专利摘要:
The present invention relates to the genomic DNA, cDNA, polypeptide sequence of CanIon, a voltage-gated ion channel protein. The present invention also relates to double allele markers of the CanIon gene. The CanIon gene can be used as a biological target for the treatment and diagnosis of schizophrenia, bipolar disorder, and other diseases and disorders.
公开号:KR20030074643A
申请号:KR10-2003-7007511
申请日:2001-12-04
公开日:2003-09-19
发明作者:다니엘 코헨;일리아 추마코브;안네-마리아 시몬;하우디 아브데라히
申请人:쟝세;
IPC主号:
专利说明:

Schizophrenia-associated voltage-gated ion channel genes and proteins {SCHIZOPHRENIA RELATED VOLTAGE-GATED ION CHANNEL GENE AND PROTEIN}
[2] Advances in the technological facilities available to clinical speakers allow for the sophisticated study of brain and nervous system function in health and disease conditions. Many neurological and pharmacological hypotheses have been proposed in terms of neurochemical and genetic mechanisms involved in central nervous system (CNS) diseases, including mental and neurodegenerative diseases. However, CNS disease presents a complex and incomprehensible etiology and overlapping, rarely characterized and difficult to measure symptoms. As a result, future treatment regimens and drug development efforts need to focus on more sophisticated and multiple genetic causes, require new assays to identify disease populations, and provide more accurate diagnosis and prognostic information for patients with CNS disease. Must be provided.
[3] CNS diseases can be accompanied by a wide range of diseases and corresponding broad genetic factors. Examples of CNS diseases include neurodegenerative disorders, mental disorders, mood disorders, autism, drug dependence and alcoholism, mental retardation and cognitive disorders, mental disorders, anxiety disorders, eating disorders, impulse control disorders, personality disorders, including personality disorders. Included. Diseases can be defined according to the Diagnosis Statistical Manual of Mental Disorder fourth edition (DSM-IV) classification.
[4] Even when considering a small subset of CNS diseases, it is clear that there is a need for new targets and improved methods of treatment of the invention due to the lack of adequate treatment and understanding of the molecular basis for schizophrenia and bipolar disorder. All currently known molecules used in the treatment of schizophrenia and bipolar disorder have side effects and their effects are limited to their symptoms. There is an urgent need for new molecules that inhibit targets involved in the etiology of schizophrenia and bipolar disorder without the associated side effects. Thus, tools that facilitate the discovery and characterization of these targets are useful.
[5] Voltage-gated ion channel
[6] Voltage-gated ion channels are a macromolecule of macromolecules that perform several functions, including regulation and maintenance of potential, secretion, and signal transduction in cell membranes. These channel proteins are involved in the regulation of neurotransmitters from neurons and play an important role in the regulation of various cellular functions including membrane excitability, muscle contraction, synaptic transmission. The major alpha-subunit of the Na + channel and the alpha-1 subunit of the Ca + channel consist of approximately 2000 amino acids and possess ionic conduction pathways. Biochemical analysis has shown that the bioactive ion channel consists of several different subunits. There are two auxiliary subunits, beta-1 and beta-2 subunits, which assist the alpha subunit of the Na + channel. Additional subunits (alpha-2, beta, gamma, sigma) in the Ca + channel were found to have regulatory action. Alpha-2 and beta-subunits appear to enhance the functional activity of Ca + alpha-1 subunits. The alpha-subunit of the K + channel binds to the beta subunit in a 1: 1 manner to induce a K + channel complex with (alpha) 4 (beta) 4 stereochemical properties (Terlau et al., Naturwissenschaften 85: 437-444 (1998). Basic structures and examples of calcium and sodium ion channels are described, for example, in Williams, et al. Science 257: 389-395 (1992); Mori, et al., Nature 350: 398-402 (1991); Koch, et al., J. Biol. Chem. 265 (29): 17786-17791 (1990). Ca + and Na + ion channel nucleic acid sequences that share high levels of sequence homology with CanIon channels and are derived from mice are described in Lee et al., FEBS Lett. 445 231: 236 (1999).
[7] Alpha subunits share sequence characteristics with all voltage-dependent cation channels and utilize the same structural motifs that comprise a six-helix bundle of translocation membrane spanning domains. In both sodium and calcium channels, the motif is repeated four times in the sequence and provides a 24-helix bundle. Amino acid sequences are highly conserved between species (eg, humans and fruit flies) and between different ion channels.
[8] There are several tissue-specific pharmacological and electrophysiologically distinct allotropes of calcium channels encoded by individual genes in the multi-gene family. In skeletal muscle, each tightly coupled assembly of alpha, beta, and gamma subunits combines with four other assemblies to form macromolecules of the pentomer. For example, the neuronal calcium channel alpha-1 subunit is the product of at least seven different genes (alpha 1 A to H). Immunocytochemical studies showed differential distribution of alpha-1 calcium channel subunits. Alpha-1A and alpha-1B are mainly expressed at the dendritic and semisynaptic ends, with alpha-1A concentrated at a greater number of nerve endings than alpha-1B. In rat and human neuromuscular junctions, alpha-1A is centrally located at the exhibition synapse, while alpha-1B and alpha-1A are present in axon-coupled Schwann cells. Alpha-1E is concentrated in the cell body and in some cases is located in the proximal dendritic processes and the terminal branches of Purkinje cells. Alpha-1C and alpha-1D are concentrated in the proximal dendritic processes of cellular bodies and central neurons.
[9] Intrinsic calcium channels were classified based on pharmacological and / or electrophysiological properties. Voltage-dependent calcium channels are divided into the following: high voltage-activated channels (HVA), including L-, N-, P-, Q-types; Intermediate channels (IVA, R-type); Low voltage-activated channels (LVA, T-type) (Morena et al., Annals NY Acad. Sci. 102-117 (1999)).
[10] The major subunit (alpha-1) belongs to a gene whose membranes can form their own functional channels when expressed in heterogeneous expression systems (Zhang et al., Neuropharmacology 32 (11): 1075-1088 (1993). In, alpha-1 subunits are expressed as multi-subunit complexes with auxiliary subunits that change the functional properties of the alpha-1 unit In many cases, coexpression of the auxiliary subunits is dependent on the biophysical properties of the channel. In particular, beta subunits have been found to significantly affect alpha-1 subunits; beta subunits have been found to alter activation properties, steady state deactivation, deactivation kinetics, and peak current.
[11] Most of the molecular diversity of the channels is caused by the presence of complex alpha-1 subunits. For example, differently cleaved forms of calcium channels are differentially expressed and possess unique susceptibility to phosphorylation by serine-threonine kinases (Hell et al., Annals NY Acad. Sci. 747: 282-293 (1994) ). Mutations in ion channel genes have been shown to be involved in a wide range of diseases, including several central nervous system diseases. Examples of ion channel mutations that cause various paroxysmal disorders, including periodic paralysis, paroxysmal ataxia, migraine, prolonged QT syndrome, and paroxysmal dyskinesia, are described in Bulman et al., Hum. Mol. Gen. 6 (10) 1679-1685 (1997). Examples of some Ca + channel mutant diseases are shown in Table A (Moreno, supra).
[12] Table A
[13] disease Calcium Channel Subunit Model Familial lateral paraplegic migraine Alpha-1A Person Paroxysmal ataxia type 2 Alpha-1A Person Spinal cerebellar ataxia type 6 Alpha-1A Person Tottering and Learner Phenotypes Alpha-1A mouse Lambert-Etonton syndrome Alpha-1A, α-1B Person Hypokalsic periodic paralysis Alpha-1S Person Muscle dysplasia Alpha-1S mouse Zucker Diabetic Hyperlipotype α-1C; α-1D rat Coma Beta-4 mouse Malignant hypothermia Alpha-2 / δ Person Stargazer gamma mouse
[14] Regulators of calcium and sodium channels are commonly used to treat a variety of diseases and conditions. For example, calcium and / or sodium channel blockers can be used to treat a variety of diseases or disorders, such as various heart diseases and disorders (eg, angina pectoris, arrhythmia), high blood pressure, migraine, neurological effects of seizures, manic, neuroleptic-induced delayed It has been found to be useful for treating or preventing one or a plurality of symptoms associated with movement disorders, bipolar disorders, pain, epilepsy and the like.
[15] Significant functional differences in the nervous system appear to exist between different ion channels. In addition, functional differences exist between different mutations in the same ion channel gene and between truncation variants of the same ion channel. Thus, despite the association of ion channels in CNS disease, it is difficult to predict which ion channel is an effective target for therapeutic intervention for a particular disease. One challenge is to provide ion channel genes that are involved in schizophrenia, bipolar disorder or diseases associated with them.
[16] Summary of the Invention
[17] The present invention is directed to a nucleic acid molecule comprising the genomic sequence of a novel human gene encoding a voltage-gated ion channel protein called CanIon. The CanIon genomic sequence includes regulatory sequences located upstream (5 'end) and downstream (3' end) of the transcription region of the gene, which regulatory sequences also belong to the present invention.
[18] The present invention also provides a complete cDNA sequence and corresponding translation product encoding the CanIon protein.
[19] Oligonucleotide probes and primers that hybridize specifically with the CanIon genome or cDNA sequences, and DNA amplification and detection methods using these primers and probes also belong to the present invention.
[20] Another object of the present invention is to introduce a recombinant vector comprising one of the above-described nucleic acid sequences, in particular a recombinant vector comprising a CanIon regulatory sequence or a sequence encoding a CanIon protein and a cell host and transgenic comprising the nucleic acid sequence or the recombinant vector. It is to provide animals except humans.
[21] The present invention also relates to dual allele markers of the CanIon gene and uses thereof.
[22] Finally, the present invention relates to a method for screening a substance or molecule that modulates the expression or activity of CanIon and a method for screening a substance or molecule that interacts with a CanIon polypeptide.
[23] Also provided are methods of using materials identified by these methods. For example, the use of CanIon channel antagonists provides a method for treating or preventing diseases or disorders, including schizophrenia or bipolar disorder.
[24] Accordingly, in one aspect the invention provides an isolated, purified or recombinant polynucleotide comprising one of the nucleotide sequences shown as SEQ ID NOs 1 to 4 or 6, or sequences complementary to these sequences.
[25] In another aspect, the present invention provides an isolated, purified or recombinant polynucleotide comprising a continuous span of at least 50 nucleotides of SEQ ID NO 4, wherein the polynucleotide encodes a biologically active CanIon polypeptide.
[26] In another aspect, the present invention provides an isolated, purified or recombinant polynucleotide encoding a human CanIon polypeptide comprising the amino acid sequence of SEQ ID NO 5 or a biologically active fragment thereof.
[27] In one embodiment, the polynucleotide described above is attached to a solid support. In another embodiment, the polynucleotide comprises a label.
[28] In another aspect, the invention provides an array of polynucleotides comprising at least one of the foregoing polynucleotides. In one embodiment, the array is addressable.
[29] In another aspect, the invention provides a recombinant vector operably linked to a promoter and comprising one of the aforementioned polynucleotides.
[30] In another aspect, the present invention provides a polynucleotide that alters the expression level of the CanIon gene in a cell. In one embodiment, the polynucleotide is inserted into a CanIon gene or CanIon genomic region. In one embodiment, the polynucleotide is inserted into a CanIon gene promoter. In one embodiment, the polynucleotide is inserted by substitution of one or more elements of homologous recombination, eg, an endogenous CanIon promoter or enhancer region.
[31] In another aspect, the invention provides a human exclusion host cell comprising one of the aforementioned recombinant vectors or polynucleotides.
[32] In another aspect, the present invention provides a mammalian host cell or human exclusion host mammal comprising a CanIon gene that is destroyed by homologous recombination with a knock out vector.
[33] In another aspect, the present invention provides an isolated, purified or recombinant polypeptide comprising the amino acid sequence shown in SEQ ID NO 5 or a biologically active fragment thereof.
[34] In another aspect, the invention provides a method of making a polypeptide, the method comprising: a) providing a cell population comprising a polynucleotide encoding a polypeptide according to the invention operably linked to a promoter; b) culturing the cell population under conditions conducive to the production of said polypeptide in cells; c) purifying the polypeptide from the cell population.
[35] In another aspect, the present invention provides a method of binding an anti-CanIon antibody to a CanIon polypeptide, the method comprising contacting the antibody with the aforementioned CanIon polypeptide under conditions such that the antibody and polypeptide can specifically bind. It is composed. In another aspect, provided are antibodies or immunologically active fragments thereof that specifically recognize CanIon proteins or epitopes.
[36] In another aspect, the present invention provides a method for detecting expression of a CanIon gene in a cell, the method comprising: a) i) a polynucleotide that hybridizes with the aforementioned CanIon polynucleotide under stringent conditions; Or ii) contacting the cell or cell extract with a polypeptide that specifically binds to one of the aforementioned CanIon polypeptides; b) identifying the presence of hybridization between polynucleotide and RNA species in said cell or cell extract, or the presence of polypeptide and protein binding in said cell or cell extract, wherein the presence of hybridization or binding is such that the CanIon gene is Imply that it is expressed within. In one embodiment, the polynucleotide is a primer and hybridization is confirmed by the presence of an amplification product comprising the sequence of said primer. In other embodiments, the polypeptide is an antibody, eg an anti-CanIon antibody.
[37] In another aspect, the present invention provides a method of identifying candidate modulators of a CanIon polypeptide, the method comprising: a) contacting a test compound with one of the aforementioned CanIon polypeptides; b) identifying whether said compound specifically binds said polypeptide, wherein identification of said compound specifically binding to said polypeptide suggests that said compound is a candidate modulator of a CanIon polypeptide.
