embodiments described herein can be adapted to nucleic acid samples isolated from any of these organisms.
  • the nucleic acid sample may be isolated from the subject and then directly utilized in a method for determining the presence of a polymorphic variant, or alternatively, the sample may be isolated and then stored (e.g., frozen) for a period of time before being subjected to analysis.
  • the presence or absence of a polymorphic variant is determined using one or both chromosomal complements represented in the nucleic acid sample. Determining the presence or absence of a polymorphic variant in both chromosomal complements represented in a nucleic acid sample from a subject having a copy of each chromosome is useful for determining the zygosity of an individual for the polymorphic variant (i.e., whether the individual is homozygous or heterozygous for the polymorphic variant). Any oligonucleotide-based diagnostic may be utilized to determine whether a sample includes the presence or absence of a polymorphic variant in a sample. For example, primer extension methods, ligase sequence determination methods (e.g., U.S.
  • mismatch sequence determination methods e.g., U.S. Pat. Nos. 5,851,770; 5,958,692; 6,110,684; and 6,183,958
  • microarray sequence determination methods restriction fragment length polymorphism (RFLP), single strand conformation polymorphism detection (SSCP) (e.g., U.S. Pat. Nos. 5,891,625 and 6,013,499)
  • PCR-based assays e.g., TAQMAN ® PCR System (Applied Biosystems)
  • nucleotide sequencing methods may be used.
  • Oligonucleotide extension methods typically involve providing a pair of oligonucleotide primers in a polymerase chain reaction (PCR) or in other nucleic acid amplification methods for the purpose of amplifying a region from the nucleic acid sample that comprises the polymorphic variation.
  • PCR polymerase chain reaction
  • One oligonucleotide primer is complementary to a region 3' of the polymorphism and the other is complementary to a region 5' of the polymorphism.
  • a PCR primer pair may be used in methods disclosed in U.S. Pat. Nos. 4,683,195; 4,683,202, 4,965,188; 5,656,493; 5,998,143; 6,140,054; WO 01/27327; and WO 01/27329 for example.
  • PCR primer pairs may also be used in any commercially available machines that perform PCR, such as any of the GENEAMP ® Systems available from Applied Biosystems. Also, those of ordinary skill in the art will be able to design oligonucleotide primers based upon a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence using knowledge available in the art.
  • extension oligonucleotide that hybridizes to the amplified fragment adjacent to the polymorphic variation.
  • adjacent refers to the 3' end of the extension oligonucleotide being often 1 nucleotide from the 5' end of the polymorphic site, and sometimes 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides from the 5' end of the polymorphic site, in the nucleic acid when the extension oligonucleotide is hybridized to the nucleic acid.
  • extension oligonucleotide then is extended by one or more nucleotides, and the number and/or type of nucleotides that are added to the extension oligonucleotide determine whether the polymorphic variant is present.
  • Oligonucleotide extension methods are disclosed, for example, in U.S. Pat. Nos. 4,656,127; 4,851,331; 5,679,524; 5,834,189; 5,876,934; 5,908,755; 5,912,118; 5,976,802; 5,981,186; 6,004,744; 6,013,431; 6,017,702; 6,046,005; 6,087,095; 6,210,891; and WO 01/20039.
  • Oligonucleotide extension methods using mass spectrometry are described, for example, in U.S. Pat. Nos. 5,547,835; 5,605,798; 5,691,141; 5,849,542; 5,869,242; 5,928,906; 6,043,031; and 6,194,144, and a method often utilized is described herein in Example 2.
  • a microarray can be utilized for determining whether a polymorphic variant is present or absent in a nucleic acid sample.
  • a microarray may include any oligonucleotides described herein, and methods for making and using oligonucleotide microarrays suitable for diagnostic use are disclosed in U.S. Pat. Nos.
  • the microarray typically comprises a solid support and the oligonucleotides may be linked to this solid support by covalent bonds or by non-covalent interactions.
  • the oligonucleotides may also be linked to the solid support directly or by a spacer molecule.
  • a microarray may comprise one or more oligonucleotides complementary to a polymorphic site set forth herein.
  • a kit also may be utilized for determining whether a polymorphic variant is present or absent in a nucleic acid sample.
  • a kit often comprises one or more pairs of oligonucleotide primers useful for amplifying a fragment of a nucleotide sequence of SEQ ID NO's:l-10 or a substantially identical sequence thereof, where the fragment includes a polymorphic site.
  • the kit sometimes comprises a polymerizing agent, for example, a thermostable nucleic acid polymerase such as one disclosed in U.S. Pat. Nos. 4,889,818 or 6,077,664.
  • the kit often comprises an elongation oligonucleotide that hybridizes to a CETP 1 PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence in a nucleic acid sample adjacent to the polymorphic site.
  • the kit includes an elongation oligonucleotide, it also often comprises chain elongating nucleotides, such as dATP, dTTP, dGTP, dCTP, and dITP, including analogs of dATP, dTTP, dGTP, dCTP and dITP, provided that such analogs are substrates for a thermostable nucleic acid polymerase and can be incorporated into a nucleic acid chain elongated from the extension oligonucleotide.
  • chain elongating nucleotides would be one or more chain terminating nucleotides such as ddATP, ddTTP, ddGTP, ddCTP, and the like.
  • the kit comprises one or more oligonucleotide primer pairs, a polymerizing agent, chain elongating nucleotides, at least one elongation oligonucleotide, and one or more chain terminating nucleotides.
  • Kits optionally include buffers, vials, microtiter plates, and instructions for use.
  • An individual identified as being at risk of osteoporosis may be heterozygous or homozygous with respect to the allele associated with low BMD.
  • a subject homozygous for an allele associated with low BMD is at a comparatively high risk of osteoporosis
  • a subject heterozygous for an allele associated with low BMD is at a comparatively intermediate risk of osteoporosis
  • a subject homozygous for an allele associated with normal or high BMD levels ⁇ i.e., a decreased risk of low BMD) is at a comparatively low risk of osteoporosis.
  • a genotype may be assessed for a complementary strand, such that the complementary nucleotide at a particular position is detected.
  • the antibody specifically binds to an epitope comprising an arginine at position 120 in a PROL4 polypeptide.
  • Pharmacogenomics is a discipline that involves tailoring a treatment for a subject according to the subject's genotype as a particular treatment regimen may exert a differential effect depending upon the subject's genotype. For example, based upon the outcome of a prognostic test described herein, a clinician or physician may target pertinent information and preventative or therapeutic treatments to a subject who would be benefited by the information or treatment and avoid directing such information and treatments to a subject who would not be benefited (e.g., the treatment has no therapeutic effect and/or the subject experiences adverse side effects).
  • a particular treatment regimen can exert a differential effect depending upon the subject's genotype.
  • a candidate therapeutic exhibits a significant interaction with a major allele and a comparatively weak interaction with a minor allele (e.g., an order of magnitude or greater difference in the interaction)
  • such a therapeutic typically would not be administered to a subject genotyped as being homozygous for the minor allele, and sometimes not administered to a subject genotyped as being heterozygous for the minor allele.
  • a candidate therapeutic is not significantly toxic when administered to subjects who are homozygous for a major allele but is comparatively toxic when administered to subjects heterozygous or homozygous for a minor allele
  • the candidate therapeutic is not typically administered to subjects who are genotyped as being heterozygous or homozygous with respect to the minor allele.
  • the methods described herein are applicable to pharmacogenomic methods for preventing, alleviating or treating low BMD conditions such as osteoporosis.
  • a nucleic acid sample from an individual may be subjected to a prognostic test described herein.
  • information for preventing or treating osteoporosis and/or one or more osteoporosis treatment regimens then may be prescribed to that subject.
  • a treatment or preventative regimen is specifically prescribed and/or administered to individuals who will most benefit from it based upon their risk of developing osteoporosis assessed by the methods described herein.
  • a treatment or preventative regimen is specifically prescribed and/or administered to individuals who will most benefit from it based upon their risk of developing osteoporosis assessed by the methods described herein.
  • certain embodiments are directed to a method for increasing BMD levels or otherwise treating osteoporosis in a subject, which comprises: detecting the presence or absence of a polymorphic variant associated with low BMD in a nucleotide sequence in a nucleic acid sample from a subject, where the nucleotide sequence comprises a polynucleotide sequence selected from the group consisting of: (a) a nucleotide sequence of SEQ ID NO's:l-10; (b) a nucleotide sequence which encodes a polypeptide consisting of an amino acid sequence encoded by a nucleotide sequence of SEQ ID NO's:l-10; (c) a nucleotide sequence which encodes a polypeptide that is 90% or more identical to an amino acid sequence encoded by a nucleotide sequence of SEQ ID NO's: 1-10, or a nucleotide sequence about 90% or more identical to a nucleotide sequence of SEQ ID NO's:l-10;
  • predisposition results may be utilized in combination with other test results to diagnose low BMD associated conditions, such as osteoporosis.
  • Certain preventative treatments often are prescribed to subjects having a predisposition to osteoporosis and where the subject is diagnosed with low BMD or is diagnosed as having symptoms indicative of early stage osteoporosis.
  • Established diagnostic techniques use x-ray and ultrasonography to measure skeletal parameters of bone size, volume and mineral density to predict fracture risk and to assess response to therapy. Such measurements give a "static" value which can be compared to normal values to aid diagnosis of low bone mass and fracture risk (Schott, Cornier et al. 1998).
  • the World Health Organization defines osteoporosis as a bone mineral density level more than 2.5 standard deviations below the young normal mean.
  • the various techniques used to measure bone mineral density include, a) Dual energy X-ray absorptiometry (DXA) - used to measure bone mass at the lumbar spine and hip, but it can also be applied to measuring total skeletal bone mass, soft-tissue composition and other regional bone measurements.
  • DXA is considered the "gold standard" for BMD measurement; b) high-resolution quantitative computed tomography QCT) - highly sensitive, accurate and specific spinal measurements.
  • An alternative method to predict fracture independently of bone mass is to measure bone turnover.
  • High turnover bone resorption and formation
  • This is a "dynamic" measurement which is assessed with biochemical markers in urine or serum and can be used effectively in therapy monitoring in preference to BNO measurements.
  • biomarkers can provide more accurate fracture predictions over bone mass measurement alone.
  • Osteoporosis-related biomarkers for bone resorption include deoxypyridinoline crosslinks
  • osteoporosis-related biomarkers for bone formation include bone alkaline phosphatase and osteocalcin.
  • the treatment sometimes is preventative (e.g., is prescribed or administered to reduce the probability that a low BMD associated condition arises or progresses), sometimes is therapeutic, and sometimes delays, alleviates or halts the progression of a low BMD associated condition.
  • Any known preventative or therapeutic treatment for alleviating or preventing the occurrence of a low BMD associated disorder is prescribed and/or administered.
  • Preventative treatment for osteoporosis is most effective at the time when bone loss is increasing and before the bones have become fragile and prone to fracturing. Strategies for the prevention of this disease include development of bone density in early adulthood (i.e., building strong bones during childhood and adolescence), and minimization of bone loss in later life.
  • osteoporosis preventative and treatment information can be specifically targeted to subjects in need thereof (e.g., those at risk of low BMD or those that have early stages of osteoporosis)
  • a method for preventing or reducing the risk of developing osteoporosis in a subject which comprises: (a) detecting the presence or absence of a polymorphic variation associated with low BMD at a polymorphic site in a nucleotide sequence in a nucleic acid sample from a subject; (b) identifying a subject with a predisposition to osteoporosis, whereby the presence of the polymorphic variation is indicative of a predisposition to osteoporosis in the subject; and (c) if such a predisposition is identified, providing the subject with information about methods or products to prevent osteoporosis or to delay the onset of osteoporosis.
  • Also provided is a method of targeting information or advertising to a subpopulation of a human population based on the subpopulation being genetically predisposed to a disease or condition which comprises: (a) detecting the presence or absence of a polymorphic variation associated with low BMD at a polymorphic site in a nucleotide sequence in a nucleic acid sample from a subject; (b) identifying the subpopulation of subjects in which the polymorphic variation is associated with low BMD; and (c) providing information only to the subpopulation of subjects about a particular product which may be obtained and consumed or applied by the subject to help prevent or delay onset of the disease or condition.
  • Pharmacogenomics methods also may be used to analyze and predict a response to a osteoporosis treatment or a drug. For example, if pharmacogenomics analysis indicates a likelihood that an individual will respond positively to a osteoporosis treatment with a particular drug, the drug may be administered to the individual. Conversely, if the analysis indicates that an individual is likely to respond negatively to treatment with a particular drug, an alternative course of treatment may be prescribed. A negative response may be defined as either the absence of an efficacious response or the presence of toxic side effects.
  • the response to a therapeutic treatment can be predicted in a background study in which subjects in any of the following populations are genotyped: a population that responds favorably to a treatment regimen, a population that does not respond significantly to a treatment regimen, and a population that responds adversely to a treatment regiment (e.g., exhibits one or more side effects). These populations are provided as examples and other populations and subpopulations may be analyzed. Based upon the results of these analyses, a subject is genotyped to predict whether he or she will respond favorably to a treatment regimen, not respond significantly to a treatment regimen, or respond adversely to a treatment regimen. [0115] The tests described herein also are applicable to clinical drug trials.