[38] In one embodiment, the method further comprises measuring the bioactivity of the CanIon polypeptide in the presence of the candidate modulator, wherein the change in bioactivity of the CanIon polypeptide in the presence of the candidate modulator is compared to the activity in the absence of the candidate modulator. It suggests that the candidate modulator is a modulator of the CanIon polypeptide.
[39] In another aspect, the present invention provides a method for identifying a modulator of a CanIon polypeptide, the method comprising: a) contacting a test compound with one of the aforementioned CanIon polypeptides; b) identifying the activity of said polypeptide in the absence of said compound, wherein identification of the difference in activity in the presence of said compound compared to activity in the absence of said compound suggests that said compound is a modulator of a CanIon polypeptide do.
[40] In one embodiment of the invention, the polypeptide is present in the cell or cell membrane and the bioactivity is voltage-gated ion channel activity.
[41] In another aspect, the present invention provides a method of preparing a pharmaceutical composition, the method comprising: a) identifying a modulator of a CanIon polypeptide using one of the methods described above; b) mixing the modulator with a physiologically acceptable carrier. Methods of using the pharmaceutical compositions are also provided.
[42] The present invention also provides the use of any one of the aforementioned CanIon modulators, polypeptides, polynucleotides or antibodies in the treatment of a human body or in the manufacture of a medicament for the treatment of a disease or condition described above.
[43] Also provided are kits for using and detecting CanIon polynucleotides and polypeptides of the invention in vitro or in vivo.
[1] The present invention relates to voltage-gated ion channel genes and their roles in proteins and diseases. The present invention relates to a polynucleotide encoding a CanIon polypeptide and a regulatory region located at the 5'- and 3'-ends of the coding region. The present invention also relates to methods of screening CanIon channel modulators (eg antagonists) and the use of such modulators for the treatment or prevention of various diseases or conditions. The invention also relates to antibodies specifically directed to such polypeptides useful as diagnostic reagents. The present invention also relates to dual allele markers of the CanIon gene useful for gene analysis.
[44] 1 is a diagram showing a BAC map of a chromosome 13q region containing a CanIon gene.
[45] 2 is a block diagram of a typical computer system.
[46] 3 is a flow diagram illustrating one embodiment of a process 200 for measuring the level of homology between the new sequence and the sequence of the database by comparing the new nucleotide or protein sequence with the sequence of the database.
[47] 4 is a flow diagram illustrating one embodiment of a process 250 in a computer to determine whether two sequences are homologous.
[48] 5 is a flow diagram illustrating an embodiment of a verification process 300 for detecting the presence of a feature in a sequence.
[49] Brief description of the sequences presented in the sequence list
[50] SEQ ID No 1 has a genomic sequence of CanIon comprising a 5 ′ regulatory region (upstream untranscribed region) and exons 1-7.
[51] SEQ ID No 2 holds the genomic sequence of CanIon comprising exons 8-27.
[52] SEQ ID No 3 contains the genomic DNA sequence of CanIon comprising exons 28-44 and 3 ′ regulatory region (downstream untranscribed region).
[53] SEQ ID No 4 carries the cDNA sequence of CanIon.
[54] SEQ ID No 5 has an amino acid sequence encoded by SEQ ID No 4.
[55] SEQ ID No 6 holds the nucleotide sequence of an amplicon comprising double allele marker A18.
[56] SEQ ID No 7 has a primer further comprising a PU 5 ′ sequence described in detail in Example 2.
[57] SEQ ID No 8 has a primer further comprising the RP 5 'sequence described in detail in Example 2.
[58] In accordance with the regulations relating to the sequence list, the following code was used in the sequence list to indicate the position of the double allele marker in the sequence and to identify each of the alleles present in the polymorphic base. The code "r" in the sequence indicates that one allele of the polymorphic base is guanine and the other allele is adenine. The code "y" in the sequence indicates that one allele of the polymorphic base is thymine and the other allele is cytosine. The code "m" in the sequence indicates that one allele of the polymorphic base is adenine and the other allele is cytosine. The code "k" in the sequence indicates that one allele of the polymorphic base is guanine and the other allele is thymine. The code "s" in the sequence indicates that one allele of the polymorphic base is guanine and the other allele is cytosine. The code "w" in the sequence indicates that one allele of the polymorphic base is adenine and the other allele is thymine. The nucleotide code of the circular allele for each dual allele marker is as follows:
[59] Double allele marker circular allele
[60] 5-124-273 ----------- A (for example)
[61] In some cases, the polymorphic base of the double allele marker changes the nature of the amino acid in the encoded polypeptide. This is shown in the attached sequence list using the VARIANT feature, the placement of Xaa at the position of the polymorphic amino acid, and the definition of Xaa as two alternative amino acids. For example, if one allele of a double allele marker is a codon CAC encoding histidine and the other allele of a double allele marker is a CAA encoding glutamine, then the sequence list for the encoded polypeptide is the position of the polymorphic amino acid. Will have Xaa. In this case, Xaa is defined as histidine or glutamine.
[62] In other cases, Xaa may represent amino acids whose identity is unclear due to nucleotide sequence multiplicity. In this case, the definition of Xaa is used with the UNSURE feature, the placement of Xaa at the location of the unknown amino acid, and 20 or a limited number of amino acids by genetic code.
[63] Concentration of schizophrenia and bipolar disorder in families, evidence of twins and adoption studies, and lack of change in global incidence suggest that schizophrenia and bipolar disorder are primarily genetic diseases, even if environmental risk factors are involved in some degree of development. do. For example, schizophrenia occurs in 1% of the population. However, if one of the grandparents has schizophrenia, the risk of getting sick is about 3%; If one parent has schizophrenia, it increases by 10%. If both parents have schizophrenia, the risk is up to 40%.
[64] Identification of genes involved in specific traits, such as specific central nervous system diseases such as schizophrenia, can be carried out through two main strategies currently being used for gene mapping: chain analysis and related studies. Chain analysis requires family studies of several patients and is currently used for the detection of single- or olgogenic inherited traits. In contrast, linkage studies examine the frequency of marker alleles in irrelevant trait (T +) individuals as compared to trait negative (T−) controls and are generally used for detection of polygenic inheritance.
[65] The genetic link or "chain" is based on the analysis of whether two adjacent sequences in the chromosome retain minimal recombination by gene exchange during meiosis. To this end, chromosomal markers, such as microsatellite markers, are precisely located in the genome. Genetic link analysis measures recombination in the chromosome markers and target genes used in the genealogical tree, the transmission of the disease, and the likelihood of delivery of the markers. Thus, if a particular allele of any marker in the disease is delivered at a higher frequency than the usual level (recombination level of 0 to 0.5), it is possible to infer that the target gene is found in the vicinity of the marker. Using this technique, we identified the location of several genes that show the genetic predisposition to familial cancer. In order to be included in gene chain studies, families affected by genetic disorders must meet the "information factor" criteria: multiple affected individuals (available from constituent DNA) and many descendants per generation.
[66] The results of previous studies supported the hypothesis that chromosome 13 is more likely to have schizophrenic loci at 13q32 (Blouin JL et al., 1998, Nature Genetics, 20: 70-73; Lin MW et al., 1997, Hum. Genet., 99 (3): 417-420; Brzustowicz et al., Am. J. Hum. Genet. 65: 1096-1103 (1999)). Although chain analysis is an effective method for detecting genes involved in traits, complementarity studies are often required to refine the analysis of the regions initially identified by the method without analytical capacity exceeding the megabase level.
[67] BAC contig (contigs) containing the candidate genomic region of the chromosome 13q-31-q33 locus was constructed with published STS located in the chromosome 13q31-33q region to screen for seven genome equivalent proprietary BAC libraries. New STSs were made from these materials and a dense physical map of the area was created. BAC was sized and mapped by in situ chromosome hybridization for validation. Minimal BACs were identified and fully sequenced, where a contiguous sequence consisting of more than 4Mb sequences was created. This mapping resulted in the identification of the CanIon gene located in a genomic region with marked schizophrenia.
[68] The CanIon amino acid sequence is characteristic of the Ca channel, a seven-component fingerprint that provides a signature for the alpha-1 subunit of the calcium channel. This fingerprint was derived from an initial alignment consisting of six sequences: motifs were derived from conserved loop regions that can distinguish them from other cation channels; Motif 1 and 2 encode the conserved loop region between transmembrane segments 4 and 5 and 5 and 6 (first internal repeat); Motif 3 corresponds to a conserved loop region between segment 6 of repeat 1 and segment 1 of repeat 2; Motif 4 encodes the conserved loop region between segments 5 and 6 of repetition 2; Motif 5 corresponds to a conserved loop region between segment 6 of repeat 3 and segment 1 of repeat 4; Motif 6 and 7 encode the conserved loop region between segments 4 and 5 and 5 and 6 of repetition sequence 4
[69] FIG. 1 depicts the BAC contics covering the Chromosome 13 region of interest, including the CanIon gene, and shows the genomic location of the CanIon gene in relation to the genetic marker of highest importance in serial studies. In particular, Blouin et al. (1998) conducted a genome wide scan of schizophrenic susceptible loci using 452 microsatellite markers in 54 complex lineages. The most prominent link between schizophrenia in the family was identified at chromosome 13q32 around marker D13S174. Brzustowics et al. (1999) evaluated microsatellite markers across chromosomes 8 and 13 in 21 enlarged Canadian lines. Markers in the chromosome 13q region showed positive LOD scores in each analytical model used: autosomal dominant and recessive according to the narrow or broad definition of schizophrenia. Maximum 3-point LOD score was obtained with marker D13S798 under recessive broad model: recombination fraction (θ) under homology. 4.42 at α = .65 and θ = 0 under heterogeneity. Referring to FIG. 1, the CanIon gene is partially located in the contiguous 'E region' and contiguous C0001A10. The CanIon gene is flanked by two markers of highest importance in chain studies. Marker D13S174 is located at contic C0001A10 and marker D13S793 is located at approximately 3.5 Mb center of CanIon gene.
[70] It is necessary to identify genes involved in schizophrenia, bipolar disorder and other CNS and cardiovascular diseases and abnormalities. In addition, it is necessary to identify new ion channels involved in the disease. These genes and proteins provide new points of intervention in the treatment of schizophrenia, bipolar disorder or other CNS diseases and other diseases, such as heart and hypertension, and enable further study and characterization of the CanIon gene and related biological pathways. . Knowledge of these genes and related biological pathways involved in these diseases and abnormalities will allow speakers to understand, for example, the etiology of schizophrenia and bipolar disorder, and will result in drugs and drug treatments for the cause of the disease. For example, compounds that block CanIon channels can be used to treat a variety of diseases or disorders, preferably schizophrenia or bipolar disorder, pain disorders, epilepsy, various cardiovascular diseases (eg, cardiac arrhythmias), manic, hypertension. There is also a need for new methods of detecting susceptibility to schizophrenia, bipolar disorder and other abnormalities and preventing or tracking the development of these diseases. Diagnostic tools are also very useful. Indeed, early identification of individuals at high risk of developing schizophrenia enables diagnostic and / or prophylactic treatment. In addition, an accurate assessment of the efficacy of the drug and the patient's tolerability will allow doctors to improve the benefit / risk ratio of treatment for schizophrenia and bipolar disorder.
[71] The present invention relates to polynucleotides and polypeptides associated with the CanIon gene. Oligonucleotide probes and primers that hybridize specifically with the CanIon genome or cDNA sequence also belong to the present invention. Another object of the present invention is to provide a recombinant vector comprising one of the nucleic acid sequences disclosed herein, in particular a recombinant vector comprising a regulatory region of CanIon or a sequence encoding CanIon protein and a host cell comprising said nucleic acid sequence or recombinant vector. To provide. The present invention also relates to methods of screening molecules for their ability to modulate the expression or activity of the CanIon gene or protein and methods of using such molecules to treat or prevent schizophrenia, bipolar disorder or various other diseases or disorders. The invention also relates to antibodies specifically directed to such polypeptides useful as diagnostic reagents.
[72] The present invention also relates to CanIon-related double allele markers and their use in gene analysis methods, including chain studies in families, chain imbalance studies in populations, and linkage studies in case control populations. An important aspect of the present invention is that the double allele markers allow for linkage studies to identify the role of genes involved in complex traits.
[73] Justice
[74] Prior to describing the invention in more detail, the following definitions are set forth in order to limit the meaning and scope of the terms used to describe the invention.
[75] "CanIon gene" as used herein includes genome, mRNA, cDNA sequences that encode CanIon protein and comprise an untranslated regulatory region of genomic DNA.
[76] As used herein, “heterologous protein” means any protein or polypeptide except CanIon protein. For example, the heterologous protein may be a compound that can be used as a marker for further testing with the CanIon regulatory region.
[77] "Isolated" means that the material is moved from its original environment (eg, inherent environment if it occurs naturally). For example, naturally-occurring polynucleotides or polypeptides present in living animals are not isolated, but those same polynucleotides, DNAs or polypeptides isolated from some or all of the coexisting material in a native system are isolated. Such polynucleotides may be part of a vector or such polynucleotides or polypeptides may be part of a composition, which is also isolated in that the vector or composition is not part of the intrinsic environment.
[78] For example, naturally-occurring polypeptides present in living animals are not isolated, but the same polynucleotides isolated from some or all of the coexisting material in a native system are isolated. Naturally-occurring chromosomes (eg, chromosomal spreads) in the definition of “isolated”; Artificial chromosome libraries; Genomic library; Excludes cDNA libraries present in in vitro nucleic acid formulations or transfection / transformed host cell formulations, where the host cells are in vitro heterogeneous formulations or plated into heterogeneous populations of single colonies. Also excluded are libraries in which the specified polynucleotide comprises less than 5% of the number of nucleic acid inserts in the vector molecule. Whole cell genomic DNA or whole cell RNA formulations (including mechanically sheared or enzymatically cleaved whole cell formulations) are also excluded. Such whole cell formulations are also excluded as in vitro formulations or heterogeneous mixtures separated by electrophoresis (blot delivery), wherein the polynucleotides of the present invention are no longer separated from heterologous polynucleotides in an electrophoretic medium (eg, agarose). Further separation by cleaving a single band from the heterogeneous band population in a gel or nylon blot).