  • One or more polymorphic variants indicative of response to an agent for treating osteoporosis or to side effects to an agent for treating osteoporosis may be identified using the methods described herein. Thereafter, potential participants in clinical trials of such an agent may be screened to identify those individuals most likely to respond favorably to the drug and exclude those likely to experience side effects. In that way, the effectiveness of drug treatment may be measured in individuals who respond positively to the drug, without lowering the measurement as a result of the inclusion of individuals who are unlikely to respond positively in the study and without risking undesirable safety problems.
  • another embodiment is a method of selecting an individual for inclusion in a clinical trial of a treatment or drug comprising the steps of: (a) obtaining a nucleic acid sample from an individual; (b) determining the identity of a polymorphic variation which is associated with a positive response to the treatment or the drug, or at least one polymorphic variation which is associated with a negative response to the treatment or the drug in the nucleic acid sample, and (c) including the individual in the clinical trial if the nucleic acid sample contains said polymorphic variation associated with a positive response to the treatment or the drug or if the nucleic acid sample lacks said polymorphic variation associated with a negative response to the treatment or the drug.
  • the polymorphic variation may be in a sequence selected individually or in any combination from the group consisting of (i) a nucleotide sequence of SEQ ID NO's:l-10; (ii) a nucleotide sequence which encodes a polypeptide consisting of an amino acid sequence encoded by a nucleotide sequence of SEQ ID NO's:l-10; (iii) a nucleotide sequence which encodes a polypeptide that is 90% or more identical to an amino acid sequence encoded by a nucleotide sequence of SEQ ID NO's:l-10, or a nucleotide sequence about 90% or more identical to a nucleotide sequence of SEQ ID NO's:l-10; and (iv) a fragment of a polynucleotide sequence of (i), (ii)
  • step (c) optionally comprises administering the drug or the treatment to the individual if the nucleic acid sample contains the polymorphic variation associated with a positive response to the treatment or the drug and the nucleic acid sample lacks said biallelic marker associated with a negative response to the treatment or the drug.
  • Also provided herein is a method of partnering between a diagnostic/prognostic testing provider and a provider of a consumable product, which comprises: (a) the diagnostic/prognostic testing provider detects the presence or absence of a polymorphic variation associated with low BMD at a polymorphic site in a nucleotide sequence in a nucleic acid sample from a subject; (b) the diagnostic/prognostic testing provider identifies the subpopulation of subjects in which the polymorphic variation is associated with low BMD; (c) the diagnostic/prognostic testing provider forwards information to the subpopulation of subjects about a particular product which may be obtained and consumed or applied by the subject to help prevent or delay onset of the disease or condition; and (d) the provider of a consumable product forwards to the diagnostic test provider a fee every time the diagnostic/prognostic test provider forwards information to the subject as set forth in step (c) above.
  • compositions comprising Osteoporosis-Directed Molecules
  • composition comprising a cell from a subject having low BMD or at risk of osteoporosis and one or more molecules specifically directed and targeted to a nucleic acid comprising a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence or amino acid sequence.
  • Such directed molecules include, but are not limited to, a compound that binds to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence or amino acid sequence referenced herein; a nucleic acid that hybridizes to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid under conditions of high stringency; a RNAi or siRNA molecule having a strand complementary to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence; an antisense nucleic acid complementary to an RNA encoded by a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence; a ribozyme that hybridizes to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence; a nucleic acid aptamer that specifically
  • the osteoporosis directed molecule interacts with a nucleic acid or polypeptide variant associated with osteoporosis, such as variants referenced herein.
  • the osteoporosis directed molecule interacts with a polypeptide involved in a signal pathway of a polypeptide encoded by a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence, or a nucleic acid comprising such a nucleotide sequence.
  • compositions sometimes include an adjuvant known to stimulate an immune response, and in certain embodiments, an adjuvant that stimulates a T-cell lymphocyte response.
  • adjuvants are known, including but not limited to an aluminum adjuvant (e.g., aluminum hydroxide); a cytokine adjuvant or adjuvant that stimulates a cytokine response (e.g., interleukin (IL)-12 and/or ⁇ -interferon cytokines); a Freund-type mineral oil adjuvant emulsion (e.g., Freund's complete or incomplete adjuvant); a synthetic lipoid compound; a copolymer adjuvant (e.g., TitreMax); a saponin; Quil A; a liposome; an oil-in-water emulsion (e.g., an emulsion stabilized by Tween 80 and pluronic polyoxyethlene/polyoxypropylene block copolymer (Syntex Adjuvant Formulation);
  • compositions are useful for generating an immune response against a osteoporosis directed molecule (e.g., an HLA-binding subsequence within a polypeptide encoded by a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence).
  • a peptide having an amino acid subsequence of a polypeptide encoded by a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence is delivered to a subject, where the subsequence binds to an HLA molecule and induces a CTL lymphocyte response.
  • the peptide sometimes is delivered to the subject as an isolated peptide or as a minigene in a plasmid that encodes the peptide.
  • Methods for identifying HLA-binding subsequences in such polypeptides are known (see e.g., publication WO02/20616 and PCT application US98/01373 for methods of identifying such sequences).
  • the cell may be in a group of cells cultured in vitro or in a tissue maintained in vitro or present in an animal in vivo (e.g., a rat, mouse, ape or human).
  • a composition comprises a component from a cell such as a nucleic acid molecule (e.g., genomic DNA), a protein mixture or isolated protein, for example.
  • a nucleic acid molecule e.g., genomic DNA
  • the aforementioned compositions have utility in diagnostic, prognostic and pharmacogenomic methods described previously and in osteoporosis therapeutics described hereafter. Certain osteoporosis directed molecules are described in greater detail below.
  • Compounds can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; peptoid libraries (libraries of molecules having the functionalities of peptides, but with a novel, non-peptide backbone which are resistant to enzymatic degradation but which nevertheless remain bioactive (see, e.g., Zuckermann et ah, J. Med. Chem.37: 2678-85 (1994)); spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; "one-bead one-compound” library methods; and synthetic library methods using affinity chromatography selection.
  • Biolibrary and peptoid library approaches are typically limited to peptide libraries, while the other approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam, Anticancer Drug Des. 12: 145, (1997)).
  • Examples of methods for synthesizing molecular libraries are described, for example, in DeWitt et ah, Proc. Natl. Acad. Sci. U.S.A. 90: 6909 (1993); Erb et ah, Proc. Natl. Acad. Sci. USA 91 : 11422 (1994); Zuckermann et ah, J. Med. Chem.
  • Libraries of compounds may be presented in solution (e.g., Houghten, Biotechniques 13: 412-421 (1992)), or on beads (Lam, Nature 354: 82-84 (1991)), chips (Fodor, Nature 364: 555-556 (1993)), bacteria or spores (Ladner, United States Patent No. 5,223,409), plasmids (Cull et ah, Proc. Natl. Acad. Sci. USA 89: 1865-1869 (1992)) or on phage (Scott and Smith, Science 249: 386-390 (1990); Devlin, Science 249: 404-406 (1990); Cwirla et ah, Proc. Natl. Acad. Sci.
  • a compound sometimes alters expression and sometimes alters activity of a polypeptide target and may be a small molecule.
  • Small molecules include, but are not limited to, peptides, peptidomimetics (e.g., peptoids), amino acids, amino acid analogs, polynucleotides, polynucleotide analogs, nucleotides, nucleotide analogs, organic or inorganic compounds (i.e., including heteroorganic and organometallic compounds) having a molecular weight less than about 10,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 5,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 1,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 500 grams per mole, and salts, esters, and other pharmaceutically acceptable forms of such compounds.
  • peptides e.g., peptoids
  • amino acids amino acid analogs
  • polynucleotides polynucleotide analogs
  • nucleotides nucleotide analogs
  • an "antisense” nucleic acid refers to a nucleotide sequence complementary to a "sense" nucleic acid encoding a polypeptide, e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence.
  • the antisense nucleic acid can be complementary to an entire coding strand (e.g., SEQ ID NO: 7-13), or to a portion thereof or a substantially identical sequence thereof.
  • the antisense nucleic acid molecule is antisense to a "noncoding region" of the coding strand of a nucleotide sequence (e.g., 5' and 3' untranslated regions in SEQ ID NO's:l-5).
  • An antisense nucleic acid can be designed such that it is complementary to the entire coding region of an mRNA encoded by a nucleotide sequence (e.g., SEQ ID NO's:l-10), and often the antisense nucleic acid is an oligonucleotide antisense to only a portion of a coding or noncoding region of the mRNA.
  • the antisense oligonucleotide can be complementary to the region surrounding the translation start site of the mRNA, e.g., between the -10 and +10 regions of the target gene nucleotide sequence of interest.
  • An antisense oligonucleotide can be, for example, about 7, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, or more nucleotides in length.
  • the antisense nucleic acids which include the riboizymes described hereafter, can be designed to target a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence, often a variant associated with low BMD, or a substantially identical sequence thereof.
  • minor alleles and major alleles can be targeted, and those associated with a higher risk of osteoporosis are often designed, tested, and administered to subjects.
  • An antisense nucleic acid can be constructed using chemical synthesis and enzymatic ligation reactions using standard procedures.
  • an antisense nucleic acid e.g., an antisense oligonucleotide
  • an antisense nucleic acid can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used.
  • Antisense nucleic acid also can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
  • antisense nucleic acids When utilized as therapeutics, antisense nucleic acids typically are administered to a subject (e.g., by direct injection at a tissue site) or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding a polypeptide and thereby inhibit expression of the polypeptide, for example, by inhibiting transcription and/or translation.
  • antisense nucleic acid molecules can be modified to target selected cells and then are administered systemically.
  • antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface, for example, by linking antisense nucleic acid molecules to peptides or antibodies which bind to cell surface receptors or antigens.
  • Antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. Sufficient intracellular concentrations of antisense molecules are achieved by incorporating a strong promoter, such as a pol II or pol III promoter, in the vector construct.
  • a strong promoter such as a pol II or pol III promoter
  • Antisense nucleic acid molecules sometimes are *-anomeric nucleic acid molecules.
  • An *- anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual * -units, the strands run parallel to each other (Gaultier et al., Nucleic Acids. Res. 15: 6625-6641 (1987)).
  • Antisense nucleic acid molecules can also comprise a 2'-o- methylribonucleotide (Inoue et al., Nucleic Acids Res. 15: 6131-6148 (1987)) or a chimeric RNA-DNA analogue (Inoue et al., FEBS Lett. 215: 327-330 (1987)).
  • Antisense nucleic acids sometimes are composed of DNA or PNA or any other nucleic acid derivatives described previously.
  • an antisense nucleic acid is a ribozyme.
  • a ribozyme having specificity for a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence can include one or more sequences complementary to such a nucleotide sequence, and a sequence having a known catalytic region responsible for mRNA cleavage (see e.g., U.S. Pat. No. 5,093,246 or Haselhoff and Gerlach, Nature 334: 585-591 (1988)).
  • a derivative of a Tetrahymena L-19 IVS RNA is sometimes utilized in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in a mRNA (see e.g., Cech et al. U.S. Patent No. 4,987,071; and Cech et al. U.S. Patent No. 5,116,742).
  • target mRNA sequences can be used to select a catalytic RNA having a specific ribonuc lease activity from a pool of RNA molecules (see e.g., Bartel & Szostak, Science 261: 1411-1418 (1993)).
  • Osteoporosis directed molecules include in certain embodiments nucleic acids that can form triple helix structures with a CETP 1 PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence or a substantially identical sequence thereof, especially one that includes a regulatory region that controls expression of a polypeptide.
  • Gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of a nucleotide sequence referenced herein or a substantially identical sequence (e.g., promoter and/or enhancers) to form triple helical structures that prevent transcription of a gene in target cells (see e.g., Helene, Anticancer Drug Des.
  • Switchback molecules are synthesized in an alternating 5 ' -3 ' , 3 ' -5 ' manner, such that they base pair with first one strand of a duplex and then the other, eliminating the necessity for a sizeable stretch of either purines or pyrimidines to be present on one strand of a duplex.
  • Osteoporosis directed molecules include RNAi and siRNA nucleic acids. Gene expression may be inhibited by the introduction of double-stranded RNA (dsRNA), which induces potent and specific gene silencing, a phenomenon called RNA interference or RNAi.
  • dsRNA double-stranded RNA
  • RNAi RNA interference
  • RNA interference RNA interference
  • siRNA refers to a nucleic acid that forms a double stranded RNA and has the ability to reduce or inhibit expression of a gene or target gene when the siRNA is delivered to or expressed in the same cell as the gene or target gene.