[79] “Purified” herein does not require absolute purity; Rather, it is understood as a relative definition. Purification of the starting material or natural material is thought to be at least one size digit, preferably two or three size digits, more preferably four or five size digits. As an example, a tablet from 0.1% to 10% concentration is 2 size digits. By way of example, individual cDNA clones from a cDNA library are typically purified to electrophoretic homogeneity. Sequences obtained from these clones could not be obtained from the library or from whole human DNA. cDNA clones are not naturally occurring and are obtained through manipulation of partially purified naturally occurring substances (messenger RNA). Conversion of mRNA to cDNA library involves the production of synthetic material (cDNA), and pure individual cDNA clones can be isolated from the synthetic library by clonal selection. Thus, creating a cDNA library from messenger RNA and subsequent isolation of individual clones from the library results in 10 4 -10 6 fold purification of the unique message.
[80] In addition, “purified” can be used to describe polypeptides or polynucleotides of the invention that are isolated from other compounds, including but not limited to polypeptides, polynucleotides, carbohydrates, lipids, and the like. "Purified" may also be used to specify the separation of monomeric polypeptides of the invention from oligotypes such as homo- or hetero-dimers, trimers, and the like. "Purified" may also be used to specify the separation of covalently linked polynucleotides from linear polynucleotides. The polypeptide is substantially pure when at least 50%, preferably 60 to 75% of the sample shows conformation with a single polynucleotide sequence. Substantially pure polypeptide or polynucleotide consists of approximately 50%, preferably 60-90%, more preferably approximately 95%, most preferably at least 99% w / w of polypeptide or polynucleotide sample, respectively. Polynucleotide purity or homogeneity with the polypeptide is indicated by methods known in the art, such as agarose or polyacrylamide gel electrophoresis of the sample, followed by single band visualization immediately after staining the gel. Higher assays for specific purposes can be provided by HPLC or other known methods. In an alternative embodiment the purification of polypeptides and polynucleotides according to the invention may be expressed in% at least "purity" with respect to heterologous polypeptides and polynucleotides (DNA, RNA or both). In suitable embodiments, the polypeptides and polynucleotides of the present invention are at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, with respect to heterologous polypeptides and polynucleotides, respectively, 95%, 96%, 96%, 98%, 99%, or 100% purity. In other suitable embodiments, the polypeptide and the polynucleotide are each from 90% to 100% purity (eg, at least 99.995% purity polypeptide or polynucleotide), or carrier, between 90% and 100% with respect to the heterologous polypeptide or polynucleotide, respectively. Purity is in the w / w ratio with respect to all compounds and molecules except for the compounds and molecules present in. Each number representing purity% up to 1/1000 digits can be claimed in a separate form of purity.
[81] "Polypeptide" herein means an amino acid polymer regardless of the length of the polymer; Thus, peptides, oligopeptides, proteins are included in the definition of polypeptide. The term does not specify or exclude post-expression modifications of the polypeptide, eg, polypeptides having glycosyl groups, acetyl groups, phosphate groups, lipid groups, and the like, are included in a polypeptide. Polypeptides having one or more amino acid analogs (eg, non-naturally occurring amino acids, amino acids naturally occurring only in unrelated organisms, modified amino acids derived from mammals, etc.), polypeptides with substituted chains, naturally occurring and naturally occurring Other variants known in the art that do not occur are also included in the polypeptide.
[82] By “recombinant polypeptide” is meant herein a polypeptide that is artificially designed and that comprises at least two polypeptide sequences that are not found in the continuous polypeptide sequence of the original native environment, or a polypeptide expressed from a recombinant polynucleotide.
[83] A "human exclusion animal" as used herein refers to a human exclusion vertebrate, bird, mammal, preferably primate, poultry (e.g. pig, goat, sheep, donkey, horse, rabbit) or rodent, more preferably rat or mouse. Means. By “animal” is meant herein any vertebrate, preferably mammal. "Animals" and "mammals" include humans unless "excluding people" is specified.
[84] By “antibody” herein is meant a polypeptide or group of polypeptides consisting of one or more binding domains, wherein the antibody binding domain is created by the folding of various domains in an antibody molecule and is an interior surface complementary to the antigenic determinant appearance of the antigen. It forms a three-dimensional binding space with shape and charge distribution, which allows for an immunological reaction with the antigen. Antibodies include recombinant proteins bearing binding domains, including Fab, Fab ', F (ab) 2 , F (ab') 2 .
[85] By “antigen determinant” herein is meant a portion of an antigen molecule, herein a CanIon polypeptide, that determines the specificity of an antigen-antibody response. "Epitope" means the antigenic determinant of a polypeptide. Epitopes consist of at least three amino acids that give the epitope a unique spatial conformation. Generally, the epitope consists of at least six such amino acids, preferably at least 8-10 such amino acids. Methods for measuring the amino acids constituting the epitope include x-ray determination; Two-dimensional nuclear magnetic resonance; Epitope mapping, for example Geysen et al. 1984; PCT Application WO 84/03564; Pepscan method as disclosed in PCT application WO 84/03506 is included.
[86] As used herein, "nucleotide sequence" refers to a polynucleotide or nucleic acid. More specifically, the "nucleotide sequence" includes the nucleic acid material itself, and thus is not limited to sequence information (ie, a sequence of letters selected from four base letters) that biochemically characterizes a particular DNA or RNA molecule.
[87] As used herein, "nucleic acid", "oligonucleotide", "polynucleotide" have the same meaning as each other, where an RNA, DNA, or RNA / DNA hybrid sequence comprising one or more nucleotides in the form of a single chain or a double helix Included. As used herein, "nucleotide" also includes sequences comprising RNA, DNA, or RNA / DNA hybrid sequences of any length in the form of a single chain or a double helix. In addition, the term “nucleotide” herein consists of a purine or pyrimidine, ribose or deoxyribose sugar component, a phosphate group or a phosphodiester bond in the case of individual nucleotides or a plurality of nucleotides, ie oligonucleotides or nucleotides of polynucleotides. In a molecule or larger nucleic acid molecule is meant an individual unit. "Nucleotides" includes "modified nucleotides" comprising at least one variant (a) another linking group, (b) an analog of purine, (c) an analog of pyrimidine, or (d) a sugar analog (see WO95 / 04064). The polynucleotide sequences of the present invention can be made using any known method and purification methods known in the art, including synthesis, recombination, ex vivo generation or combinations thereof.
[88] Regulatory sequences, such as sequences "operably linked" to a promoter, mean that the regulatory elements are present in the nucleic acid in the correct position and orientation to regulate RNA polymerase initiation and expression of the nucleic acid of interest. By "operably linked" is meant herein a chain of polynucleotide elements in a functional correlation. For example, a promoter or enhancer is operably linked to a coding sequence and affects the transcription of the coding sequence.
[89] “Ttrait” and “phenotype” have the same meaning herein and refer to the visible, detectable or otherwise measurable properties of a microorganism, such as the symptoms or susceptibility to a disease. In general, “trait” or “phenotype” means a symptom or susceptibility to a disease, a beneficial response associated with treatment, or a side effect associated with treatment. Suitably, these traits include psychiatric disorders such as schizophrenia or bipolar disorder; Other CNS or neurological diseases such as epilepsy or pain disease; Cardiovascular diseases such as angina, high blood pressure, arrhythmia; Any aspect, feature, or characteristic of one of these diseases or conditions may be, but is not limited to.
[90] "Allele" means a variation of the nucleotide sequence. Double allele polymorphisms take two forms. Diploid microorganisms may be homozygous or heterozygous for one allele type.
[91] As used herein, "heterozygosity rate" refers to the frequency of individuals in a population that is heterozygous for a particular allele. In a dual allele system, the heterozygosity rate is on average 2P a (1-P a ), where P a is the frequency of the least common allele. Genetic markers useful for genetic research possess a suitable level of heterozygosity that allows for a reasonable acceptance of the likelihood that a randomly selected individual is heterozygous.
[92] "Genetic" as used herein means the nature of the allele present in the individual or sample. Suitably, genotype refers to a double allele marker allele present in an individual or sample, eg, the allele of a double allele marker in the CanIon gene or genomic region. "Genotyping" of a sample or individual to a "dual allele marker" includes determining a particular allele or a specific nucleotide held by the individual in a biallelic marker.
[93] By “mutation” herein is meant a difference in DNA sequence between different genomes or individuals between frequencies of less than 1%.
[94] As used herein, "haplotype" means a combination of alleles present in an individual or sample. Suitably, a haplotype refers to a combination of double allele marker alleles found in any individual and associated with a phenotype.
[95] "Polymorphism" as used herein means the generation of two or more genomic sequences or alleles between different genomes or individuals. "Polymorphic" means a condition in which two or more variations in a particular genomic sequence are found in a population. "Polymorphic site" is the locus in which a mutation occurs. Single nucleotide polymorphism is the substitution of one nucleotide for another nucleotide at a polymorphic site. Deletion of a single nucleotide or addition of a single nucleotide also results in single nucleotide polymorphism. As used herein, "single nucleotide polymorphism" refers to single nucleotide substitution whenever possible. Typically, polymorphic sites between different individuals may be occupied by two different nucleotides. "Dual allele polymorphism" and "double allele marker" as used herein mean a single nucleotide polymorphism that has the same meaning and has two alleles at a fairly high frequency in a population. "Dual allele marker allele" means a nucleotide variation present at a biele marker site. Typically, the less common frequency of alleles of the dual allele marker according to the present invention is at least 1%, preferably at least 10%, more preferably at least 20% (ie at least 0.32 heterozygosity), most Preferably at least 30% (ie at least 0.42 release). A lesser common allele marker with a frequency of more than 30% is called the "high quality double allele marker."
[96] The position of nucleotides in the polynucleotide relative to the center of the polynucleotide is described in the following manner. If the polynucleotide contains an odd number of nucleotides, the nucleotides that are equally distanced from the 3 'and 5' ends of the polynucleotide are considered to be the "center" of the polynucleotide and are located at the nucleotides immediately adjacent to the centered nucleotides or at the center itself. Nucleotides are considered to be located within 1 nucleotide of the center. One of the five nucleotide positions in the middle of a polynucleotide having an odd number of nucleotides is considered to be located within 2 nucleotides of the center. Having a nucleotide, there is a non-nucleotide bond in the center of the polynucleotide, so one of the two center nucleotides is considered to be located "within 1 nucleotide from the center." And one of the four nucleotides in the middle of the polynucleotide is considered to be located “within two nucleotides from the center.” In the case of polymorphism, including substitution, deletion or addition of one or more nucleotides, a polymorphic substituted, inserted The distance between the 3 'end of the polynucleotide and the distance from the substituted or deleted polynucleotide and the distance between the 5' end of the polynucleotide and the distance from the substituted, inserted or deleted polynucleotide of the polymorphism is 0 or 1 nucleotide A polymorphic, allele, or dual allele marker is located at the "center" of the polynucleotide, if this difference is 0-3, the polymorphism is considered to be "within 1 nucleotide from the center." If 5, the polymorphism is "within two nucleotides from the center" Considered to be. If this difference is 0 to 7, the polymorphism is considered to be positioned "within 3 nucleotides from the center."
[97] “Upstream” herein means a position from the particular reference point to the 5 ′ terminal direction of the polynucleotide, or in the case of a gene, from the coding sequence to the promoter direction.
[98] As used herein, "base pair" and "Watson & Crick base pair" mean nucleotides that have the same meaning and are hydrogen bonded to each other due to sequence homology in a manner similar to that found in double-stranded DNA, where thymine or uracil residues are two hydrogens Binding to adenine residues and cytosine residues to guanine residues by three hydrogen bonds (Stryer, L., Biochemistry, 4 th edtion, 1995)
[99] "Complementarity" or "complement thereof" means a polynucleotide sequence capable of forming Watson & Crick base pairs with other specified polynucleotides in the region of complementarity. In the present invention, when each base is paired with a complementary base in the first polynucleotide, the first polynucleotide is said to be complementary to the second polynucleotide. Complementary bases are A and T (or A and U), or C and G. As used herein, "complement" is used synonymously with "complementary nucleotide", "complementary nucleic acid", "complementary nucleotide sequence. These terms refer to a pair of purely sequence-based polynucleotides, not to specific conditions under which two polynucleotides actually react. Apply.
[100] Mutations and Shorts
[101] The present invention relates to mutations and fragments of the above-described polynucleotides, in particular the CanIon gene, which bears at least one dual allele marker according to the invention.
[102] Variation of a polynucleotide herein is a polynucleotide different from a reference polynucleotide. Variations of the polynucleotides are naturally occurring variants, for example naturally occurring allelic variants or variants that are not known to occur naturally. Such non-naturally occurring variations of polynucleotides can be made by mutagenesis techniques, including those applied to polynucleotides, cells or microorganisms. In general, differences are limited and variations with reference nucleotide sequences are generally similar and homologous in many regions.
[103] Variations of the polynucleotide according to the present invention include polynucleotide fragments of at least 12 consecutive nucleotides of SEQ ID No 1 to 4 nucleotide sequences or at least 95% homologous to SEQ ID No 1 to 4 nucleotide sequences or SEQ ID No 1 to 4 nucleotide sequences and SEQ ID No 1 to Preferably at least 99% homologous, more preferably 99.5% homologous, most preferably at least 99.8% homologous to a polynucleotide fragment of at least 12 consecutive nucleotide sequences of a 4 nucleotide sequence or SEQ ID No 1 to 4 nucleotide sequences One nucleotide sequence is included, but is not limited to these.
[104] Nucleotide changes present in variant polynucleotides may be silent, meaning that they do not change the amino acids encoded by the polynucleotide. However, nucleotide changes may also result in amino acid substitutions, additions, deletions, fusions, truncations in the polypeptide encoded by the reference sequence. Substitutions, deletions or additions may involve one or more nucleotides. The variation may occur in the coding region, the non-coding region or both. Changes in the coding region can lead to conservative or non-conservative amino acid substitutions, deletions or additions.
[105] In a particularly suitable embodiment of the present invention, the polynucleotide encodes a polypeptide that substantially retains the same biological function or activity as the mature CanIon protein, or a polypeptide that reduces secondary biological activity while maintaining or increasing specific biological activity.
[106] A polynucleotide fragment is a polynucleotide having a sequence that is completely homologous to any nucleotide sequence, preferably the nucleotide sequence of the CanIon gene and portions of its variations. The fragment may be part of an intron or exon of the CanIon gene. This may be part of the control region of CanIon. Suitably, such fragments comprise a dual allele marker that is unbalanced with at least one of the dual allele markers A1 to A17 or complements thereof, or at least one of the dual allele markers A1 to A17.
[107] Such fragments may be “free-standing”, that is to say not fused to a portion or other polynucleotide of another polynucleotide, or may form a portion or region in a larger single polynucleotide. Indeed, some of these fragments are present on larger polynucleotides.
[108] Alternatively, such fragments may consist of at least 8, 10, 12, 15, 18, 20, 25, 35, 40, 50, 70, 80, 100, 250, 500 or 1000 contiguous nucleotides. One set of suitable fragments carries at least one of the dual alleles A1 to A17 of the CanIon gene or complement thereof.
[109] 2-polypeptide
[110] The invention also relates to variants, fragments, analogs, derivatives of the aforementioned polypeptides, including mutated CanIon proteins.
[111] Variations include those in which one or more amino acid residues are substituted with conserved or non-conserved amino acid residues and such substituted amino acid residues are encoded or not encoded by the genetic code; 2) one or more amino acid residues bearing a substituent; 3) mutations in which the mutated CanIon is fused with another compound, such as a compound that increases the half-life of the polypeptide (eg, polyethylene glycol); Or 4) the added amino acid may be a fusion to a mutant CanIon, eg, a leader or secretory sequence or a sequence used for purification of a mutated CanIon or shear protein (preprotein) sequence.
[112] A polypeptide fragment is a polypeptide having a sequence that is fully homologous to any polypeptide sequence, preferably a portion of the polypeptide encoded by the CanIon gene and variants thereof.
[113] In the case of amino acid substitutions in the amino acid sequence of a polypeptide according to the invention, one or several amino acids may be substituted with "equivalent" amino acids. An “equivalent” amino acid herein is any capable of replacing amino acids having similar properties, such that one skilled in the art of peptide chemistry can expect that the secondary structure and hydropathic properties of the polypeptide will not substantially change. It means amino acid. In general, the following groups of amino acids represent equivalent changes: (1) Ala, Pro, Gly, Glu, Asp, Gln, Asn, Ser, Thr; (2) Cys, Ser, Tyr, Thr; (3) Val, Il 3, Leu, Met, Ala, Phe; (4) Lys, Arg, His; (5) Phe, Tyr, Trp, His.
[114] Certain embodiments of the modified CanIon peptide molecule according to the present invention include peptide molecules that resist proteolysis, namely -CONH-peptide bonds and (CH2NH) reduced bonds, (NHCO) retro involute bonds, (CH2- O) methylene-oxy bond, (CH2-S) thiomethylene bond, (CH2CH2) carba bond, (CO-CH2) ketomethylene bond, (CHOH-CH2) hydroxyethylene bond, (NN) bond, E-alsen bond Or peptides substituted with —CH═CH bonds, but are not limited to these. In addition, the present invention includes fragments or variations thereof in which a human CanIon polypeptide or at least one peptide bond is modified as described above.
[115] Such fragments may be “free-standing”, ie, fused to a portion or other polypeptide of another polypeptide, or may form a portion or region in a larger single polypeptide. In fact, some fragments are included in larger single polypeptides.
[116] Representative examples of other polypeptide fragments in the present invention are polypeptides having approximately 5, 6, 7, 8, 9, 10-15, 10-20, 15-40, or 30-55 amino acids. Suitably, the polypeptide fragment has at least one amino acid mutation in the CanIon protein.
[117] Homology Between Nucleic Acids or Polypeptides
[118] As used herein, "% sequence homology" and "% homology" have the same meaning and mean a comparison between a polynucleotide and a polypeptide and are determined by comparing two optimally aligned sequences in a comparison window, wherein the polynucleotide on the comparison window or Portions of a polypeptide sequence may retain additions or deletions (ie gaps) as compared to a reference sequence (no additions or deletions) for optimal alignment of the two sequences. % Sequence homology measures the total number of positions where identical nucleic acid bases or amino acid residues occur in both sequences, yielding the total number of concatenated positions, dividing the total number of concatenated positions by the total number of positions in the comparison window, Calculate by multiplying by. Homogeneity is assessed using various sequence comparison algorithms and programs known in the art. Such algorithms and programs include TBLASTN, BLASTIP, FASTA, TFASTA, CLUSTALW (Pearson and Lipman, 1988; Altschul et al., 1990; Thompson et al., 1994; Higgins et al., 1996; Altschul et al., 1990; Altschul et al., 1993), but is not limited to these. In particularly suitable embodiments, protein and nucleic acid sequence homogeneity is assessed using the Basic Local Alignment Tool (“BLAST”) known in the art (Karlin and Altschul, 1990; Altschul et al., 1990 1993, 1997).
[119] In particular, five specific BLAST programs are used to perform the following tasks:
[120] (1) BLASTP and BLAST3 compare the protein sequence database and amino acid query sequence;
[121] (2) BLASTN compares the nucleotide sequence database with the nucleotide query sequence;
[122] (3) BLASTX compares the six-frame putative translation product of the protein sequence database with the query nucleotide sequence (both strands);
[123] (4) TBLASTN compares query protein sequences with translated nucleotide sequence databases in six translation frames (both strands);
[124] (5) TBLASTX compares the six-frame translation of the nucleotide sequence database with the six-frame translation of the nucleotide query sequence.
[125] The BLAST program identifies homologous sequences by identifying similar segments, which are then referred to as "high-scoring segment pairs" between query amino or nucleic acid sequences and test sequences obtained from a protein or nucleic acid sequence database. High-scoring segment pairs are identified (ie aligned), preferably with a scoring matrix known in the art. The scoring matrix used is preferably the BLOSUM62 matrix (Gonnet et al., 1992; Henikoff and Henikoff, 1993). Although less preferred, PAM or PAM250 matrices are also available (Schwartz and Dayhoff, eds., 1978). The BLAST program evaluates the statistical significance of all identified high-scoring segment pairs and selects segments that satisfy the user-specific significance limit, eg, user-specific% of homogeneity, if possible. Suitably, the statistical significance of the high-scoring segment pairs is assessed using Karlin's statistical significance formula (Karlin and Altschul, 1990).
[126] The BLAST program can be used as a default variable or as a user-supplied parameter.
[127] Strict Hybridization Conditions
[128] Stringent hybridization conditions for defining hybridizing nucleic acids according to the present invention are as follows:
[129] The hybridization step is achieved at 65 ° C. in the presence of 6 × SSC buffer, 5 × Denhardt solution, 0.5% SDS, 100 μg / ml salmon sperm DNA.
[130] Following the hybridization step, four wash steps are performed:
[131] Wash twice for 5 min at 65 ° C. in the presence of 2 × SSC and 0.1% SDS buffer;
[132] Wash once for 30 min at 65 ° C. in the presence of 2 × SSC and 0.1% SDS buffer;
[133] Wash once for 10 min at 65 ° C. in the presence of 2 × SSC and 0.1% SDS buffer.
[134] These hybridization conditions are suitable for nucleic acid molecules consisting of approximately 20 nucleotides. The above hybridization conditions can be adjusted according to the length of the desired nucleic acid by techniques known to those skilled in the art. For example, suitable hybridization conditions can be adjusted according to what is disclosed in the book of Hames and Higgins (1985).
[135] Genome sequence of the CanIon gene
[136] The present invention relates to the genomic sequence of CanIon. The present invention includes the CanIon gene, CanIon genomic sequence comprising SEQ ID Nos. 1-3 sequences, complementary sequences thereof, fragments and variants thereof. These polynucleotides can be purified, isolated or recombinant.
[137] In addition, the present invention provides an isolated, purified comprising a nucleotide sequence having at least 70, 75, 80, 85, 90, or 95% nucleotide homology with the nucleotide sequence of SEQ ID Nos. 1 to 3, a complementary sequence thereof, or a fragment thereof. Or recombinant polynucleotides are included. In general, nucleotide differences associated with SEQ ID No 1 to 3 nucleotide sequences are distributed randomly throughout the nucleic acid. Nevertheless, preferred nucleic acids are nucleic acids whose nucleotide differences associated with SEQ ID Nos. 1 to 3 nucleotide sequences are located outside the coding sequence included in the exon. These nucleic acids, fragments and variants thereof can be used as oligonucleotide primers or probes for detecting the presence of a CanIon gene copy in a test sample, or alternatively for amplifying a target nucleotide sequence in a CanIon sequence.
[138] Another object of the present invention relates to an isolated, purified, recombinant nucleic acid that hybridizes with the SEQ ID No 1 to 3 nucleotide sequences, complementary sequences or variants thereof under the stringent hybridization conditions described above. In suitable embodiments, the purified, isolated or recombinant nucleic acid is specifically hybridized with polynucleotides of the human CanIon gene, and more suitably the nucleic acid may hybridize with the nucleotides of the human CanIon gene, but not of the mouse CanIon gene. It may hybridize with nuclear line sequences.
[139] Particularly suitable nucleic acids of the invention include those isolated having at least 12, 15, 18, 20, 25, 35, 40, 50, 70, 80, 100, 250, 500 or 1000 contiguous nucleotides of SEQ ID Nos. And purified, recombinant polynucleotides. Nucleic acid fragments of any size and sequence may also include the polynucleotides identified in this section.
[140] CanIon genomic nucleic acid consists of 44 exons. Exon positions in SEQ ID Nos. 1 to 3 are described in detail in Table B below.
[141] TABLE B
[142]
[143]
[144]
[145] Thus, the invention includes isolated, purified, recombinant polynucleotides comprising a nucleotide sequence selected from 44 exons of the CanIon gene or a complement sequence thereof. The present invention also provides a purified, isolated, recombinant nucleic acid comprising a combination of at least two exons of the CanIon gene, wherein the polynucleotides are 5 'in the nucleic acid in the same order as in SEQ ID Nos. 1 to 3 From end to 3 'end.
[146] Intron 1 represents the nucleotide sequence located between exon 1 and exon 2. The location of the introns is described in detail in Table A. Accordingly, the present invention includes isolated, purified, recombinant polynucleotides comprising a nucleotide sequence selected from the 43 introns of the CanIon gene or a complement sequence thereof.
[147] Although the title of this section is the "genomic sequence of the CanIon gene", nucleic acid fragments of any size and sequence in contact with the CanIon genomic sequence or between two or more such genomic sequences on either side may also be composed of the polynucleotides identified in this section. Can be.
[148] CanIon cDNA Sequence
[149] Expression of the CanIon gene produces at least one type of mRNA whose nucleic acid sequence is shown in SEQ ID No 4.
[150] Another object of the invention is an isolated, purified, recombinant nucleic acid comprising the SEQ ID No 4 nucleotide sequence, its complementary sequence, allelic variants, fragments. Suitably, polynucleotides of the present invention include isolated, purified, recombinant CanIon cDNA comprising the sequence of SEQ ID No 4. Particularly suitably, nucleic acids of the invention include at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 500, Isolated, purified, recombinant polynucleotides comprising a continuous span of 1000 nucleotides or complement thereof. In a suitable embodiment, the span comprises a CanIon-related double allele marker selected from A12 to A16.
[151] In addition, the present invention relates to a polynucleotide having at least 95% nucleotide homology, preferably 99% nucleotide homology, more preferably 99.5% nucleotide homology, and most preferably 99.8% nucleotide homology with SEQ ID No 4 polynucleotide. It relates to an isolated or purified nucleic acid comprising a nucleotide, its complementary sequence or a biologically active fragment.
[152] Another object of the present invention relates to an isolated, purified, recombinant nucleic acid comprising a polynucleotide, complementary sequence, variant or biologically active fragment thereof that hybridizes with the SEQ ID No 4 polynucleotide under the strict conditions described above.
[153] Another object of the invention relates to an isolated, purified, recombinant polynucleotide encoding a CanIon polypeptide comprising a contiguous span of at least six amino acids of SEQ ID No 5, wherein the contiguous span is SEQ ID No 5 Amino acid positions of 277, 338, 574, 678, 680, 683, 691, 692, 695, 696, 894, 1480, 1481, 1483, 1484, 1485, 1630, 1631, 1632, 1636, 1660, 1667, 1707, 1709 At least one, two, three, five or ten. Furthermore, at least 80, 85, 90, 95, 98, and an isolated, purified, recombinant polynucleotide encoding the amino acid sequence of SEQ ID No 5, a derivative or biologically active fragment thereof, and the amino acid sequence of SEQ ID No 5; Isolated, purified, recombinant polynucleotides encoding CanIon polypeptides that are 99, 99.5 or 99.8% homologous are included in the present invention.