  • siRNA refers to short double-stranded RNA formed by the complementary strands. Complementary portions of the siRNA that hybridize to form the double stranded molecule often have substantial or complete identity to the target molecule sequence.
  • an siRNA refers to a nucleic acid that has substantial or complete identity to a target gene and forms a double stranded siRNA.
  • the targeted region When designing the siRNA molecules, the targeted region often is selected from a given DNA sequence beginning 50 to 100 nucleotides downstream of the start codon. See, e.g., Elbashir et al,. Methods 26:199-213 (2002). Initially, 5' or 3' UTRs and regions nearby the start codon were avoided assuming that UTR-binding proteins and/or translation initiation complexes may interfere with binding of the siRNP or RISC endonuclease complex. Sometimes regions of the target 23 nucleotides in length conforming to the sequence motif AA(Nl 9)TT (N, an nucleotide), and regions with approximately 30% to 70% G/C-content (often about 50% G/C-content) often are selected.
  • AA(Nl 9)TT N, an nucleotide
  • the sequence of the sense siRNA sometimes corresponds to (N19) TT or N21 (position 3 to 23 of the 23-nt motif), respectively. In the latter case, the 3' end of the sense siRNA often is converted to TT.
  • the rationale for this sequence conversion is to generate a symmetric duplex with respect to the sequence composition of the sense and antisense 3' overhangs.
  • the antisense siRNA is synthesized as the complement to position 1 to 21 of the 23-nt motif. Because position 1 of the 23-nt motif is not recognized sequence-specifically by the antisense siRNA, the 3 '-most nucleotide residue of the antisense siRNA can be chosen deliberately.
  • the penultimate nucleotide of the antisense siRNA (complementary to position 2 of the 23-nt motif) often is complementary to the targeted sequence.
  • TT often is utilized.
  • Respective 21 nucleotide sense and antisense siRNAs often begin with a purine nucleotide and can also be expressed from pol III expression vectors without a change in targeting site. Expression of RNAs from pol III promoters often is efficient when the first transcribed nucleotide is a purine.
  • the sequence of the siRNA can correspond to the full length target gene, or a subsequence thereof.
  • the siRNA is about 15 to about 50 nucleotides in length ⁇ e.g., each complementary sequence of the double stranded siRNA is 15-50 nucleotides in length, and the double stranded siRNA is about 15-50 base pairs in length, sometimes about 20-30 nucleotides in length or about 20-25 nucleotides in length, e.g., 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.
  • the siRNA sometimes is about 21 nucleotides in length.
  • siRNA molecules sometimes is of a different chemical composition as compared to native RNA that imparts increased stability in cells (e.g., decreased susceptibility to degradation), and sometimes includes one or more modifications in siSTABLE RNA described at the http address www.dharmacon.com.
  • Antisense, ribozyme, RNAi and siRNA nucleic acids can be altered to form modified nucleic acid molecules.
  • the nucleic acids can be altered at base moieties, sugar moieties or phosphate backbone moieties to improve stability, hybridization, or solubility of the molecule.
  • the deoxyribose phosphate backbone of nucleic acid molecules can be modified to generate peptide nucleic acids (see Hyrup et al., Bioorganic & Medicinal Chemistry 4 (1): 5-23 (1996)).
  • peptide nucleic acid refers to a nucleic acid mimic such as a DNA mimic, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained.
  • the neutral backbone of a PNA can allow for specific hybridization to DNA and RNA under conditions of low ionic strength. Synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described, for example, in Hyrup et al., (1996) supra and Perry-O'Keefe et al, Proc. Natl. Acad. Sci. 93: 14670-675 (1996).
  • PNA nucleic acids can be used in prognostic, diagnostic, and therapeutic applications.
  • PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, for example, inducing transcription or translation arrest or inhibiting replication.
  • PNA nucleic acid molecules can also be used in the analysis of single base pair mutations in a gene, (e.g., by PNA-directed PCR clamping); as "artificial restriction enzymes" when used in combination with other enzymes, (e.g., Sl nucleases (Hyrup (1996) supra)); or as probes or primers for DNA sequencing or hybridization (Hyrup et ah, (1996) supra; Perry-O'Keefe supra).
  • oligonucleotides may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across cell membranes (see e.g., Letsinger et ah, Proc. Natl. Acad. Sci. USA 86: 6553-6556 (1989); Lemaitre et ah, Proc. Natl. Acad. Sci. USA 84: 648-652 (1987); PCT Publication No. W088/09810) or the blood- brain barrier (see, e.g., PCT Publication No. WO 89/10134).
  • peptides e.g., for targeting host cell receptors in vivo
  • agents facilitating transport across cell membranes see e.g., Letsinger et ah, Proc. Natl. Acad. Sci. USA 86: 6553-6556 (1989); Lemaitre et ah, Proc. Natl. Acad
  • oligonucleotides can be modified with hybridization-triggered cleavage agents (See, e.g., Krol et ah, Bio-Techniques 6: 958- 976 (1988)) or intercalating agents. (See, e.g., Zon, Pharm. Res. 5: 539-549 (1988) ).
  • the oligonucleotide may be conjugated to another molecule, (e.g., a peptide, hybridization triggered cross- linking agent, transport agent, or hybridization-triggered cleavage agent).
  • molecular beacon oligonucleotide primer and probe molecules having one or more regions complementary to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence or a substantially identical sequence thereof, two complementary regions one having a fluorophore and one a quencher such that the molecular beacon is useful for quantifying the presence of the nucleic acid in a sample.
  • Molecular beacon nucleic acids are described, for example, in Lizardi et ah, U.S. Patent No. 5,854,033; Nazarenko et ah, U.S. Patent No. 5,866,336, and Livak et ah, U.S. Patent 5,876,930.
  • antibody refers to an immunoglobulin molecule or immunologically active portion thereof, i.e., an antigen-binding portion.
  • immunologically active portions of immunoglobulin molecules include F(ab) and F(ab') 2 fragments which can be generated by treating the antibody with an enzyme such as pepsin.
  • An antibody sometimes is a polyclonal, monoclonal, recombinant (e.g., a chimeric or humanized), fully human, non-human (e.g., murine), or a single chain antibody.
  • An antibody may have effector function and can fix complement, and is sometimes coupled to a toxin or imaging agent.
  • a full-length polypeptide or antigenic peptide fragment encoded by a nucleotide sequence referenced herein can be used as an immunogen or can be used to identify antibodies made with other immunogens, e.g., cells, membrane preparations, and the like.
  • An antigenic peptide often includes at least 8 amino acid residues of the amino acid sequences encoded by a nucleotide sequence referenced herein, or substantially identical sequence thereof, and encompasses an epitope.
  • Antigenic peptides sometimes include 10 or more amino acids, 15 or more amino acids, 20 or more amino acids, or 30 or more amino acids. Hydrophilic and hydrophobic fragments of polypeptides sometimes are used as immunogens.
  • Epitopes encompassed by the antigenic peptide are regions located on the surface of the polypeptide (e.g., hydrophilic regions) as well as regions with high antigenicity.
  • regions located on the surface of the polypeptide e.g., hydrophilic regions
  • an Emini surface probability analysis of the human polypeptide sequence can be used to indicate the regions that have a particularly high probability of being localized to the surface of the polypeptide and are thus likely to constitute surface residues useful for targeting antibody production.
  • the antibody may bind an epitope on any domain or region on polypeptides described herein.
  • chimeric, humanized, and completely human antibodies are useful for applications which include repeated administration to subjects.
  • Chimeric and humanized monoclonal antibodies comprising both human and non-human portions, can be made using standard recombinant DNA techniques.
  • Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in Robinson et al International Application No. PCT/US86/02269; Akira, et al European Patent Application 184,187; Taniguchi, M., European Patent Application 171,496; Morrison et al European Patent Application 173,494; Neuberger et al PCT International Publication No.
  • Completely human antibodies are particularly desirable for therapeutic treatment of human patients.
  • Such antibodies can be produced using transgenic mice that are incapable of expressing endogenous immunoglobulin heavy and light chains genes, but which can express human heavy and light chain genes. See, for example, Lonberg and Huszar, Int. Rev. Immunol. 13: 65-93 (1995); and U.S. Patent Nos. 5,625,126; 5,633,425; 5,569,825; 5,661,016; and 5,545,806.
  • companies such as Abgenix, Inc. (Fremont, CA) and Medarex, Inc. (Princeton, NJ), can be engaged to provide human antibodies directed against a selected antigen using technology similar to that described above.
  • Completely human antibodies that recognize a selected epitope also can be generated using a technique referred to as "guided selection.”
  • a selected non-human monoclonal antibody e.g., a murine antibody
  • This technology is described for example by Jespers et al, Bio/Technology 12: 899-903 (1994).
  • An antibody can be a single chain antibody.
  • a single chain antibody scFV
  • Single chain antibodies can be dimerized or multimerized to generate multivalent antibodies having specificities for different epitopes of the same target polypeptide.
  • Antibodies also may be selected or modified so that they exhibit reduced or no ability to bind an Fc receptor.
  • an antibody may be an isotype or subtype, fragment or other mutant, which does not support binding to an Fc receptor (e.g., it has a mutagenized or deleted Fc receptor binding region).
  • an antibody may be conjugated to a therapeutic moiety such as a cytotoxin, a therapeutic agent or a radioactive metal ion.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to cells.
  • Examples include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1 dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homo logs thereof.
  • Therapeutic agents include, but are not limited to, antimetabolites ⁇ e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thiotepa chlorambucil, melphalan, carmustine (BCNU) and lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis- dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g
  • Antibody conjugates can be used for modifying a given biological response.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a polypeptide such as tumor necrosis factor, ⁇ -interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator; or, biological response modifiers such as, for example, lymphokines, interleukin-1 ("IL-I”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors.
  • an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Patent No. 4,676,
  • An antibody e.g., monoclonal antibody
  • an antibody can be used to isolate target polypeptides by standard techniques, such as affinity chromatography or immunoprecipitation.
  • an antibody can be used to detect a target polypeptide (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the polypeptide.
  • Antibodies can be used diagnostically to monitor polypeptide levels in tissue as part of a clinical testing procedure, e.g., to determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling ⁇ i.e., physically linking) the antibody to a detectable substance ⁇ i.e., antibody labeling).
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin;
  • an example of a luminescent material includes luminol;
  • bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125 1, 131 1, 35 S or 3 H.
  • an antibody can be utilized as a test molecule for determining whether it can treat osteopo
  • An antibody can be made by immunizing with a purified antigen, or a fragment thereof, e.g., a fragment described herein, a membrane associated antigen, tissues, e.g., crude tissue preparations, whole cells, preferably living cells, lysed cells, or cell fractions.
  • a purified antigen or a fragment thereof, e.g., a fragment described herein, a membrane associated antigen, tissues, e.g., crude tissue preparations, whole cells, preferably living cells, lysed cells, or cell fractions.
  • osteoporosis treatments include bone-active phosphonates (See, e.g., U.S. Patent No.
  • bisphosphonates e.g., alendronate (Fosamax®) and risedronate (Actonel®); calcitonin (Miacalcin®); estrogen and hormone therapy, e.g., estrogens (Climara®, Estrace®, Estraderm®, Estratab®, Ogen®, Ortho-Est®, Vivelle®, Premarin®, and others) and estrogens and progestins (brand names, such as ActivellaTM, FemHrt®, Premphase®, Prempro®, and others); parathyroid hormone, e.g.
  • estrogens Climara®, Estrace®, Estraderm®, Estratab®, Ogen®, Ortho-Est®, Vivelle®, Premarin®, and others
  • estrogens and progestins brand names, such as ActivellaTM, FemHrt®, Premphase®, Prempro®, and others
  • parathyroid hormone e.g.
  • teriparatide ⁇ PTH 1-34 ⁇ (Forteo®); as well as, estrogen and estrogen receptor modulators (SERMs), e.g., raloxifene HCl (Evista®), sodium fluoride and vitamin D metabolites.
  • SERMs estrogen and estrogen receptor modulators
  • Any of the above therapeutics may be administered alone or in combination (e.g., bone-active phosphonates and estrogen and hormone therapy).
  • Current therapeutic approaches were largely developed in the absence of defined molecular targets or even a solid understanding of disease pathogenesis. Therefore, provided are methods of identifying candidate therapeutics that target biochemical pathways related to the development of osteoporosis.
  • the methods comprise contacting a test molecule with a target molecule in a system.
  • a "target molecule” as used herein refers to a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid, a substantially identical nucleic acid thereof, or a fragment thereof, and an encoded polypeptide of the foregoing.
  • the methods also comprise determining the presence or absence of an interaction between the test molecule and the target molecule, where the presence of an interaction between the test molecule and the nucleic acid or polypeptide identifies the test molecule as a candidate osteoporosis therapeutic.
  • the interaction between the test molecule and the target molecule may be quantified.
  • Test molecules and candidate therapeutics include, but are not limited to, compounds, antisense nucleic acids, siRNA molecules, ribozymes, polypeptides or proteins encoded by a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleotide sequence, or a substantially identical sequence or fragment thereof, and immunotherapeutics ⁇ e.g., antibodies and HLA-presented polypeptide fragments).
  • a test molecule or candidate therapeutic may act as a modulator of target molecule concentration or target molecule function in a system.
  • a “modulator” may agonize ⁇ i.e., up-regulates) or antagonize ⁇ i.e., down-regulates) a target molecule concentration partially or completely in a system by affecting such cellular functions as DNA replication and/or DNA processing ⁇ e.g., DNA methylation or DNA repair), RNA transcription and/or RNA processing ⁇ e.g., removal of intronic sequences and/or translocation of spliced mRNA from the nucleus), polypeptide production ⁇ e.g., translation of the polypeptide from mRNA), and/or polypeptide post-translational modification ⁇ e.g., glycosylation, phosphorylation, and proteolysis of pro-polypeptides).
  • DNA processing e.g., DNA methylation or DNA repair
  • RNA transcription and/or RNA processing e.g., removal of intronic sequences and/or translocation of spliced mRNA from the nucleus
  • polypeptide production
  • a modulator may also agonize or antagonize a biological function of a target molecule partially or completely, where the function may include adopting a certain structural conformation, interacting with one or more binding partners, ligand binding, catalysis ⁇ e.g., phosphorylation, dephosphorylation, hydrolysis, methylation, and isomerization), and an effect upon a cellular event ⁇ e.g., effecting progression of osteoporosis).
  • a candidate therapeutic increases BMD.
  • system refers to a cell free in vitro environment and a cell-based environment such as a collection of cells, a tissue, an organ, or an organism.
  • a system is "contacted” with a test molecule in a variety of manners, including adding molecules in solution and allowing them to interact with one another by diffusion, cell injection, and any administration routes in an animal.
  • interaction refers to an effect of a test molecule on test molecule, where the effect sometimes is binding between the test molecule and the target molecule, and sometimes is an observable change in cells, tissue, or organism.
  • Test molecule/target molecule interactions can be detected and/or quantified using assays known in the art. For example, an interaction can be determined by labeling the test molecule and/or the target molecule, where the label is covalently or non-covalently attached to the test molecule or target molecule.
  • the label is sometimes a radioactive molecule such as 125 1, 131 I 5 35 S or 3 H, which can be detected by direct counting of radioemission or by scintillation counting.
  • enzymatic labels such as horseradish peroxidase, alkaline phosphatase, or luciferase may be utilized where the enzymatic label can be detected by determining conversion of an appropriate substrate to product.
  • a microphysiometer ⁇ e.g., Cytosensor
  • LAPS light-addressable potentiometric sensor
  • cells typically include a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid, an encoded polypeptide, or substantially identical nucleic acid or polypeptide thereof, and are often of mammalian origin, although the cell can be of any origin.
  • Whole cells, cell homogenates, and cell fractions e.g., cell membrane fractions
  • soluble and/or membrane bound forms of the polypeptide may be utilized.
  • membrane-bound forms of the polypeptide it may be desirable to utilize a solubilizing agent.
  • solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-dodecylmaltoside, octanoyl-N-methylglucamide, decanoyl-N-methylglucamide, Triton® X-IOO, Triton® X-114, Thesit®, Isotridecypoly(ethylene glycol ether) n , 3-[(3-cholamidopropyl)dimethylamminio]-l-propane sulfonate
  • CHAPS 3-[(3-cholamidopropyl)dimethylamminio]-2-hydroxy-l-propane sulfonate
  • CHOPSO 3-[(3-cholamidopropyl)dimethylamminio]-2-hydroxy-l-propane sulfonate
  • N-dodecy l-N,N-dimethy 1-3 -ammonio- 1 -propane sulfonate or N-dodecy l-N,N-dimethy 1-3 -ammonio- 1 -propane sulfonate .
  • An interaction between a test molecule and target molecule also can be detected by monitoring fluorescence energy transfer (FET) (see, e.g., Lakowicz et al., U.S. Patent No. 5,631,169; Stavrianopoulos et al. U.S. Patent No. 4,868,103).
  • FET fluorescence energy transfer
  • a fluorophore label on a first, "donor” molecule is selected such that its emitted fluorescent energy will be absorbed by a fluorescent label on a second, "acceptor” molecule, which in turn is able to fluoresce due to the absorbed energy.
  • the "donor" polypeptide molecule may simply utilize the natural fluorescent energy of tryptophan residues.
  • Labels are chosen that emit different wavelengths of light, such that the "acceptor” molecule label may be differentiated from that of the "donor". Since the efficiency of energy transfer between the labels is related to the distance separating the molecules, the spatial relationship between the molecules can be assessed. In a situation in which binding occurs between the molecules, the fluorescent emission of the "acceptor" molecule label in the assay should be maximal.
  • An FET binding event can be conveniently measured through standard fluorometric detection means well known in the art (e.g., using a fluorimeter).
  • determining the presence or absence of an interaction between a test molecule and a target molecule can be effected by monitoring surface plasmon resonance (see, e.g., Sjolander & Urbaniczk, Anal. Chem. 63: 2338-2345 (1991) and Szabo et al, Curr. Opin. Struct. Biol. 5: 699-705 (1995)).
  • surface plasmon resonance or “biomolecular interaction analysis (BIA)” can be utilized to detect biospecif ⁇ c interactions in real time, without labeling any of the interactants (e.g., BIAcore).
  • the target molecule or test molecules are anchored to a solid phase, facilitating the detection of target molecule/test molecule complexes and separation of the complexes from free, uncomplexed molecules.
  • the target molecule or test molecule is immobilized to the solid support.
  • the target molecule is anchored to a solid surface, and the test molecule, which is not anchored, can be labeled, either directly or indirectly, with detectable labels discussed herein.
  • test molecules may be desirable to immobilize a target molecule, an anti-target molecule antibody, and/or test molecules to facilitate separation of target molecule/test molecule complexes from uncomplexed forms, as well as to accommodate automation of the assay.
  • the attachment between a test molecule and/or target molecule and the solid support may be covalent or non-covalent (see, e.g., U.S. Patent No. 6,022,688 for non-covalent attachments).
  • the solid support may be one or more surfaces of the system, such as one or more surfaces in each well of a microtiter plate, a surface of a silicon wafer, a surface of a bead (see, e.g., Lam, Nature 354: 82-84 (1991)) that is optionally linked to another solid support, or a channel in a microfluidic device, for example.
  • Types of solid supports, linker molecules for covalent and non-covalent attachments to solid supports, and methods for immobilizing nucleic acids and other molecules to solid supports are well known (see, e.g., U.S. Patent Nos. 6,261,776; 5,900,481; 6,133,436; and 6,022,688; and WIPO publication WO 01/18234).
  • target molecule may be immobilized to surfaces via biotin and streptavidin.
  • biotinylated target polypeptide can be prepared from biotin-NHS (N- hydroxy-succinimide) using techniques known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • a target polypeptide can be prepared as a fusion polypeptide.
  • glutathione-S-transferase/target polypeptide fusion can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St.
  • the non-immobilized component is added to the coated surface containing the anchored component.
  • an indirect label can be used to detect complexes anchored on the surface, e.g., by adding a labeled antibody specific for the immobilized component, where the antibody, in turn, can be directly labeled or indirectly labeled with, e.g., a labeled anti-Ig antibody.
  • an assay is performed utilizing antibodies that specifically bind target molecule or test molecule but do not interfere with binding of the target molecule to the test molecule.
  • Such antibodies can be derivitized to a solid support, and unbound target molecule may be immobilized by antibody conjugation.
  • Methods for detecting such complexes include immunodetection of complexes using antibodies reactive with the target molecule, as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the target molecule.
  • Cell free assays also can be conducted in a liquid phase.
  • reaction products are separated from unreacted components, by any of a number of standard techniques, including but not limited to: differential centrifugation (see, e.g., Rivas, G., and Minton, Trends Biochem SciAug;18(8): 284-7 (1993)); chromatography (gel filtration chromatography, ion-exchange chromatography); electrophoresis (see, e.g., Ausubel et ah, eds. Current Protocols in Molecular Biology , J.
  • modulators of target molecule expression are identified.
  • a cell or cell free mixture is contacted with a candidate compound and the expression of target mRNA or target polypeptide is evaluated relative to the level of expression of target mRNA or target polypeptide in the absence of the candidate compound.
  • the candidate compound is identified as an agonist of target mRNA or target polypeptide expression.
  • the candidate compound is identified as an antagonist or inhibitor of target mRNA or target polypeptide expression.
  • binding partners that interact with a target molecule are detected.
  • the target molecules can interact with one or more cellular or extracellular macromolecules, such as polypeptides in vivo, and these interacting molecules are referred to herein as "binding partners.”
  • Binding partners can agonize or antagonize target molecule biological activity.
  • test molecules that agonize or antagonize interactions between target molecules and binding partners can be useful as therapeutic molecules as they can up-regulate or down-regulated target molecule activity in vivo and thereby treat osteoporosis.
  • Binding partners of target molecules can be identified by methods known in the art. For example, binding partners may be identified by lysing cells and analyzing cell lysates by electrophoretic techniques. Alternatively, a two-hybrid assay or three-hybrid assay can be utilized (see, e.g., U.S. Patent No. 5,283,317; Zervos et al., Cell 72:223-232 (1993); Madura et al, J. Biol. Chem. 268: 12046-12054 (1993); Bartel et al, Biotechniques 14: 920-924 (1993); Iwabuchi et al, Oncogene 8: 1693-1696 (1993); and Brent WO94/10300).
  • a two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains.
  • the assay often utilizes two different DNA constructs.
  • a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid (sometimes referred to as the "bait") is fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GAL-4).
  • a DNA sequence from a library of DNA sequences that encodes a potential binding partner (sometimes referred to as the "prey”) is fused to a gene that encodes an activation domain of the known transcription factor.
  • a CETP, PROL4, GRID2, PDE4D or GPX3/TNIP1 nucleic acid can be fused to the activation domain. If the "bait" and the “prey” molecules interact in vivo, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) which is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to identify the potential binding partner.
  • a reporter gene e.g., LacZ
  • a reaction mixture containing the target molecule and the binding partner is prepared, under conditions and for a time sufficient to allow complex formation.
  • the reaction mixture often is provided in the presence or absence of the test molecule.
  • the test molecule can be included initially in the reaction mixture, or can be added at a time subsequent to the addition of the target molecule and its binding partner. Control reaction mixtures are incubated without the test molecule or with a placebo. Formation of any complexes between the target molecule and the binding partner then is detected.
  • Decreased formation of a complex in the reaction mixture containing test molecule as compared to in a control reaction mixture indicates that the molecule antagonizes target molecule/binding partner complex formation.
  • increased formation of a complex in the reaction mixture containing test molecule as compared to in a control reaction mixture indicates that the molecule agonizes target molecule/binding partner complex formation.
  • complex formation of target molecule/binding partner can be compared to complex formation of mutant target molecule/binding partner (e.g., amino acid modifications in a target polypeptide). Such a comparison can be important in those cases where it is desirable to identify test molecules that modulate interactions of mutant but not non-mutated target gene products.
  • the assays can be conducted in a heterogeneous or homogeneous format.
  • target molecule and/or the binding partner are immobilized to a solid phase, and complexes are detected on the solid phase at the end of the reaction.
  • homogeneous assays the entire reaction is carried out in a liquid phase.
  • the order of addition of reactants can be varied to obtain different information about the molecules being tested.
  • test compounds that agonize target molecule/binding partner interactions can be identified by conducting the reaction in the presence of the test molecule in a competition format.
  • test molecules that agonize preformed complexes e.g., molecules with higher binding constants that displace one of the components from the complex, can be tested by adding the test compound to the reaction mixture after complexes have been formed.
  • the target molecule or the binding partner is anchored onto a solid surface (e.g., a microtiter plate), while the non-anchored species is labeled, either directly or indirectly.
  • the anchored molecule can be immobilized by non-covalent or covalent attachments.
  • an immobilized antibody specific for the molecule to be anchored can be used to anchor the molecule to the solid surface.
  • the partner of the immobilized species is exposed to the coated surface with or without the test molecule. After the reaction is complete, unreacted components are removed (e.g., by washing) such that a significant portion of any complexes formed will remain immobilized on the solid surface.
  • the detection of label immobilized on the surface is indicative of complex.
  • an indirect label can be used to detect complexes anchored to the surface; e.g., by using a labeled antibody specific for the initially non-immobilized species.
  • test compounds that inhibit complex formation or that disrupt preformed complexes can be detected.
  • the reaction can be conducted in a liquid phase in the presence or absence of test molecule, where the reaction products are separated from unreacted components, and the complexes are detected (e.g., using an immobilized antibody specific for one of the binding components to anchor any complexes formed in solution, and a labeled antibody specific for the other partner to detect anchored complexes).
  • test compounds that inhibit complex or that disrupt preformed complexes can be identified.
  • a homogeneous assay can be utilized. For example, a preformed complex of the target gene product and the interactive cellular or extracellular binding partner product is prepared.
  • One or both of the target molecule or binding partner is labeled, and the signal generated by the label(s) is quenched upon complex formation (e.g., U.S. Patent No. 4,109,496 that utilizes this approach for immunoassays).
  • Addition of a test molecule that competes with and displaces one of the species from the preformed complex will result in the generation of a signal above background. In this way, test substances that disrupt target molecule/binding partner complexes can be identified.