[154] The cDNA of SEQ ID No 4 comprises a 5′-UTR region starting at position 1 nucleotide of SEQ ID No 4 and ending at position 65 nucleotide. The cDNA of SEQ ID No 4 comprises a 3′-UTR region starting at position nucleotide 5283 and ending at position 6799.
[155] As a result, the present invention relates to an isolated, purified, recombinant nucleic acid comprising the nucleotide sequence of CanIon cDNA 5′UTR, its complementary sequence or allelic variant. The present invention also relates to an isolated, purified, recombinant nucleic acid comprising the nucleotide sequence of CanIon cDNA 3′UTR, its complementary sequence or allelic variant thereof.
[156] Although the title of this section is "CanIon cDNA sequence," nucleic acid fragments of any size and sequence that border the CanIon's genomic sequence on either side or between two or more such genomic sequences may also consist of the polynucleotides identified in this section. .
[157] Coding region
[158] The CanIon open reading frame is included in the corresponding mRNA of SEQ ID No 4 cDNA. More specifically, an effective CanIon coding sequence (CDS) comprises a region between position 66 nucleotides (first nucleotide of an ATG codon) and position 5282 (last nucleotide of a TGA codon) of SEQ ID No 4. The present invention provides a polypeptide comprising at least 6 amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably 12, 15, 20, 25, 30, 40, 50, 100 consecutive spans. It relates to an isolated, purified, recombinant polynucleotide encoding.
[159] The aforementioned polynucleotides comprising the coding sequence of the CanIon gene can be expressed in a desired host cell or host microorganism under the control of a suitable expression signal. The expression signal is an expression signal contained in a regulatory region on the CanIon gene of the present invention or an exogenous regulatory nucleic acid sequence. Such polynucleotides may be inserted into a vector for expression and / or amplification under a suitable expression signal.
[160] Regulatory sequence of CanIon
[161] As mentioned above, the genome sequence of the CanIon gene carries regulatory sequences in the non-coding 5-side region and the non-coding 3-side region flanked by the CanIon coding region comprising 44 exons of the gene.
[162] Polynucleotides derived from the 5 'and 3' regulatory regions are useful for detecting the presence of one or more copies of a CanIon nucleotide sequence or fragment thereof in a test sample.
[163] Promoter activity of the 5 ′ regulatory region included in CanIon can be assessed as described below.
[164] In order to identify relevant biologically active polynucleotide fragments or variants of SEQ ID No 1, those skilled in the art will appreciate the use of a recombinant vector carrying a marker gene (ie, beta galactosidase, chloramphenicol acetyl transferase, etc.) to Sambrook et. al. (Sambrook, 1989), wherein expression of the marker gene can be detected under the control of a biologically active polynucleotide fragment or variant of SEQ ID No 1. Genomic sequences located upstream of the first exon of the CanIon gene may be selected from appropriate promoter reporter vectors such as pSEAP-Basic, pSEAP-Enhancer, pβgal-Basic, pβgal-Enhancer, pEGEP-1 promoter reporter vector (Clontech); Or cloned with pGL2-Basic, pGL3-Basic promoter-free luciferase reporter gene vector (Promega). In short, each of these promoter reporter vectors is a multicloning site located upstream of a reporter gene that encodes an easily analytical protein, such as a released alkaline phosphatase, luciferase, β-galactosidase or green fluorescent protein. Holds. The upstream sequence of the CanIon coding region is inserted into the cloning site upstream of the reporter gene in both directions and introduced into a suitable host cell. The level of reporter protein is compared to the level obtained from a vector without an insert at the cloning site. The presence of elevated expression levels in the vector carrying the insert as compared to the level in the control vector suggests the presence of a promoter in the insert. If desired, the upstream sequence can be cloned into a vector carrying an enhancer to increase the level of transcription from the weak promoter sequence. Significantly higher expression levels than those observed in vectors without inserts suggest that the promoter sequence is present in the inserted upstream sequence.
[165] Promoter sequences in upstream genomic DNA are further defined by constructing nested 5 'and / or 3' deletions in upstream DNA by conventional techniques such as exonuclease III or suitable restriction endonuclease cleavage. . The resulting deletion fragment can be inserted into a promoter reporter vector to confirm whether the deletion reduces or eliminates promoter activity (Coles et al. (1998)). In this way, boundaries can be defined. If desired, potential individual regulatory sites in the promoter can be identified, either individually or in combination, by specific site mutagenesis or linker scanning that extinguish the potential transcription factors in the promoter. The effect of these mutations on the level of transcription can be confirmed by inserting the mutations into the cloning site on the promoter reporter vector. Assays of this type are known in the art (WO 97/17359, US Patent No. 5, 374,544; EP 582 796; US Patent No. 5,698,389; US 5,643,746; US Patent No. 5,502,176; US Patent 5,266,488;).
[166] The strength and specificity of the promoter of the CanIon gene can be assessed through the expression level of a detectable polynucleotide operably linked to the CanIon promoter in different types of cells and tissues. The detectable polynucleotide may be a polynucleotide that specifically hybridizes with a predetermined oligonucleotide probe, or a polynucleotide that encodes a detectable protein, including a CanIon polypeptide, fragment or variant thereof. Assays of this type are known in the art (US Patent No. 5,502,176; US Patent No. 5,266,488). Some of these methods are detailed below.
[167] Polynucleotides having regulatory elements located at the 5 'and 3' ends of the CanIon coding region can be effectively used to modulate the transcriptional and translational activity of heterologous polynucleotides of interest.
[168] Accordingly, the present invention relates to isolated or purified nucleic acids, complementary sequences thereof, biologically active fragments or variants comprising polynucleotides selected from 5 ′ and 3 ′ regulatory regions. In one aspect, the “5 ′ regulatory region” is present in the nucleotides at positions 1-2000 of SEQ ID No 1. In one aspect, the “3 ′ regulatory region is in the nucleotide sequence at positions 45842 to 47841 of SEQ ID No 3.
[169] In addition, the present invention provides at least 95% nucleotide homology, preferably 99% nucleotide homology, more preferably 99.5% nucleotide homology, most preferably 99.8% with a polynucleotide selected from the 5 'and 3' regulatory regions It relates to isolated or purified nucleic acids, complementary sequences, variants or biologically active fragments comprising polynucleotides having nucleotide homology.
[170] Another object of the invention is an isolated, purified comprising a polynucleotide that hybridizes to a polynucleotide selected from the nucleotide sequences of the 5 'and 3' regulatory regions, complementary sequences thereof, variants or biologically active fragments under the strict conditions described above. And the recombinant nucleic acid.
[171] Suitably, the fragment of the 5 ′ regulatory region has approximately 1500 to 1000, preferably 500, more preferably 400, even more preferably 300 and most preferably 200 nucleotides.
[172] Suitably, the fragment of the 3 'regulatory region is at least 50, 100, 150, 200, 300, 400 bases.
[173] A “biologically active” polynucleotide derivative of SEQ ID Nos. 1 and 3 comprises a polynucleotide comprising or consisting of a fragment of said polynucleotide which serves as a regulatory region for expressing a recombinant polypeptide or recombinant polynucleotide in a recombinant host cell. to be. It can act as an enhancer or inhibitor.
[174] In the present invention, such nucleic acid or poly when the regulatory polynucleotide has a nucleotide sequence containing transcriptional and translational control information and said sequence is "operably linked" to the desired polynucleotide or to the nucleotide sequence encoding the desired polypeptide. Nucleotides “function” as regulatory regions for expressing recombinant polypeptides or recombinant polynucleotides.
[175] Regulatory nucleotides of the invention can be made from the nucleotide sequences of SEQ ID Nos. 1 to 3 by cleavage with appropriate restriction enzymes, as found in Sambrook et al. (1989). Regulatory polynucleotides can also be made by cleaving SEQ ID Nos. 1 to 3 with an exonuclease enzyme, for example Bal31 (Wabiko et al., 1986). In addition, these regulatory polynucleotides can be made by nucleic acid chemical synthesis as disclosed herein.
[176] Regulatory polynucleotides according to the invention can be part of a recombinant expression vector that can be used to express coding sequences in a desired host cell or host microorganism. Recombinant expression vectors according to the invention are presented herein.
[177] Suitably, the 5'-regulating polynucleotide of the present invention comprises the 5-untranslated region (5'-UTR) of a CanIon cDNA, a biologically active fragment or variant thereof.
[178] Suitably, the 3'-regulating polynucleotide of the present invention comprises the 3-untranslated region (3'-UTR) of a CanIon cDNA, a biologically active fragment or variant thereof.
[179] Another object of the present invention is to provide an isolated or purified nucleic acid consisting of:
[180] a) a nucleic acid comprising a regulatory nucleotide sequence selected from:
[181] (i) the polynucleotide of the 5 ′ regulatory region or its complementary sequence
[182] Nucleotide sequences comprising;
[183] (ii) at least 95% of the nucleotide sequence of the 5 'regulatory region
[184] Polynucleotides having nucleotide homology or complementarities thereof
[185] Nucleotide sequences comprising sequences;
[186] (iii) the nucleotide sequence of the 5 'regulatory region under stringent hybridization conditions;
[187] Comprising the polynucleotide to be hybridized or its complementary sequence
[188] Nucleotide sequence;
[189] (iv) biologically active fragments or variants of (iii), (ii) and (iii) nucleotides
[190] b) a polynucleotide operably linked to a nucleic acid as defined above (a) and encoding a desired polypeptide or nucleic acid;
[191] c) optionally, a nucleic acid comprising a 3'-regulating polynucleotide, preferably a 3'-regulating polynucleotide of the CanIon gene.
[192] In certain embodiments of nucleic acids as defined above, the nucleic acid comprises a 5'-untranslated region (5'-UTR) of a CanIon cDNA, a biologically active fragment or variant thereof.
[193] In another specific embodiment of the nucleic acid as defined above, the nucleic acid comprises a 3'-untranslated region (3'-UTR) of a CanIon cDNA, a biologically active fragment or variant thereof.
[194] Regulatory polynucleotides, biologically active fragments or variants thereof, of the 5'-untranslated region (5'-UTR) are operably linked to the 5'-terminus of the polynucleotide encoding the desired polypeptide or polynucleotide.
[195] Regulatory polynucleotides, biologically active fragments or variants thereof, of the 3'-untranslated region (3'-UTR) are operably linked to the 3'-terminus of the polynucleotide encoding the desired polypeptide or polynucleotide.
[196] Desired polypeptides encoded by the aforementioned nucleic acids can exhibit a variety of properties or origins, including proteins of eukaryotic and prokaryotic origin. Polypeptides expressed under the control of the CanIon regulatory region include bacterial, fungal or viral antigens. In addition, eukaryotic proteins such as intracellular proteins such as “housekeeping” proteins; Membrane-bound proteins such as receptors; Excluded proteins such as endogenous mediators (eg cytokines) are included. The desired polypeptide may be a CanIon protein, in particular a protein consisting of the amino acid sequence of SEQ ID No 5, fragments or variants thereof.
[197] Desired nucleic acids, generally RNA molecules, encoded by the aforementioned polynucleotides are complementary to the desired coding polynucleotides, eg, CanIon coding sequences, and are therefore useful as antisense polynucleotides.
[198] Such polynucleotides may be included in a recombinant expression vector for expressing a desired polypeptide or a desired nucleic acid in a host cell or host microorganism. Suitable recombinant vectors for including such polynucleotides have been described above.
[199] Polynucleotide structure
[200] As used herein, "polynucleotide construct" and "recombinant polynucleotide" have the same meaning and are linear or circular isolated or purified comprising at least two nucleotide sequences that are artificially designed and are not found as contiguous nucleotide sequences in the original native environment. Means polynucleotide.
[201] DNA constructs regulate temporal and spatial CanIon gene expression in recombinant cell hosts and transgenic animals
[202] In order to investigate the physiological and phenotypic consequences of the lack of CanIon protein synthesis at the cellular and multicellular microbial levels, the present invention provides a specific allele of the CanIon genomic sequence or cDNA and the CanIon nucleotide sequence of SEQ ID Nos. 1 to 4 or fragments thereof. In this context, DNA constructs and recombinant vectors are provided that enable conditional expression of said genomic sequence or cDNA copy that retains substitutions, deletions or additions of one or more bases, wherein base substitutions, deletions, additions are exon, intron or regulatory sequences. , Preferably the 5′-regulatory sequence, the exon of the CanIon genomic sequence or the CanIon cDNA of SEQ ID No 4. In a suitable embodiment, the CanIon sequence comprises a double allele marker according to the invention. In a suitable embodiment, the CanIon sequence comprises one of the dual allele markers, preferably one of the allele markers A1 to A17, according to the present invention.
[203] The present invention includes recombinant vectors consisting of any one of the polynucleotides disclosed in the present invention. More specifically, the polynucleotide construct according to the present invention consists of any one of the polynucleotides identified in the "Genomic Sequence of the CanIon Gene" section, the "CanIon cDNA Sequence" section, the "coding region" section, and the "oligonucleotide probe and primer" section. Can be.
[204] A preferred first DNA construct is a tetracycline resistant operon tet derived from E. Coli transposon Tn10 that regulates CanIon gene expression, as identified by Gossen et al. (1992, 1995) and Furth et al. (1994). Based on. Such DNA has seven tet operator sequences derived from Tn10, which sequences are fused to the minimum promoter or 5'-regulatory sequence of the CanIon gene, and the minimum promoter or CanIon regulatory sequence is a sense or antisense oligonucleotide. Or is operably linked to a polynucleotide encoding a polypeptide, including a CanIon polypeptide or peptide fragment thereof. This DNA construct encodes a wild type (tTA) or mutant (rTA) inhibitor in which the same cell is fused to the activating domain of viral protein VP16 of herpes simplex virus under the control of a promoter, for example HCMVIE1 enhancer / promoter or MMTV-LTR. When included, the nucleotide sequence functions as a conditional expression system for the nucleotide sequence of interest. Indeed, preferred DNA constructs of the invention consist of a polynucleotide comprising a tet operator sequence and a polynucleotide comprising a sequence encoding a tTA or rTA inhibitor.