  • Candidate therapeutics for treating osteoporosis are identified from a group of test molecules that interact with a target molecule.
  • Test molecules are normally ranked according to the degree with which they modulate (e.g., agonize or antagonize) a function associated with the target molecule (e.g., DNA replication and/or processing, RNA transcription and/or processing, polypeptide production and/or processing, and/or biological function/activity), and then top ranking modulators are selected.
  • pharmacogenomic information described herein can determine the rank of a modulator. The top 10% of ranked test molecules often are selected for further testing as candidate therapeutics, and sometimes the top 15%, 20%, or 25% of ranked test molecules are selected for further testing as candidate therapeutics.
  • Candidate therapeutics typically are formulated for administration to a subject.
  • Formulations and pharmaceutical compositions typically include in combination with a pharmaceutically acceptable carrier one or more target molecule modulators.
  • the modulator often is a test molecule identified as having an interaction with a target molecule by a screening method described above.
  • the modulator may be a compound, an antisense nucleic acid, a ribozyme, an antibody, or a binding partner.
  • formulations may comprise a target polypeptide or fragment thereof in combination with a pharmaceutically acceptable carrier.
  • the term "pharmaceutically acceptable carrier” includes solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Supplementary active compounds can also be incorporated into the compositions. Pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • a pharmaceutical composition typically is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • Oral compositions generally include an inert diluent or an edible carrier.
  • the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules, e.g., gelatin capsules.
  • Oral compositions can also be prepared using a fluid carrier for use as a mouthwash.
  • Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • Molecules can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • active molecules are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. Materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 5 o/ED 5O .
  • Molecules which exhibit high therapeutic indices are preferred. While molecules that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such molecules lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC 50 i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • a therapeutically effective amount of protein or polypeptide ranges from about 0.001 to 30 mg/kg body weight, sometimes about 0.01 to 25 mg/kg body weight, often about 0.1 to 20 mg/kg body weight, and more often about 1 to 10 mg/kg, 2 to 9 mg/kg, 3 to 8 mg/kg, 4 to 7 mg/kg, or 5 to 6 mg/kg body weight.
  • the protein or polypeptide can be administered one time per week for between about 1 to 10 weeks, sometimes between 2 to 8 weeks, often between about 3 to 7 weeks, and more often for about 4, 5, or 6 weeks.
  • treatment of a subject with a therapeutically effective amount of a protein, polypeptide, or antibody can include a single treatment or, preferably, can include a series of treatments.
  • polypeptide formulations featured herein is a method for treating osteoporosis in a subject, which comprises contacting one or more cells in the subject with a first polypeptide, where the subject comprises a second polypeptide having one or more polymorphic variations associated with low BMD, and where the first polypeptide comprises fewer polymorphic variations associated with low BMD than the second polypeptide.
  • the first and second polypeptides are encoded by a nucleic acid which comprises a nucleotide sequence in SEQ ID NO's:l-10; a nucleotide sequence which encodes a polypeptide consisting of an amino acid sequence encoded by a nucleotide sequence referenced in SEQ ID NO's:l-10; a nucleotide sequence which encodes a polypeptide that is 90% or more identical to an amino acid sequence encoded by a nucleotide sequence of SEQ ID NO's:l-10 and a nucleotide sequence 90% or more identical to a nucleotide sequence in SEQ ID NO's:l-10.
  • the subject often is a human.
  • a dosage of 0.1 mg/kg of body weight (generally 10 mg/kg to 20 mg/kg) is often utilized. If the antibody is to act in the brain, a dosage of 50 mg/kg to 100 mg/kg is often appropriate. Generally, partially human antibodies and fully human antibodies have a longer half-life within the human body than other antibodies. Accordingly, lower dosages and less frequent administration is often possible. Modifications such as lipidation can be used to stabilize antibodies and to enhance uptake and tissue penetration (e.g., into the brain). A method for lipidation of antibodies is described by Cruikshank et al, J. Acquired Immune Deficiency Syndromes and Human Retrovirology 14:193 (1997).
  • Antibody conjugates can be used for modifying a given biological response, the drug moiety is not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a polypeptide such as tumor necrosis factor, .alpha.-interferon, .beta.-interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator; or, biological response modifiers such as, for example, lymphokines, interleukin-1 ("IL-I”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors.
  • a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin
  • a polypeptide such as tumor necrosis factor, .alpha.-interferon, .beta.-interferon, nerve
  • exemplary doses include milligram or microgram amounts of the compound per kilogram of subject or sample weight, for example, about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram. It is understood that appropriate doses of a small molecule depend upon the potency of the small molecule with respect to the expression or activity to be modulated.
  • a physician, veterinarian, or researcher may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained.
  • the specific dose level for any particular animal subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, any drug combination, and the degree of expression or activity to be modulated.
  • gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see, e.g., U.S. Patent 5,328,470) or by stereotactic injection (see e.g., Chen et al, (1994) Proc. Natl. Acad. Sci. USA Pi:3054-3057).
  • Pharmaceutical preparations of gene therapy vectors can include a gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
  • the complete gene delivery vector can be produced intact from recombinant cells (e.g., retroviral vectors) the pharmaceutical preparation can include one or more cells which produce the gene delivery system. Examples of gene delivery vectors are described herein. Therapeutic Methods
  • a therapeutic formulation described above can be administered to a subject in need of a therapeutic for inducing a desired biological response.
  • Therapeutic formulations can be administered by any of the paths described herein. With regard to both prophylactic and therapeutic methods of treatment, such treatments may be specifically tailored or modified, based on knowledge obtained from pharmacogenomic analyses described herein.
  • treatment is defined as the application or administration of a therapeutic formulation to a subject, or application or administration of a therapeutic agent to an isolated tissue or cell line from a subject with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect osteoporosis, symptoms of osteoporosis or a predisposition towards osteoporosis.
  • a therapeutic formulation includes, but is not limited to, small molecules, peptides, antibodies, ribozymes and antisense oligonucleotides.
  • Administration of a therapeutic formulation can occur prior to the manifestation of symptoms characteristic of low BMD, such that osteoporosis is prevented or delayed in its progression.
  • the appropriate therapeutic composition can be determined based on screening assays described herein.
  • modulators include, but are not limited to, small organic or inorganic molecules; antibodies (including, for example, polyclonal, monoclonal, humanized, anti-idiotypic, chimeric or single chain antibodies, and Fab, F(ab') 2 and Fab expression library fragments, scFV molecules, and epitope-binding fragments thereof); and peptides, phosphopeptides, or polypeptides.
  • antisense and ribozyme molecules that inhibit expression of the target gene can also be used to reduce the level of target gene expression, thus effectively reducing the level of target gene activity.
  • triple helix molecules can be utilized in reducing the level of target gene activity.
  • Antisense, ribozyme and triple helix molecules are discussed above. It is possible that the use of antisense, ribozyme, and/or triple helix molecules to reduce or inhibit mutant gene expression can also reduce or inhibit the transcription (triple helix) and/or translation (antisense, ribozyme) of mRNA produced by normal target gene alleles, such that the concentration of normal target gene product present can be lower than is necerney for a normal phenotype.
  • nucleic acid molecules that encode and express target gene polypeptides exhibiting normal target gene activity can be introduced into cells via gene therapy method.
  • the target gene encodes an extracellular polypeptide
  • nucleic acid molecules may be utilized in treating or preventing osteoporosis.
  • Aptamers are nucleic acid molecules having a tertiary structure which permits them to specifically bind to ligands ⁇ see, e.g., Osborne, et al, Curr. Opin. Chem. Biol.1(1): 5-9 (1997); and Patel, D. J., Curr. Opin. Chem. Biol. Jun;l(l): 32-46 (1997)).
  • nucleic acid molecules for osteoporosis treatment is gene therapy, which can also be referred to as allele therapy.
  • a gene therapy method for treating osteoporosis in a subject which comprises contacting one or more cells in the subject or from the subject with a nucleic acid having a first nucleotide sequence.
  • Genomic DNA in the subject comprises a second nucleotide sequence having one or more polymorphic variations associated with low BMD ⁇ e.g., the second nucleic acid has a nucleotide sequence in SEQ ID NO's:l-10).
  • the first and second nucleotide sequences typically are substantially identical to one another, and the first nucleotide sequence comprises fewer polymorphic variations associated with low BMD than the second nucleotide sequence.
  • the first nucleotide sequence may comprise a gene sequence that encodes a full- length polypeptide or a fragment thereof.
  • the subject is often a human. Allele therapy methods often are utilized in conjunction with a method of first determining whether a subject has genomic DNA that includes polymorphic variants associated with low BMD.
  • a method which comprises contacting one or more cells in the subject or from the subject with a polypeptide encoded by a nucleic acid having a first nucleotide sequence.
  • Genomic DNA in the subject comprises a second nucleotide sequence having one or more polymorphic variations associated with low BMD ⁇ e.g., the second nucleic acid has a nucleotide sequence in SEQ ID NO's:l-10).
  • the first and second nucleotide sequences typically are substantially identical to one another, and the first nucleotide sequence comprises fewer polymorphic variations associated with low BMD than the second nucleotide sequence.
  • the first nucleotide sequence may comprise a gene sequence that encodes a full-length polypeptide or a fragment thereof. The subject is often a human.
  • antibodies can be generated that are both specific for target molecules and that reduce target molecule activity. Such antibodies may be administered in instances where antagonizing a target molecule function is appropriate for the treatment of osteoporosis.
  • the target molecule is intracellular and whole antibodies are used, internalizing antibodies may be preferred.
  • Lipofectin or liposomes can be used to deliver the antibody or a fragment of the Fab region that binds to the target antigen into cells. Where fragments of the antibody are used, the smallest inhibitory fragment that binds to the target antigen is preferred. For example, peptides having an amino acid sequence corresponding to the Fv region of the antibody can be used. Alternatively, single chain neutralizing antibodies that bind to intracellular target antigens can also be administered.
  • Such single chain antibodies can be administered, for example, by expressing nucleotide sequences encoding single-chain antibodies within the target cell population ⁇ see, e.g., Marasco et al, Proc. Natl. Acad. ScL USA 90: 7889-7893 (1993)).
  • Modulators can be administered to a patient at therapeutically effective doses to treat osteoporosis.
  • a therapeutically effective dose refers to an amount of the modulator sufficient to result in amelioration of symptoms of osteoporosis.
  • Toxicity and therapeutic efficacy of modulators can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 ZED 50 .
  • Modulators that exhibit large therapeutic indices are preferred. While modulators that exhibit toxic side effects can be used, care should be taken to design a delivery system that targets such molecules to the site of affected tissue in order to minimize potential damage to uninfected cells, thereby reducing side effects.
  • Data obtained from cell culture assays and animal studies can be used in formulating a range of dosages for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage can vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC 50 i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms
  • levels in plasma can be measured, for example, by high performance liquid chromatography.
  • Another example of effective dose determination for an individual is the ability to directly assay levels of "free" and "bound” compound in the serum of the test subject.
  • Such assays may utilize antibody mimics and/or "biosensors” that have been created through molecular imprinting techniques.
  • Molecules that modulate target molecule activity are used as a template, or "imprinting molecule”, to spatially organize polymerizable monomers prior to their polymerization with catalytic reagents. The subsequent removal of the imprinted molecule leaves a polymer matrix which contains a repeated "negative image” of the compound and is able to selectively rebind the molecule under biological assay conditions.
  • Such "imprinted" affinity matrixes can also be designed to include fluorescent groups whose photon-emitting properties measurably change upon local and selective binding of target compound. These changes readily can be assayed in real time using appropriate fiberoptic devices, in turn allowing the dose in a test subject to be quickly optimized based on its individual IC 50 .
  • An example of such a "biosensor” is discussed in Kriz et al, Analytical Chemistry 67: 2142-2144 (1995).
  • CETP, PROL4, GRID2, PDE4D and GPX3 nucleic acids or polypeptides and variants thereof are utilized for screening test molecules for those that interact with CETP, PROL4, GRID2, PDE4D and GPX3 molecules.
  • Test molecules identified as being interactors with target polypeptides can be screened further as osteoporosis therapeutics.
  • Blood samples were collected from individuals with low BMD, which were referred to as case samples. Also, blood samples were collected from individuals with high BMD (i.e., not diagnosed with osteoporosis or any form of osteoporosis-related disease); these samples served as gender and age- matched controls. All of the samples were of Caucasian (English) descent. A database was created that listed all phenotypic trait information gathered from individuals for each case and control sample. Genomic DNA was extracted from each of the blood samples for genetic analyses.
  • the solution was incubated at 37°C or room temperature if cell clumps were visible after mixing until the solution was homogeneous.
  • 2 ml of protein precipitation was added to the cell lysate.
  • the mixtures were vortexed vigorously at high speed for 20 sec to mix the protein precipitation solution uniformly with the cell lysate, and then centrifuged for 10 minutes at 3000 x g.
  • the supernatant containing the DNA was then poured into a clean 15 ml tube, which contained 7 ml of 100% isopropanol.
  • the samples were mixed by inverting the tubes gently until white threads of DNA were visible.
  • DNA was quantified by placing samples on a hematology mixer for at least 1 hour.
  • DNA was serially diluted (typically 1 :80, 1 :160, 1 :320, and 1 :640 dilutions) so that it would be within the measurable range of standards.
  • 125 ⁇ l of diluted DNA was transferred to a clear U-bottom microtitre plate, and 125 ⁇ l of IX TE buffer was transferred into each well using a multichannel pipette.
  • the DNA and IX TE were mixed by repeated pipetting at least 15 times, and then the plates were sealed. 50 ⁇ l of diluted DNA was added to wells A5-H12 of a black flat bottom microtitre plate.
  • the plate was placed into a Fluoroskan Ascent Machine (microplate fluorometer produced by Labsystems) and the samples were allowed to incubate for 3 minutes before the machine was run using filter pairs 485 nm excitation and 538 nm emission wavelengths. Samples having measured DNA concentrations of greater than 450 ng/ ⁇ l were re-measured for conformation. Samples having measured DNA concentrations of 20 ng/ ⁇ l or less were re-measured for confirmation. Pooling Strategies
  • Samples were placed into one of two groups based on BMD levels.
  • the two groups were made up of individuals with low BMD levels and individuals with high BMD levels.
  • a select set of samples from each group were utilized to generate pools, and one pool was created for each group.
  • Each individual sample in a pool was represented by an equal amount of genomic DNA. For example, where 25 ng of genomic DNA was utilized in each PCR reaction and there were 200 individuals in each pool, each individual would provide 125 pg of genomic DNA.
  • samples for a pool were based upon the following criteria: the sample was derived from an individual of Caucasian paternal and maternal descent; the database included relevant phenotype information for the individual; case samples were derived from individuals with low BMD; control samples were derived from individuals with normal or high BMD and no history of osteoporosis or osteoporosis-related diseases; and sufficient genomic DNA was extracted from each blood sample for all allelotyping and genotyping reactions performed during the study.
  • Phenotype information included pre- or post-menopausal, familial predisposition, country or origin of mother and father, diagnosis with osteoporosis (date of primary diagnosis, age of individual as of primary diagnosis, osteoporosis-related fracture), biochemical measurements of markers of bone resorption (bone-specific alkaline Phosphatase, Urinary C-telopeptide of type I collagen, serum osteocalcin), current medication status (thyroid medication, hormone replacement therapy, steroid usage, bisphosphonates and cytotoxic agents for rheumatic diseases). Samples that met the inclusion criteria and did not meet the exclusion criteria were added to appropriate pools based on gender and disease status.
  • the selection process yielded the pools set forth in Table 2, which were used in the studies that follow.
  • the average (mean) T-score in the High group is 1.56, and in the Low group -2.1 which means that on average individuals in the high BMD pool are 1.56 standard deviations above the average BMD in young females, whereas individuals in the low BMD pool are on average 2.1 standard deviations below the normal young value.
  • a whole-genome screen was performed to identify particular SNPs associated with low BMD.
  • two groups of samples were utilized, which included samples from female individuals having low BMD (osteoporosis cases), and samples from female individuals having high BMD levels (controls).
  • the initial screen of each pool was performed in an allelotyping study, in which certain samples in each group were pooled. By pooling DNA from each group, an allele frequency for each SNP in each group was calculated. These allele frequencies were then compared to one another. Particular SNPs were considered as being associated with low BMD when allele frequency differences calculated between case and control pools were statistically significant. SNP disease association results obtained from the allelotyping study were then validated by genotyping each associated SNP across all samples from each pool.
  • results of the genotyping were then analyzed, allele frequencies for each group were calculated from the individual genotyping results, and a p-value was calculated to determine whether the case and control groups had statistically significantly differences in allele frequencies for a particular SNP.
  • genotyping results agreed with the original allelotyping results, the SNP disease association was considered validated at the genetic level.
  • a whole-genome SNP screen began with an initial screen of approximately 25,000 SNPs over each set of disease and control samples using a pooling approach. The pools studied in the screen are described in Example 1.
  • the SNPs analyzed in this study were part of a set of 25,488 SNPs confirmed as being statistically polymorphic as each is characterized as having a minor allele frequency of greater than 10%.
  • the SNPs in the set reside in genes or in close proximity to genes, and many reside in gene exons. Specifically, SNPs in the set are located in exons, introns, and within 5,000 base- pairs upstream of a transcription start site of a gene.
  • SNPs were selected according to the following criteria: they are located in ESTs; they are located in Locuslink or Ensembl genes; and they are located in Genomatix promoter predictions. SNPs in the set were also selected on the basis of even spacing across the genome, as depicted in Table 3. An additional 3088 SNPs were included with these 25,488 SNPs and these additional SNPs had been chosen on the basis of gene location, with preference to non-synonymous coding SNPs located in disease candidate genes.
  • allelic variants associated with low BMD The allelic variants identified from the SNP panel described in Table 3 are summarized below in Table 4.
  • Table 4 includes information pertaining to the incident polymorphic variant associated with low BMD identified herein. Public information pertaining to the polymorphism and the genomic sequence that includes the polymorphism are indicated. The genomic sequences identified in Table 4 may be accessed at the http address www.ncbi.nih.gov/entrez/query.fcgi, for example, by using the publicly available SNP reference number (e.g., rs 1801706).
  • the "Contig Position” provided in Table 4 corresponds to a nucleotide position set forth in the contig sequence, and designates the polymorphic site corresponding to the SNP reference number.
  • the sequence containing the polymorphisms also may be referenced by the "Sequence Identification" set forth in Table 4.
  • the "Sequence Identification” corresponds to cDNA sequence that encodes associated target polypeptides (e.g., CETP) of the invention.
  • the position of the SNP within the cDNA sequence is provided in the "Sequence Position" column of Table 4. Also, the allelic variation at the polymorphic site and the allelic variant identified as associated with low BMD is specified in Table 4. All nucleotide sequences referenced and accessed by the parameters set forth in Table 4 are incorporated herein by reference.
  • a MassARRAY® system (Sequenom, Inc.) was utilized to perform SNP genotyping in a high-throughput fashion. This genotyping platform was complemented by a homogeneous, single-tube assay method (liMETM or homogeneous MassEXTENDTM (Sequenom, Inc.)) in which two genotyping primers anneal to and amplify a genomic target surrounding a polymorphic site of interest. A third primer (the MassEXTENDTM primer), which is complementary to the amplified target up to but not including the polymorphism, was then enzymatically extended one or a few bases through the polymorphic site and then terminated.
  • SpectroDESIGNERTM software (Sequenom, Inc.) was used to generate a set of PCR primers and a MassEXTENDTM primer was used to genotype the polymorphism.
  • Table 5 shows PCR primers and Table 6 shows extension primers used for analyzing polymorphisms.
  • the initial PCR amplification reaction was performed in a 5 ⁇ l total volume containing IX PCR buffer with 1.5 mM MgCl 2 (Qiagen), 200 ⁇ M each of dATP, dGTP, dCTP, dTTP (Gibco-BRL), 2.5 ng of genomic DNA, 0.1 units of HotStar DNA polymerase (Qiagen), and 200 nM each of forward and reverse PCR primers specific for the polymorphic region of interest.
  • a primer extension reaction was initiated by adding a polymorphism-specific MassEXTENDTM primer cocktail to each sample.
  • Each MassEXTENDTM cocktail included a specific combination of dideoxynucleotides (ddNTPs) and deoxynucleotides (dNTPs) used to distinguish polymorphic alleles from one another.
  • ddNTPs dideoxynucleotides
  • dNTPs deoxynucleotides
  • the MassEXTENDTM reaction was performed in a total volume of 9 ⁇ l, with the addition of IX ThermoSequenase buffer, 0.576 units of ThermoSequenase (Amersham Pharmacia), 600 nM MassEXTENDTM primer, 2 mM of ddATP and/or ddCTP and/or ddGTP and/or ddTTP, and 2 mM of dATP or dCTP or dGTP or dTTP.
  • the deoxy nucleotide (dNTP) used in the assay normally was complementary to the nucleotide at the polymorphic site in the amplicon. Samples were incubated at 94°C for 2 minutes, followed by 55 cycles of 5 seconds at 94 0 C, 5 seconds at 52°C, and 5 seconds at 72 0 C.
  • samples were desalted by adding 16 ⁇ l of water (total reaction volume was 25 ⁇ l), 3 mg of SpectroCLEANTM sample cleaning beads (Sequenom, Inc.) and allowed to incubate for 3 minutes with rotation. Samples were then robotically dispensed using a piezoelectric dispensing device (SpectroJETTM (Sequenom, Inc.)) onto either 96-spot or 384-spot silicon chips containing a matrix that crystallized each sample (SpectroCHIP ® (Sequenom, Inc.)).
  • MALDI-TOF mass spectrometry (Biflex and Autoflex MALDI-TOF mass spectrometers (Bruker Daltonics) can be used) and SpectroTYPER RTTM software (Sequenom, Inc.) were used to analyze and interpret the SNP genotype for each sample.
  • Variations identified in the target genes are provided in their respective genomic sequences (see SEQ ID NOs: 1-4) Minor allelic frequencies for these polymorphisms was verified as being 10% or greater by determining the allelic frequencies using the extension assay described above in a group of samples isolated from 92 individuals originating from the state of Utah in the United States, Venezuela and France (Coriell cell repositories).
  • Odds ratio results are shown in Table 7 (and other Tables below).
  • An odds ratio is an unbiased estimate of relative risk which can be obtained from most case-control studies.
  • Relative risk (RR) is an estimate of the likelihood of disease in the exposed group (susceptibility allele or genotype carriers) compared to the unexposed group (not carriers). It can be calculated by the following equation:
  • IA is the incidence of disease in the A carriers and /a is the incidence of disease in the non-carriers.
  • RR > 1 indicates the A allele increases disease susceptibility.
  • RR ⁇ 1 indicates the a allele increases disease susceptibility.
  • An odds ratio can be interpreted in the same way a relative risk is interpreted and can be directly estimated using the data from case-control studies, i.e., case and control allele frequencies.
  • the higher the odds ratio value the larger the effect that particular allele has on the development of low BMD. Possessing an allele associated with a relatively high odds ratio translates to having a higher risk of developing or having low BMD.
  • the "genome letter” corresponds to the particular allele that appears in NCBI's build 34 genomic sequence of the region (chromosome 16: positions 56743155-56840953), and the "deduced iupac” corresponds to the single letter IUPAC code for the CTEP polymorphic variants as they appear in SEQ ID NO:1. Also, the “genome letter” may differ from the alleles (A1/A2) provided in Table 8 (and in subsequent Tables that provide the same information) if the genome letter is on one strand and the alleles are on the complementary strand, thus having different strand orientations (i.e., reverse vs forward).
  • allelotyping results were considered particularly significant with a calculated p-value of less than or equal to 0.05 for allelotype results. These values are indicated in bold.
  • the allelotyping p- values were plotted in Figure 1.
  • the position of each SNP on the chromosome is presented on the x- axis.
  • the y-axis gives the negative logarithm (base 10) of the p-value comparing the estimated allele in the case group to that of the control group.
  • the minor allele frequency of the control group for each SNP designated by an X or other symbol on the graphs in Figure 1 can be determined by consulting Table 11. For example, the left-most X on the left graph is at position 56743155. By proceeding down the Table from top to bottom and across the graphs from left to right the allele frequency associated with each symbol shown can be determined.
  • the broken horizontal lines are drawn at two common significance levels, 0.05 and 0.01.
  • the vertical broken lines are drawn every 20kb to assist in the interpretation of distances between SNPs.
  • Two other lines are drawn to expose linear trends in the association of SNPs to the disease.
  • the light gray line (or generally bottom-most curve) is a nonlinear smoother through the data points on the graph using a local polynomial regression method (W.S. Cleveland, E. Grosse and W.M. Shyu (1992) Local regression models. Chapter 8 of Statistical Models in S eds J.M. Chambers and TJ. Hastie, Wadsworth & Brooks/Cole.).
  • the black line provides a local test for excess statistical significance to identify regions of association. This was created by use of a 10kb sliding window with lkb step sizes. Within each window, a chi-square goodness of fit test was applied to compare the proportion of SNPs that were significant at a test wise level of 0.01, to the proportion that would be expected by chance alone (0.05 for the methods used here). Resulting p-values that were less than 10 "8 were truncated at that value.
  • allelotyping results were considered particularly significant with a calculated p-value of less than or equal to 0.05 for allelotype results. These values are indicated in bold.
  • the allelotyping p- values were plotted in Figure 2.
  • the position of each SNP on the chromosome is presented on the x- axis.