[205] In certain embodiments, the conditionally expressing DNA construct has a sequence encoding a mutant tetracycline inhibitor rTA, wherein expression of the polynucleotide of interest is silent in the absence of tetracycline and induced in the presence thereof.
[206] DNA constructs enabling homologous recombination: substitution vectors
[207] A preferred second DNA construct is constructed from the 5'-end to the 3'-end as follows: (a) a first nucleotide sequence comprised in the CanIon genomic sequence; (b) a nucleotide sequence comprising a positive selection marker, such as a neomycin resistance marker; (c) a second nucleotide sequence included in the CanIon genomic sequence and (a) located in the genome downstream of the CanIon nucleotide sequence.
[208] In a suitable embodiment, the DNA construct consists of a negative selection marker located (a) upstream of the nucleotide sequence or (c) downstream of the nucleotide sequence. Suitably, the negative selection markers include thymidine kinase (tk) gene (Thomas et al., 1986), hygromycin beta gene (Te Riele et al., 1990), hprt gene (Van der Lugt et al., 1991; Reid et al., 1990) or diphtheria toxin A fragment (Dt-A) gene (Nada et al., 1993; Yagi et al. 1990). Preferably, a positive selection marker is placed within the CanIon exon sequence to break the sequence encoding the CanIon protein. These substitution vectors are described in Thomas et al. (1986; 1987); Mansour et al., (988); Koller et al. (1992).
[209] The first and second nucleotide sequences (a) and (c) may be independently positioned in a sequence comprising a CanIon control sequence, intron sequence, exon sequence, or control sequence, intron sequence and / or exon sequence. The size of the nucleotide sequences (a) and (c) is 1 to 50 kb, preferably 1 to 10 kb, more preferably 2 to 6 kb, most preferably 2 to 4 kb.
[210] DNA constructs enabling homologous recombination: Cre-LoxP system
[211] These novel DNA constructs utilize a site specific recombination system of P1 phage. P1 phage carry a recombinase called Cre that specifically interacts with the 34 base pair loxP. The loxP site consists of two 13 bp Palindrom sequences separated by an 8 bp conserved sequence (Hoess et al., 1986). Recombination by Cre enzyme between two co-orientated loxP sites results in deletion of the DNA fragment.
[212] The Cre-loxP system used in pairs with homologous recombination techniques was first described in Gu et al. (1993, 1994). In short, the nucleotide sequence inserted into the target location of the genome has at least two loxP sites located at each end of the nucleotide sequence that are coordinating and cleaved from the recombinant genome. Cleavage requires the presence of a recombinase ( Cre ) enzyme in the nucleus of the recombinant cell host. Recombinase enzymes can be prepared by (a) direct injection of Cre enzymes into the cells of interest, as found in Araki et al. (1995), or by lipofection of these enzymes into cells, as found in Baubonis et al. (1993). Culturing the recombinant cell host in a culture medium containing; (b) transfecting the host cell with a vector comprising a Cre coding sequence operably linked to a promoter functioning in a recombinant cell host, wherein the promoter is selectively induced in the cell host, and the vector is described in Gu et al. (1993) and Sauer et al. (1988) introduced into a recombinant cell host; (c) introducing a polynucleotide comprising a Cre coding sequence operably linked to a promoter functioning in a recombinant host cell into the genome of the cell host, wherein the promoter is selectively derived from the host cell and the polynucleotide is Gu et As described in al. (1994), random insertion or homologous recombination is inserted into the genome of the cell host.
[213] In certain embodiments, a vector comprising a sequence inserted into the CanIon gene by homologous recombination is constructed so that the selection markers flank the loxPs in the same orientation, and are inserted by homologous recombination while removing the selection markers with Cre enzyme treatment. It is possible to make the CanIon sequence survive. Once again, two selection markers are needed: a positive selection marker that selects recombination phenomena and a negative selection marker that selects homologous recombination phenomena. Vectors and methods using the Cre-loxP system are described in Zou et al. (1994).
[214] Thus, a preferred third DNA construct of the present invention is constructed from the 5'-end to the 3'-end as follows: (a) a first nucleotide sequence comprising the CanIon genomic sequence; (b) a nucleotide sequence comprising a polynucleotide encoding a positive selection marker, said nucleotide sequence further comprising two sequences defining a site recognized by the recombinase, eg, a loxP site, said two sites Is located in the same orientation; a second nucleotide sequence consisting of (c) a CanIon genomic sequence and (a) downstream of the CanIon nucleotide sequence in the genome.
[215] Suitably, the sequence defining the site recognized by the recombinase, eg, the loxP site, is present at (b) in the nucleotide sequence adjacent to the nucleotide sequence in which condition cleavage is desired. In certain embodiments, two loxP sites are located in each positive selection mark sequence to allow cleavage at the desired time point after homologous recombination.
[216] In a suitable embodiment of the method using the third DNA construct described above, the cleavage of the polynucleotide fragment flanked by two sites, preferably two loxP sites, recognized by the recombinase is described in Gu et al. (1994). As shown in the recombinant host cell genome, the Cre enzyme is operably linked to a promoter sequence, preferably an inducible promoter, more preferably a tissue-specific promoter sequence, most preferably an inducible and tissue-specific promoter sequence. It is executed at the desired time due to the presence of the encoding sequence.
[217] The presence of the Cre enzyme in the genome of a recombinant host cell is shown by Gu et al. (1994), which is a first transduction comprising two transgenic animals, a CanIon-derived sequence carrying a loxP site as described above. Animal and a second transgenic animal comprising a Cre coding sequence operably linked to a suitable promoter sequence.
[218] In addition, the spatial-temporal regulation of Cre enzyme expression, as identified by Graham (1995) and Kanegae et al. (1995), possesses the Cre gene to enable cellular infection or organ in vivo infection, and delivery of Cre enzyme. Adenovirus-based vectors can be achieved.
[219] The above-described DNA construct may be adapted to introduce a modified copy of a nucleotide sequence of the present invention, preferably a CanIon genomic sequence or a CanIon cDNA sequence, most preferably a CanIon genome or cDNA sequence, at a predetermined position in the target genome, to modify the target gene. It can be used to induce substitution of the target gene copy (hit homologous recombination) by another copy that is homologous enough to allow copy generation or homologous recombination to occur. In certain embodiments, the aforementioned DNA constructs can be used to introduce a CanIon genomic sequence or a CanIon cDNA sequence comprising at least one dual allele marker.
[220] Nuclear Antisense DNA Constructs
[221] Other compositions containing a vector of the invention comprise an oligonucleotide fragment of the nucleic acid sequence SEQ ID No 4, preferably a fragment comprising the initiation codon of the CanIon gene, as an antisense tool that inhibits the expression of the corresponding CanIon gene. Preferred methods of using other antisense polynucleotides in the present invention are those found in Sczakiel et al. (1995) or those disclosed in WO 95/24223.
[222] Suitably, the antisense tool is selected from polynucleotides (15-200 bp) complementary to the 5 'end of CanIon mRNA. In one embodiment, a combination of individual antisense polynucleotides complementary to different portions of the desired target gene is used.
[223] Preferred antisense polynucleotides according to the present invention are complementary to the sequence of the CanIon mRNA carrying the translation initiation codon ATG or the conjugation site. Another preferred antisense polynucleotide according to the present invention is complementary to the conjugation site of CanIon mRNA.
[224] Suitably, the antisense polynucleotides of the present invention have a 3 'polyadenylation signal that is substituted with a self-cleaving ribozyme sequence as identified by Liu et al. (1994), thus the RNA polymerase II transcript is 3'. There is no poly (A) at the end, and these antisense polynucleotides cannot be released into the nucleus. In a suitable embodiment, these CanIon antisense polynucleotides are ribozyme cassettes with histone stem-loop structures that stabilize cleaved transcripts from 3′-5 ′ exonuclease digestion, such as the structures identified in Eckner et al. (1991). Include in.
[225] Oligonucleotide Probes and Primers
[226] Polynucleotides derived from the CanIon gene are useful for detecting the presence of one or more copies of SEQ ID Nos. 1 to 4 and 6 nucleotide sequences, fragments, complements or variants thereof in a test sample.
[227] Particularly preferred probes and primers of the invention are at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 500 of SEQ ID Nos. , An isolated, purified, recombinant polynucleotide comprising a continuous span of 1000 or 2000 nucleotides or complement thereof. More preferred probes and primers of the invention carry an isolated, purified, recombinant polynucleotide wherein the continuous span comprises a biallelic marker selected from A1 to A17.
[228] Another object of the invention is an isolated, purified, recombinant nucleic acid comprising the nucleotide sequence of SEQ ID No 4, its complement sequence, allelic variants, fragments. In addition, preferred probes and primers of the invention carry a purified, isolated, recombinant CanIon cDNA comprising SEQ ID 4. Particularly preferred probes and primers of the invention are at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 500, 1000 of SEQ ID 4 Or, isolated, purified, recombinant polynucleotides comprising 2000, 3000, 4000, 5000, or 6000 contiguous nucleotides or complements thereof. More preferred probes and primers of the invention include at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 500, 1000 of SEQ ID 4 , Isolated, purified, recombinant polynucleotide comprising a contiguous span of 2000, 3000, 4000, 5000, or 6000 nucleotides or complement thereof, wherein the contiguous span comprises a biallelic marker selected from A1 to A17 do.
[229] In another embodiment, the probes and primers of the invention are at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300 of SEQ ID 6 Or an isolated, purified, recombinant polynucleotide comprising a continuous span of 400 nucleotides or complement thereof. In a suitable embodiment, the continuous span of SEQ ID No 6 comprises the biallelic marker A18.
[230] Thus, the present invention relates to nucleic acid probes that specifically hybridize with human CanIon nucleotide sequences of SEQ ID Nos. 1 and 3, complementary sequences or variants thereof under stringent hybridization conditions.
[231] In one embodiment, the invention includes an isolated, purified, recombinant polynucleotide consisting of a contiguous span of 8 to 50 nucleotides of one of SEQ ID Nos 1 to 4 and 6, wherein the span is present in the sequence. Retains a CanIon-associated bi-allelic marker, wherein the CanIon-related bi-allelic marker is selected from A1 to A18 and its complement, or optionally a heterologous allele marker. Optionally, the continuous span is 18 to 35 nucleotides and the dual allele marker is within 4 nucleotides from the polynucleotide center; Optionally, the continuous span is 25 nucleotides and a double allele marker is at the polynucleotide center; Optionally, the 3 'end of the continuous nucleotide is at the 3' end of the nucleotide; Optionally, the 3 'end of the continuous nucleotide is present at the 3' end of the nucleotide and the double allele marker is present at the 3 'end of the polynucleotide. In a suitable embodiment, the probe consists of a sequence selected from the following sequences: P1 to P18 and their complementary sequences.
[232] In another embodiment, the invention includes an isolated, purified, recombinant polynucleotide consisting of a contiguous span of 8 to 50 nucleotides of SEQ ID Nos. 1 to 4 or complement thereof, wherein the 3 'of the contiguous span The terminus is located at the 3 'end of the polynucleotide, and the 3' end of the polynucleotide is located within 20 nucleotides upstream of the CanIon-related double allele marker in the sequence; Optionally, the CanIon-related double allele marker is selected from A1 to A18 and its complement or, optionally, a dual allele marker of chain imbalance; Optionally, the 3 'end of the polynucleotide is located at 1 nucleotide upstream of the CanIon-related double allele marker in the sequence; Optionally, said polynucleotide consists of a sequence selected from the following sequences: D1 to D18 and E1 to E18.
[233] In another embodiment, the present invention includes an isolated, purified, recombinant polynucleotide consisting of the sequence selected from the following sequences: B1 to B17 and C1 to C17.
[234] In another embodiment, the present invention relates to an enzyme-based mismatch detection method for identifying the identity of nucleotides in hybridization assays, sequencing, CanIon-related double allele markers in SEQ ID Nos. 1 to 4 and 6 or complements thereof. Polynucleotides used and polynucleotides used to amplify segments of nucleotides comprising CanIon-related double allele markers in SEQ ID Nos. 1 to 4 and 6 or complements thereof.
[235] The present invention is directed to the identification of nucleotide entities in CanIon-related double allele markers, preferably to hybridization assays, sequencing, microsequence or enzyme-based mismatch detection assays and CanIon-related double allele markers. It relates to the use of a polynucleotide according to the invention in the amplification of a nucleotide segment comprising.
[236] Probes or primers according to the invention may comprise 8 to 1000 nucleotides, more specifically, at least 12, 15, 18, 20, 25, 35, 40, 50, 60, 70, 80, 90, 100, 250, 500, 1000 Has a nucleotide of. More specifically, the length of these probes and primers is 8, 10, 15, 20 or 30 to 100, preferably 10 to 50, more preferably 15 to 30 nucleotides. Shorter probes and primers tend to lack specificity for the target nucleic acid sequence and generally require lower temperatures to form a stable hybrid complex with the template. Longer probes and primers are expensive to make and can sometimes self-hybridize to form hairpin structures. Suitable lengths of primers and probes under certain assay conditions can be empirically determined by those skilled in the art. Suitable probes or primers are selected from the nucleotide sequences of P1 to P18 and their complementary sequences, B1 to B17, C1 to C17, D1 to D18, E1 to E18, wherein the individual positions in the sequence listing are shown in Tables 1, 2, and 3, respectively It consists of a nucleic acid comprising a polynucleotide.
[237] The formation of a stable hybrid depends on the melting point (Tm) of the DNA. Tm depends on the length of the primer or probe, the ionic power of the solution, and the G + C content. The higher the G + C content of the primer or probe, the higher the melting point, since the G: C pair has three hydrogen bonds while A: T has two hydrogen bonds. The GC content in the probe of the present invention is 10 to 75%, preferably 35 to 60%, more preferably 40 to 55%.
[238] Primers and probes may be prepared by any suitable method, such as cloning and restriction of suitable sequences and phosphodiester methods of Narang et al. (1979), phosphodiester methods of Brown et al. (1979), Beaucage et al. 1981) diethylphosphoramidite, a solid chemical synthesis method such as the solid support method disclosed in EP 0 707 592.
[239] Detection probes generally include nucleic acid sequences or uncharged nucleic acid analogs, for example peptide nucleic acids disclosed in international patent application WO 92/20702, US Pat. No. 5, 185,44; Morpholino analogs described in 5,034,506, 5,142,047. The probe is "not expandable" in that no additional dNTPs can be added. In essence, the analogs are non-expandable, and nucleic acid probes can be made non-expandable by modifying the 3 'end so that hydroxyl groups can no longer participate in the kidney. For example, the 3 'end of the probe can be functionalized with a capture or detection label to consume or block hydroxyl groups. Alternatively, the 3 ′ hydroxyl group can be cleaved, substituted or modified (U.S. Patent No. 07 / 049,061, filed April 19, 1993).
[240] The polynucleotides of the present invention can be labeled by incorporating labels known in the art that can be detected by microscopic, photochemical, biochemical, immunochemical or chemical means. For example, useful labels include radioactive materials (e.g. 32 P, 35 S, 3 H, 125 I), fluorescent dyes (e.g. 5-bromodiisoxiuridine, fluresin, acetylaminofluorene, deoxygenin) or Biotin is included. Suitably, the polynucleotide labels the 3 'and 5' ends. Non-radioactive labels of nucleic acid fragments are described in French Patent No. FR-7810975, Urdea et al (1988); Sanchez-Pescador et al. (1988). In addition, the probes according to the present invention may have structural features that allow for signal amplification, which may be, for example, branched chain DNA as disclosed in Urdea et al. (1991) or EP 0 225 807 (Chiron). Probe.
[241] Labels can be used to capture primers to facilitate fixation of primers or primer extension products, such as amplified DNA, to solid supports. The capture label is attached to a primer or probe, which may be a specific binding member that forms a binding pair with specific binding members of a solid phase reagent (eg, biotin and streptavidin). Therefore, depending on the type of label included by the polynucleotide or probe, it is possible to capture or detect the target DNA. In addition, the polynucleotides, primers or probes presented herein function as capture labels. For example, if the binding member of a solid phase reagent is a nucleic acid sequence, a capture label is selected that binds to the complementarity region of the primer or probe to immobilize the primer or probe on the solid phase. When the polynucleotide probe itself functions as a binding member, such probe carries a sequence or "tail" that is not complementary to the target. If the polynucleotide primer itself functions as a capture label, at least a portion of the primer freely hybridizes with the nucleic acid in the solid phase. DNA labeling techniques are known to those skilled in the art.
[242] The probes of the present invention are useful for many purposes. These can be used in particular for Southern hybridization to genomic DNA. Probes can also be used to detect PCR amplification products. They can also be used to detect mismatches in the CanIon gene or mRNA using other techniques. These may be used, for example, to detect the expression of the CanIon gene in northern blots.
[243] Polynucleotides, primers, probes of the present invention can be easily immobilized on a solid phase. Solid supports are known to those skilled in the art, including the well walls of test trays, test tubes, polystyrene beads, magnetic beads, nitrocellulose strips, membranes, microparticles (eg latex particles), sheep (or other animal) red blood cells, dura A duracyte and the like. Solid support is not critical and can be chosen by one skilled in the art. Thus, latex particles, microparticles, magnetic or non-magnetic beads, membranes, plastic tubes, walls of microtiter wells, glass or silicon chips, sheep (or other animal) red blood cells, durasite are all suitable. Suitable methods for immobilizing nucleic acids on solid phases include ionic bonds, hydrophobic bonds, covalent bonds, and the like. By solid support is meant herein a material that is insoluble or insoluble in a subsequent reaction. Solid supports can be selected for their internal ability to attract and immobilize capture reagents. Alternatively, the solid phase can carry additional receptors with the ability to attract and immobilize the capture reagents. Additional receptors may include the charged reagents themselves or oppositely charged materials with respect to the charged materials conjugated to the capture reagents. Alternatively, the receptor molecule may be any specific binding member that is immobilized (attached) to the solid support and has the ability to immobilize the capture reagent through a specific binding reaction. The receptor molecule allows indirect binding of the capture reagent to the solid support material prior to or during the assay. Thus, the solid phase may be a plastic, derived plastic, magnetic or non-magnetic material, glass or silicon surface of test tubes, microtiter wells, sheets, beads, microparticles, chips, red blood cells known to those skilled in the art, dura Site and other forms. The polynucleotides of the invention can be fixed or attached to a single solid support either individually to the solid support or as a collection of 2, 5, 8, 10, 12, 15, 20 or 25 individual polynucleotides according to the invention. In addition, other polynucleotides except the polynucleotides according to the present invention can be attached to the same solid support as one or a plurality of polynucleotides according to the present invention.
[244] As a result, the present invention relates to a method for detecting the presence of a nucleic acid comprising a nucleotide sequence selected from SEQ ID Nos. 1 to 4 and 6, a fragment or variant thereof, and a complementary sequence thereof in a sample. It consists of steps.
[245] a) contacting an assay sample with a nucleic acid probe or a plurality of nucleic acid probes that can hybridize with a nucleotide sequence comprised in a nucleic acid selected from the nucleic acids of SEQ ID Nos. 1-4 and 6, fragments or variants thereof, and complementary sequences thereof;
[246] b) Detecting the hybrid complex formed between the probe and the nucleic acid in the sample.
[247] The present invention also relates to a kit for detecting the presence of a nucleic acid comprising a nucleotide sequence selected from SEQ ID Nos. 1 to 4 and 6, a fragment or variant thereof, and a complementary sequence thereof in a sample, wherein the kit is as follows. It is composed of:
[248] a) a nucleic acid probe or a plurality of nucleic acid probes capable of hybridizing with a nucleotide sequence included in a nucleic acid selected from SEQ ID Nos. 1 to 4 and 6, fragments or variants thereof, and complementary sequences thereof;
[249] b) optionally, reagents necessary to carry out the hybridization reaction.
[250] In a suitable first embodiment of this detection method and kit, the nucleic acid probe or the plurality of nucleic acid probes is labeled with a detectable molecule. In a suitable second embodiment of the methods and kits, the nucleic acid probe or the plurality of nucleic acid probes is immobilized on a substrate. In a third embodiment, a nucleic acid probe or a plurality of nucleic acid probes is selected from P1 to P18 and their complementary sequences, B1 to B17, C1 to C17, D1 to D18, E1 to E18, or A1 to A18 and their complements. A sequence selected from the selected biallelic markers.
[251] Oligonucleotide Analysis
[252] A substrate comprising a plurality of oligonucleotide primers or probes according to the invention can be used to detect or amplify a target sequence in the CanIon gene, and can be used to detect mutations in the coding or non-coding sequences of the CanIon gene.
[253] Any of the polynucleotides set forth herein may be attached at overlapping regions or random locations in the solid support. Alternatively, the polynucleotides of the present invention can be attached to an ordered array, where each polynucleotide is attached to a separate position on a solid phase that does not overlap with the attachment position of another polynucleotide. Suitably, this ordered array of polynucleotides is "addressable" where the individual locations are recorded and accessible as part of the analysis process. Typically, an addressable polynucleotide array consists of a plurality of oligonucleotide probes that bind to the surface of a substrate at different known locations. Knowledge of the exact location of each polynucleotide makes these "addressable" arrays particularly useful for hybridization analysis. Addressable array techniques known in the art can be utilized for the polynucleotides of the present invention. Particular embodiments of such polynucleotide arrays are Genechips , which is described in US Patent 5,143,854; PCT International Applications WO 90/15070 and 92/10092 are generally described. These arrays can be made by mechanical synthesis methods or by light induced synthesis methods incorporating a combination of lithography and solid phase oligonucleotide synthesis (Fodor et al., 1991). Immobilization of oligonucleotide arrays on solid supports has been made possible by the development of "Very Large Scale Immobilized Polymer Synthesis" (VLSIPS ) technology, where the probes are immobilized on a solid surface of the chip in a high density array. Examples of VLSIPS technology are described in US Patent 5,143,854; 5,412,087; PCT Application WO 90/15070; WO 92/10092; WO 95/11995, which describes a method of forming oligonucleotide arrays through techniques such as light induced synthesis techniques. In preparing a strategy for providing nucleotide arrays immobilized on solid supports, additional presentation strategies have been developed for aligning and displaying oligonucleotide arrays on chips to maximize hybridization patterns and sequence information. Examples of such presentation strategies are described in PCT application WO 94/12305; WO 94/11530; WO 97/29212; It is disclosed in WO 97/31256.
[254] In another embodiment of the oligonucleotide array according to the invention, the oligonucleotide probe matrix can be usefully used to detect mutations occurring in the CanIon gene, preferably in its regulatory region. To this end, probes design specific probes with nucleotide sequences that hybridize to genes carrying known mutations (eg, deletion, insertion or addition of one or multiple nucleotides). Known mutation means a mutation in the CanIon gene identified according to techniques utilized by, for example, Huang et al. (1996) or Samson et al. (1996).
[255] Another technique used to detect mutations in the CanIon gene is high density DNA arrays. Each oligonucleotide probe constituting the unit element of a high density DNA array is designed to match a particular sequence of CanIon genomic DNA or cDNA. Therefore, an array comprising oligonucleotides complementary to the target gene sequence is used to confirm the homology of the wild gene sequence with the target sequence, to determine its content, and to detect differences between the reference wild type gene sequence of the target sequence and the CanIon gene. . In this design a 4L tiled array runs with a set of four probes (A, C, G, T), preferably 15-nucleotide oligomers. In each set of four probes, complete complement hybridizes more strongly than mismatched probes. As a result, nucleic acid targets of length L scan mutations into a tiled array containing 4L probes, and the entire probe set includes all possible mutations in known wild type reference sequences. Hybridization signals of the 15-mer probe set tiled arrays perturb a single base change in the target sequence. As a result, there is a characteristic disappearance or “footprint” of the signal in the probe adjacent to the mutation site. This technique is described in Chee et al. (1996).
[256] As a result, the present invention relates to an array of nucleic acid molecules comprising at least one of the aforementioned polynucleotides as a probe and a primer. Suitably, the present invention relates to an array of nucleic acid molecules comprising two or more of the aforementioned polynucleotides as probes and primers.
[257] Another object of the invention is P1 to P18, B1 to B17, C1 to C17, D1 to D18, E1 to E18, complementary sequences thereof, at least 8, 10, 12, 15, 18, 20, 25, 30 or 40 thereof. And an array of nucleic acid sequences comprising one or more sequences selected from at least one sequence having a dual allele marker selected from A1 to A18 and complements thereof.
[258] In addition, the present invention provides P1 to P18, B1 to B17, C1 to C17, D1 to D18, E1 to E18, complementary sequences thereof, at least 8, 10, 12, 15, 18, 20, 25, 30 or 40 thereof. It relates to an array of nucleic acid sequences comprising two or more sequences selected from contiguous nucleotide fragments and at least two sequences having a dual allele marker selected from A1 to A18 and complement thereof.
[259] CanIon Protein and Polypeptide Fragments
[260] As used herein, "CanIon polypeptide" includes all proteins and polypeptides of the invention. Polypeptides encoded by the polynucleotides according to the invention and fusion peptides comprising such polypeptides are also included in the invention. The present invention includes CanIon proteins derived from humans, including isolated or purified CanIon proteins consisting of the SEQ ID No 5 sequence.
[261] The present invention relates to polypeptides encoded by the nucleotide sequences of SEQ ID Nos. 1 to 4 and 6, complementary sequences or fragments thereof.
[262] The present invention includes at least six amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably 12, 15, 20, 25, 30, 40, 50, 100, 150, 200, 300, Isolated, purified, recombinant polypeptides comprising a continuous span of 400, 500, 700, 1000, 1200, 1400, 1600, or 1700 amino acids. In another suitable embodiment, the continuous span of amino acids comprises a site of mutation or functional mutation, including deletion, addition, substitution or truncation of the amino acid in the CanIon protein sequence.
[263] In a suitable embodiment, the present invention includes at least 6 amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably 12, 15, 20, 25, 30, 40, 50, 100, 150 Isolated, purified, recombinant polypeptides comprising a continuous span of 200, 300, 400, 500, 700, 1000, 1200, 1400, 1600, or 1700 amino acids, wherein the continuous span of amino acids is amino acid position 277 At least 1, 338, 574, 678, 680, 683, 691, 692, 695, 696, 894, 1480, 1481, 1483, 1484, 1485, 1630, 1631, 1632, 1636, 1660, 1667, 1707, 1709 Have 2, 3, 5 or 10 Suitably, the continuous span of SEQ ID No 5 comprises an alanine residue at position 277; Serine residues at position 338; Valine residues at position 574; Leucine residue at position 678; Serine residues at position 680; Threonine residue at position 683; Histidine residues at position 691; Serine residues at position 692; Alserine residue at position 695; An alanine residue at position 696; Isoleucine residues at position 894; Lysine residues at position 1480; An arginine residue at position 1481; Glycine residues at position 1483; Valine residues at position 1484; Isoleucine residues at position 1485; Asparagine residue at position 1630; A serine residue at position 1631; Methionine residues at the 1632 position; Threonine residue at position 1636; An alanine residue at position 1660; Phenylalanine residue at position 1667; Threonine residue at position 1707; An alanine residue at position 1709. Also shown are polynucleotides encoding one of these polypeptides.