  • the y-axis gives the negative logarithm (base 10) of the p-value comparing the estimated allele in the case group to that of the control group.
  • the minor allele frequency of the control group for each SNP designated by an X or other symbol on the graphs in Figure 2 can be determined by consulting Table 15. For example, the left-most X on the left graph is at position 10842129. By proceeding down the Table from top to bottom and across the graphs from left to right the allele frequency associated with each symbol shown can be determined.
  • the broken horizontal lines are drawn at two common significance levels, 0.05 and 0.01.
  • the vertical broken lines are drawn every 20kb to assist in the interpretation of distances between SNPs.
  • Two other lines are drawn to expose linear trends in the association of SNPs to the disease.
  • the light gray line (or generally bottom-most curve) is a nonlinear smoother through the data points on the graph using a local polynomial regression method (W.S. Cleveland, E. Grosse and W.M. Shyu (1992) Local regression models. Chapter 8 of Statistical Models in S eds J.M. Chambers and TJ. Hastie, Wadsworth & Brooks/Cole.).
  • the black line provides a local test for excess statistical significance to identify regions of association. This was created by use of a 10kb sliding window with lkb step sizes. Within each window, a chi-square goodness of fit test was applied to compare the proportion of SNPs that were significant at a test wise level of 0.01, to the proportion that would be expected by chance alone (0.05 for the methods used here). Resulting p-values that were less than 10 "8 were truncated at that value.
  • allelotyping results were considered particularly significant with a calculated p-value of less than or equal to 0.05 for allelotype results. These values are indicated in bold.
  • the allelotyping p- values were plotted in Figure 3.
  • the position of each SNP on the chromosome is presented on the x- axis.
  • the y-axis gives the negative logarithm (base 10) of the p-value comparing the estimated allele in the case group to that of the control group.
  • the minor allele frequency of the control group for each SNP designated by an X or other symbol on the graphs in Figure 3 can be determined by consulting Table 19. For example, the left-most X on the left graph is at position 44917643. By proceeding down the Table from top to bottom and across the graphs from left to right the allele frequency associated with each symbol shown can be determined.
  • the broken horizontal lines are drawn at two common significance levels, 0.05 and 0.01.
  • the vertical broken lines are drawn every 20kb to assist in the interpretation of distances between SNPs.
  • Two other lines are drawn to expose linear trends in the association of SNPs to the disease.
  • the light gray line (or generally bottom-most curve) is a nonlinear smoother through the data points on the graph using a local polynomial regression method (W.S. Cleveland, E. Grosse and W.M. Shyu (1992) Local regression models. Chapter 8 of Statistical Models in S eds J.M. Chambers and TJ. Hastie, Wadsworth & Brooks/Cole.).
  • the black line provides a local test for excess statistical significance to identify regions of association. This was created by use of a 10kb sliding window with lkb step sizes. Within each window, a chi-square goodness of fit test was applied to compare the proportion of SNPs that were significant at a test wise level of 0.01, to the proportion that would be expected by chance alone (0.05 for the methods used here). Resulting p-values that were less than 10 "s were truncated at that value. [0245] Finally, the exons and introns of the genes in the covered region are plotted below each graph at the appropriate chromosomal positions. The gene boundary is indicated by the broken horizontal line. The exon positions are shown as thick, unbroken bars. An arrow is place at the 3 ' end of each gene to show the direction of transcription.
  • allelotyping results were considered particularly significant with a calculated p-value of less than or equal to 0.05 for allelotype results. These values are indicated in bold.
  • the allelotyping p- values were plotted in Figure 4. The position of each SNP on the chromosome is presented on the x- axis. The y-axis gives the negative logarithm (base 10) of the p-value comparing the estimated allele in the case group to that of the control group.
  • the minor allele frequency of the control group for each SNP designated by an X or other symbol on the graphs in Figure 4 can be determined by consulting Table 23. For example, the left-most X on the left graph is at position 58309549. By proceeding down the Table from top to bottom and across the graphs from left to right the allele frequency associated with each symbol shown can be determined.
  • the broken horizontal lines are drawn at two common significance levels, 0.05 and 0.01.
  • the vertical broken lines are drawn every 20kb to assist in the interpretation of distances between SNPs.
  • Two other lines are drawn to expose linear trends in the association of SNPs to the disease.
  • the light gray line (or generally bottom-most curve) is a nonlinear smoother through the data points on the graph using a local polynomial regression method (W.S. Cleveland, E. Grosse and W.M. Shyu (1992) Local regression models. Chapter 8 of Statistical Models in S eds J.M. Chambers and TJ. Hastie, Wadsworth & Brooks/Cole.).
  • the black line provides a local test for excess statistical significance to identify regions of association. This was created by use of a 10kb sliding window with lkb step sizes. Within each window, a chi-square goodness of fit test was applied to compare the proportion of SNPs that were significant at a test wise level of 0.01, to the proportion that would be expected by chance alone (0.05 for the methods used here). Resulting p-values that were less than 10 "8 were truncated at that value.
  • allelotyping results were considered particularly significant with a calculated p-value of less than or equal to 0.05 for allelotype results. These values are indicated in bold.
  • the allelotyping p- values were plotted in Figure 4. The position of each SNP on the chromosome is presented on the x- axis. The y-axis gives the negative logarithm (base 10) of the p-value comparing the estimated allele in the case group to that of the control group.
  • the minor allele frequency of the control group for each SNP designated by an X or other symbol on the graphs in Figure 4 can be determined by consulting Table 27. For example, the left-most X on the left graph is at position 150385031. By proceeding down the Table from top to bottom and across the graphs from left to right the allele frequency associated with each symbol shown can be determined.
  • the broken horizontal lines are drawn at two common significance levels, 0.05 and 0.01.
  • the vertical broken lines are drawn every 20kb to assist in the interpretation of distances between SNPs.
  • Two other lines are drawn to expose linear trends in the association of SNPs to the disease.
  • the light gray line (or generally bottom-most curve) is a nonlinear smoother through the data points on the graph using a local polynomial regression method (W.S. Cleveland, E. Grosse and W.M. Shyu (1992) Local regression models. Chapter 8 of Statistical Models in S eds J.M. Chambers and TJ. Hastie, Wadsworth & Brooks/Cole.).
  • the black line provides a local test for excess statistical significance to identify regions of association. This was created by use of a 10kb sliding window with lkb step sizes. Within each window, a chi-square goodness of fit test was applied to compare the proportion of SNPs that were significant at a test wise level of 0.01, to the proportion that would be expected by chance alone (0.05 for the methods used here). Resulting p-values that were less than 10 "8 were truncated at that value. [0257] Finally, the exons and introns of the genes in the covered region are plotted below each graph at the appropriate chromosomal positions. The gene boundary is indicated by the broken horizontal line. The exon positions are shown as thick, unbroken bars. An arrow is place at the 3' end of each gene to show the direction of transcription.
  • cDNA is cloned into a pIVEX 2.3-MCS vector (Roche Biochem) using a directional cloning method.
  • a cDNA insert is prepared using PCR with forward and reverse primers having 5' restriction site tags (in frame) and 5-6 additional nucleotides in addition to 3' gene-specific portions, the latter of which is typically about twenty to about twenty-five base pairs in length.
  • a Sal I restriction site is introduced by the forward primer and a Sma I restriction site is introduced by the reverse primer.
  • the ends of PCR products are cut with the corresponding restriction enzymes ⁇ i.e., Sal I and Sma I) and the products are gel-purified.
  • the pIVEX 2.3-MCS vector is linearized using the same restriction enzymes, and the fragment with the correct sized fragment is isolated by gel- purification. Purified PCR product is ligated into the linearized pIVEX 2.3-MCS vector and E. coli cells transformed for plasmid amplification. The newly constructed expression vector is verified by restriction mapping and used for protein production.
  • E. coli lysate is reconstituted with 0.25 ml of Reconstitution Buffer, the Reaction Mix is reconstituted with 0.8 ml of Reconstitution Buffer; the Feeding Mix is reconstituted with 10.5 ml of Reconstitution Buffer; and the Energy Mix is reconstituted with 0.6 ml of Reconstitution Buffer.
  • 0.5 ml of the Energy Mix was added to the Feeding Mix to obtain the Feeding Solution.
  • 0.75 ml of Reaction Mix, 50 ⁇ l of Energy Mix, and 10 ⁇ g of the template DNA is added to the E. coli lysate.
  • the reaction device (Roche Biochem) 1 ml of the Reaction Solution is loaded into the reaction compartment.
  • the reaction device is turned upside-down and 10 ml of the Feeding Solution is loaded into the feeding compartment. All lids are closed and the reaction device is loaded into the RTS500 instrument. The instrument is run at 3O 0 C for 24 hours with a stir bar speed of 150 rpm.
  • the pTVEX 2.3 MCS vector includes a nucleotide sequence that encodes six consecutive histidine amino acids on the C-terminal end of the target polypeptide for the purpose of protein purification.
  • Target polypeptide is purified by contacting the contents of reaction device with resin modified with Ni 2+ ions.
  • Target polypeptide is eluted from the resin with a solution containing free Ni 2+ ions.
  • Nucleic acids are cloned into DNA plasmids having phage recombination cites and target polypeptides are expressed therefrom in a variety of host cells.
  • Alpha phage genomic DNA contains short sequences known as attP sites
  • E. coli genomic DNA contains unique, short sequences known as attB sites. These regions share homology, allowing for integration of phage DNA into E. coli via directional, site-specific recombination using the phage protein Int and the E. coli protein IHF. Integration produces two new att sites, L and R, which flank the inserted prophage DNA.
  • Phage excision from E. coli genomic DNA can also be accomplished using these two proteins with the addition of a second phage protein, Xis.
  • DNA vectors have been produced where the integration/excision process is modified to allow for the directional integration or excision of a target
  • a first step is to transfer the nucleic acid insert into a shuttle vector that contains attL sites surrounding the negative selection gene, ccdB (e.g. pENTER vector, Invitrogen, Inc.). This transfer process is accomplished by digesting the nucleic acid from a DNA vector used for sequencing, and to ligate it into the multicloning site of the shuttle vector, which will place it between the two attL sites while removing the negative selection gene ccdB.
  • a second method is to amplify the nucleic acid by the polymerase chain reaction (PCR) with primers containing attB sites. The amplified fragment then is integrated into the shuttle vector using Int and IHF.
  • PCR polymerase chain reaction
  • a third method is to utilize a topoisomerase- mediated process, in which the nucleic acid is amplified via PCR using gene-specific primers with the
  • 5' upstream primer containing an additional CACC sequence e.g., TOPO ® expression kit (Invitrogen, Inc.)
  • TOPO ® expression kit Invitrogen, Inc.
  • the PCR amplified fragment can be cloned into the shuttle vector via the attL sites in the correct orientation.
  • the nucleic acid can be cloned into an expression vector having attR sites.
  • Several vectors containing attR sites for expression of target polypeptide as a native polypeptide, N-fusion polypeptide, and C-fusion polypeptides are commercially available (e.g., pDEST (Invitrogen, Inc.)), and any vector can be converted into an expression vector for receiving a nucleic acid from the shuttle vector by introducing an insert having an attR site flanked by an antibiotic resistant gene for selection using the standard methods described above. Transfer of the nucleic acid from the shuttle vector is accomplished by directional recombination using Int, IHF, and Xis (LR clonase).
  • the desired sequence can be transferred to an expression vector by carrying out a one hour incubation at room temperature with Int, IHF, and Xis, a ten minute incubation at 37 0 C with proteinase K, transforming bacteria and allowing expression for one hour, and then plating on selective media. Generally, 90% cloning efficiency is achieved by this method.
  • expression vectors are pDEST 14 bacterial expression vector with att7 promoter, pDEST 15 bacterial expression vector with a T7 promoter and a N-terminal GST tag, pDEST 17 bacterial vector with a T7 promoter and a N-terminal polyhistidine affinity tag, and pDEST 12.2 mammalian expression vector with a CMV promoter and neo resistance gene. These expression vectors or others like them are transformed or transfected into cells for expression of the target polypeptide or polypeptide variants. These expression vectors are often transfected, for example, into murine-transformed a adipocyte cell line 3T3-L1, (ATCC), human embryonic kidney cell line 293, and rat cardiomyocyte cell line H9C2.
  • genomic nucleotide sequence for a CTEP region.
  • the genomic nucleotide sequence is set forth in SEQ ID NO: 1.
  • the following nucleotide representations are used throughout: "A” or “a” is adenosine, adenine, or adenylic acid; “C” or “c” is cytidine, cytosine, or cytidylic acid; “G” or “g” is guanosine, guanine, or guanylic acid; “T” or “t” is thymidine, thymine, or thymidylic acid; and “I” or “i” is inosine, hypoxanthine, or inosinic acid.
  • SNPs are designated by the following convention: “R” represents A or G, “M” represents A or C; “W” represents A or T; “Y” represents C or T; “S” represents C or G; “K” represents G or T; "V” represents A, C or G; “H” represents A, C, or T; “D” represents A, G, or T; "B” represents C, G, or T; and "N” represents A, G, C, or T.
  • 25261 atatgctatt ggttaatccc gtagtaatgc ttaacttcac tagtaatcaa atatatgtga 25321 agtaaagata tttttgcaat caaagtagtg acattaaaaa taataacctg tttgattttg 25381 agactgcaac aggacggata tttttcact attaacatat aggtttatta tatctacaga 25441 gcaatttatc aattctgcaa ggaatttatc aagtttctcc ccattggaat tttccttaaa 25501 aataatttac atactgctgt ctctatatat tagtcacact ggacctttag ttctggact 25561 accctt

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Analytical Chemistry (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention a trait à des procédés pour l'identification de risque de faible densité minérale osseuse chez un sujet, à des réactifs et des trousses pour la mise en oeuvre des procédés, à des procédés pour l'identification d'agents thérapeutiques candidats pour le traitement de troubles liés à la faible densité minérale osseuse, tels que l'ostéoporose, et à des procédés thérapeutiques et prophylactiques applicables à l'ostéoporose. Ces modes de réalisations reposent sur une analyse de variations polymorphes dans des séquences nucléotidiques dans le génome humain.
PCT/US2005/047411 2004-12-31 2005-12-29 Procede pour l'identification de risque de faible densite minerale osseuse et traitements correspondants WO2006074061A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/578,392 US20070292849A1 (en) 2004-12-31 2005-12-29 Methods for Identifying Risk of Low Bmd and Treatments Thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US64096604P 2004-12-31 2004-12-31
US60/640,966 2004-12-31

Publications (2)

Publication Number Publication Date
WO2006074061A2 true WO2006074061A2 (fr) 2006-07-13
WO2006074061A3 WO2006074061A3 (fr) 2009-06-04

Family

ID=36648061

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/047411 WO2006074061A2 (fr) 2004-12-31 2005-12-29 Procede pour l'identification de risque de faible densite minerale osseuse et traitements correspondants

Country Status (1)

Country Link
WO (1) WO2006074061A2 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024102688A1 (fr) * 2022-11-07 2024-05-16 New York Society For The Relief Of The Ruptured And Crippled, Maintaining The Hospital For Special Surgery Compositions pour le traitement de troubles de l'ostéoclastogenèse et/ou de la polyarthrite rhumatoïde

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE NCBI 03 July 2003 Database accession no. rs7500979 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024102688A1 (fr) * 2022-11-07 2024-05-16 New York Society For The Relief Of The Ruptured And Crippled, Maintaining The Hospital For Special Surgery Compositions pour le traitement de troubles de l'ostéoclastogenèse et/ou de la polyarthrite rhumatoïde

Also Published As

Publication number Publication date
WO2006074061A3 (fr) 2009-06-04

Similar Documents

Publication Publication Date Title
AU2017267184B2 (en) Method for assessing a prognosis and predicting the response of patients with malignant diseases to immunotherapy
US6605432B1 (en) High-throughput methods for detecting DNA methylation
KR101708544B1 (ko) 세포 증식 질환을 분석하기 위한 방법 및 핵산
KR102046668B1 (ko) 암 대상자의 예후를 결정하기 위한 방법 및 핵산
US20230056182A1 (en) Use of adeno-associated viral vectors to correct gene defects/ express proteins in hair cells and supporting cells in the inner ear
CN107223159A (zh) 源自特定细胞类型的dna的检测及相关方法
US20090305284A1 (en) Methods for Identifying Risk of Breast Cancer and Treatments Thereof
AU2016351889A1 (en) Detection of foetal chromosomal aneuploidies using DNA regions that are differentially methylated between the foetus and the pregnant female
CA2936612A1 (fr) Polymorphismes atf6 associes a une infarctus du myocarde, methode de detection et utilisations associees
AU779411B2 (en) Biallelic markers derived from genomic regions carrying genes involved in arachidonic acid metabolism
KR20120034651A (ko) 전립선암 마커로서의 포스포디에스테라제 4d7
KR20150092739A (ko) 예측변수 인자들을 이용하여 동정된 환자 부분모집단에서 암 치료를 위한 마시티닙의 용도
KR20220025749A (ko) 대장암의 검출
KR20060045950A (ko) 혈액학적 악성종양에 대한 예후
KR20220024184A (ko) 대장암의 검출
CN108026587A (zh) 用于治疗癌症的新颖生物标记和方法
KR20130123357A (ko) 저산소증과 관련된 질환의 진단방법 및 키트
EP1729930A2 (fr) Procedes d'identification de risque d'osteoarthrite et traitements correspondants
WO2006022629A1 (fr) Procédés d’identification de risque de diabète de type ii et leurs traitements
AU2018360287B2 (en) Method for determining the response of a malignant disease to an immunotherapy
US6797475B2 (en) Detection of polymorphisms in the human 5-lipoxygenase gene
WO2006022636A1 (fr) Méthodes d’identification du risque d’apparition de diabètes de type ii et traitements associés
WO2006022634A1 (fr) Méthodes d’identification du risque d’apparition de diabètes de type ii et traitements associés
US20070292849A1 (en) Methods for Identifying Risk of Low Bmd and Treatments Thereof
KR101474053B1 (ko) 골다공증 또는 골다공성 골절 발생 위험도 예측용 다형성 마커

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 10578392

Country of ref document: US

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 05857249

Country of ref document: EP

Kind code of ref document: A2