[264] In addition, the present invention provides isolated, purified, comprising an amino acid sequence having at least 70, 75, 80, 85, 90, 95, 98, or 99% amino acid homology with the amino acid sequence of SEQ ID No 5, Recombinant polypeptides are included.
[265] The CanIon protein is preferably isolated from human or mammalian tissue samples or expressed from human or mammalian genes. CanIon polypeptides of the invention can be made by conventional methods known in the art. Polynucleotides encoding the desired polypeptides are ligated into an expression vector suitable for any convenient host. Eukaryotic and prokaryotic host systems are used to produce recombinant polypeptides. The polypeptide is then separated from the lysed cells or culture medium and purified to the level required for the desired purpose. Purification is carried out by any technique known in the art, for example fractional extraction, salt fractionation, chromatography, centrifugation and the like.
[266] In addition, shorter protein fragments are made by chemical synthesis. Alternatively, the proteins of the present invention are extracted from cells or tissues of humans or non-human animals. Methods of purifying proteins are known in the art, including particle destruction, fractional extraction and ion exchange chromatography, affinity chromatography, density settling, gel electrophoresis using detergents or chaotropic drugs. Isolation of the polypeptide by
[267] CanIon cDNAs, including SEQ ID No 4, can be used to express CanIon proteins and polypeptides. Nucleic acid encoding the expressed CanIon protein or polypeptide is operably linked to a promoter on the expression vector using conventional cloning techniques. The CanIon insert in the expression vector may consist of the entire coding sequence for the CanIon protein or a portion thereof.
[268] Expression vectors are mammalian, yeast, insect or bacterial expression systems known in the art. Commercially available vectors and expression systems are available from a variety of suppliers, including Genetics Institute (Cambridge, MA), Stratagenen (La Jolla, California), Promega (Madison, Wisconsin), Invitrogen (San Diego, California). If desired, the codon environment and codon pairs of sequences are optimized for the particular expression microorganism into which the expression vector is introduced to enhance expression and promote proper protein folding (U.S. Patent No. 5,082,767).
[269] In one embodiment, the entire coding sequence of CanIon cDNA is operably linked to a promoter on an expression vector via the poly A signal of the cDNA. Alternatively, in the absence of methionine serving as the initiation site in a nucleic acid encoding a portion of the CanIon protein, conventional techniques can be used to introduce the initiating methionine into the side at the first codon of the nucleic acid. Similarly, in the absence of a poly A signal in an insert derived from CanIon cDNA, the sequence cleaves the poly A signal from pSG5 (Stratagene) with the BglI and SalI restriction endonuclease enzymes and converts it to the mammalian vector pXT1 ( Can be integrated into a Stratagene to add to the construct. pXT1 comprises a portion of a gag gene derived from LRT and Moloney murine leukemia virus. The presence of LTR in the construct allows for effective and stable transfection. The vector carries a simple herpes thymidine kinase promoter and a selectable neomycin gene. The nucleic acid encoding the CanIon protein or portion thereof has a restriction endonuclease sequence for BglII at the 5 'end of Pst I and the corresponding cDNA 3' primer that bears the CanIon cDNA of SEQ ID No 5 and is incorporated into the 5 'primer From a bacterial vector carrying a, it is obtained by PCR with oligonucleotide primers complementary to the CanIon cDNA or a portion thereof, wherein the sequence encoding the CanIon protein or portion thereof is secured to be precisely located relative to the poly A signal. Purified fragments obtained by PCR reaction are cleaved with Pst I, blunt ended with exonuclease, cleaved with BglII, purified, ligated to pXT1 carrying a poly A signal and cleaved with BglII.
[270] The ligation product can be transfected into NIH 3T3 cells using lipofectin (Life Technologies, Inc., Grand Island, New York) under the conditions specified in the product specification. Positive transfectants are selected by culturing transfected cells in 600 ug / ml G418 (Sigma, St. Louis, Missouri).
[271] The process can also be utilized to express mutant CanIon proteins or portions thereof that are responsible for the detectable phenotype.
[272] The expressed protein can be purified by conventional purification techniques, such as ammonium sulfate precipitation or chromatographic separations based on size or charge. Proteins encoded by nucleic acid inserts can also be purified using standard immunochromatographic techniques. In this process, a solution containing the expressed CanIon protein or portion thereof, such as a cell extract, is added to a column with antibodies to the CanIon protein or portion thereof. The expressed protein is bound to an immunochromatography column. The column is then washed to remove non-specifically bound proteins. The specifically bound expressed protein is then released from the column and recovered by standard techniques.
[273] To confirm expression of a CanIon protein or portion thereof, a protein expressed from a host cell having an expression vector comprising an insert encoding the CanIon protein or portion thereof is expressed in a host cell having an expression vector without the insert. Can be compared with protein. The presence of a band in a sample of cells carrying an expression vector carrying an insert that does not appear in a sample of cells carrying an expression vector without an insert suggests that the CanIon protein or portion thereof is expressed. Generally, such bands have the expected mobility in CanIon proteins or portions thereof. However, the band may have a different mobility than expected as a result of modifications such as glycosylation, ubiquitation or enzymatic cleavage.
[274] Antibodies that can specifically recognize expressed CanIon proteins or portions thereof are described below.
[275] If antibody production is not possible, the nucleic acid encoding the CanIon protein or portion thereof is incorporated into an expression vector designed for use in purification schemes utilizing chimeric polypeptides. In this strategy, nucleic acids encoding CanIon proteins or portions thereof are inserted in alignment with the gene encoding the other half of the chimera. The other half of the chimeras are β-globin or nickel binding polypeptide encoding sequences. The chromatography matrix with the antibody against β-globin or nickel attached thereto is then used to purify the chimeric protein. The protease cleavage site is engineered between the β-globin gene or nickel binding polypeptide and the CanIon protein or a portion thereof. Thus, the two polypeptides of the chimera are separated from each other by protease cleavage.
[276] An expression vector useful for making β-globin chimeric proteins is pSG5 (Stratagene), which encodes rabbit β-globin. Intron II of the rabbit β-globin gene promotes cleavage of the expressed transcript and the polyadenylation signal integrated into the construct increases the level of expression. These techniques are known to those skilled in the art of molecular biology. Standard methods are published in method texts such as Davis et al. (1986), and many methods are available from Stratagene, Life Technologies, Inc., or Promega. Polypeptides can also be made from constructs using in vitro translation systems such as the In vitro Express Translation Kit (Stratagene).
[277] Antibodies That Bind to CanIon Polypeptides of the Invention
[278] The CanIon polypeptide or whole protein can be used to make an antibody that can specifically bind an expressed CanIon protein or fragment thereof.
[279] The antibody composition of the present invention may specifically or selectively bind to the CanIon protein of SEQ ID No 5. The antibody composition that specifically binds to the first variant of CanIon is at least 5%, 10%, relative to the full length first variant of CanIon protein over the full length second variant of CanIon protein in ELISA, RIA or other antibody-based binding assays. 15%, 20%, 25%, 50% or 100% higher binding affinity should be shown. In suitable embodiments, the antibody composition may specifically bind to human CanIon protein.
[280] In a suitable embodiment, the invention relates to at least 6 amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably 12, 15, 20, 25, 30, 40, 50, 100, 200 It relates to a polyclonal or monoclonal antibody composition that specifically binds to an epitope-containing polypeptide comprising a continuous span of 300, 400, 500, 700 or 1000 amino acids. In a suitable embodiment, the continuous span is amino acid positions 277, 338, 574, 678, 680, 683, 691, 692, 695, 696, 894, 1480, 1481, 1483, 1484, 1485, 1630, of SEQ ID No 5 At least 1, 2, 3, 5 or 10 of 1631, 1632, 1636, 1660, 1667, 1707, 1709.
[281] Any CanIon polypeptide or whole protein can be used to make an antibody that can specifically bind an expressed CanIon protein or fragment thereof as disclosed herein.
[282] Epitopes consist of at least three amino acids that give the epitope a unique spatial conformation. Generally, the epitope consists of at least six such amino acids, preferably at least 8-10 such amino acids. In a suitable embodiment, the antigenic epitope consists of 3 to 50 amino acids. Fragments that function as epitopes can be made by any conventional means. Epitopes can be identified by Jameson-Wolf antigen analysis using the PROTEAN computer program (Version 4.0 Windows, DNASTAR, Inc., 1228 South Park Street Madison, Wis.) Using default variables.
[283] The invention also relates to purified or isolated antibodies, fragments or variants thereof that are composed of epitopes of mutated CanIon proteins and specifically bind to mutated CanIon proteins. In another suitable embodiment, the invention relates to an antibody capable of binding to a polypeptide consisting of at least 10 contiguous amino acids of the CanIon protein and having at least one amino acid that can be encoded by a transgenic mutation.
[284] Animals other than humans or mammals expressing a different kind of CanIon than those in which antibody binding is required and animals that do not express CanIon (ie, CanIon hitting animals) are particularly useful for making antibodies. CanIon-targeted animals recognize most of the exposed regions of the CanIon protein as foreign antigens, thus making antibodies with a broader CanIon epitope. In addition, smaller polypeptides having 10 to 30 amino acids may be useful for achieving specific binding to any one of the CanIon proteins. In addition, the humoral immune system of an animal producing a kind of CanIon similar to this antigenic sequence will recognize the difference between the native CanIon and the antigenic sequence and will produce antibodies to unique sites in the antigenic sequence. This technique is particularly useful for obtaining antibodies that specifically bind to any one of the CanIon proteins.
[285] Antibody formulations made by this protocol are useful for quantitative immunoassays to measure the concentration of antigen-bearing material in biological samples; They are also used to semi-quantitatively or qualitatively identify the presence of antigen in a biological sample. Antibodies can also be used in therapeutic compositions that kill cells or reduce levels of protein in the body.
[286] Antibodies of the invention can be labeled with radioactive, fluorescent or enzymatic labels known in the art.
[287] Accordingly, the present invention relates to a method for specifically detecting the presence of a CanIon polypeptide according to the present invention in a biological sample, which comprises the following steps:
[288] a) contacting a biological sample with a polyclonal or monoclonal antibody consisting of the amino acid sequence of SEQ ID No 5 and specifically binding to a CanIon polypeptide, peptide fragment or variant thereof;
[289] b) detect the antigen-antibody complex formed.
[290] The invention also relates to a diagnostic kit for detecting the in vitro presence of a CanIon polypeptide according to the invention in a sample, the kit consisting of:
[291] a) a polyclonal or monoclonal antibody, peptide fragment or variant thereof consisting of the amino acid sequence of SEQ ID No 5 and which specifically binds to a CanIon polypeptide;
[292] b) reagents which allow detection of the antigen-antibody complexes formed, which reagents optionally carry a label, or, in particular, if the aforementioned monoclonal or polyclonal antibodies are not self-labeled, I can recognize it.
[293] Accordingly, the present invention relates to antibodies and T-cell antigen receptors (TCRs), which include polypeptides, more specifically IgG (including IgG1, IgG2, IgG3, IgG4), IgA (including IgA1, IgA2), IgD, IgE, IgM And specifically binds to an epitope of a polypeptide according to the invention, including but not limited to those including IgY. In a suitable embodiment, the antibody is a human antigen binding antibody fragment of the invention, which includes Fab, Fab ', F (ab) 2, F (ab') 2, Fd, single-chain Fvs (scFv), single-chain Fragments comprising antibodies, disulfide-linked Fvs (sdFv), V L or V H are included, but are not limited to these. The antibody may be derived from any animal, including birds and mammals. Suitably the antibody is derived from human, murine, rabbit, goat, guinea pig, camel, horse or chicken.
[294] Antigen-binding antibody fragments, including single-chain antibodies, may consist of the variable region alone or with all or a portion of the following regions: hinge region, CH1, CH2, CH3 domains. The present invention also encompasses any combination of variable and hinge regions, CH1, CH2, CH3 domains. The present invention also includes chimeric, print, human monoclonal and polyclonal antibodies that specifically bind to the polypeptides of the invention. The invention also includes anti-idiotype antibodies to the antibodies of the invention.
[295] Antibodies of the invention may be monospecific, bispecific, trispecific or multispecific. Multispecific antibodies may be specific for different epitopes of polypeptides according to the invention or specific to both the polypeptides according to the invention and to heterogeneous compositions such as heterologous polypeptides or solid support substances (see WO 93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, A. et al. (1991) J. Immunol. 147: 60-69; US Patents 5,573,920, 4,474,893, 5,601,819, 4,714,681, 4,925,648; Kostelny, SA et al. (1992) J. Immunol. 148: 1547-1553).
[296] Antibodies of the invention are described in terms of the epitope or epitope-bearing region of a polypeptide according to the invention that is recognized or specifically binds to the antibody. In the case of a protein according to the invention, the antibody is a full-length protein encoded by the nucleic acid of the invention, a mature protein encoded by the nucleic acid of the invention (ie, a protein produced by cleavage of a signal peptide), of the invention It can specifically bind to a signal peptide encoded by a nucleic acid or any other polypeptide of the invention. Thus, epitopes carrying such epitopes or portions of polypeptides can be described, for example, by the N-terminal and C-terminal positions, the size of contiguous amino acid residues, and the like. Antibodies that specifically bind epitopes or polypeptides of the invention can also be distinguished individually. Accordingly, the present invention relates to antibodies that specifically bind to and enable discrimination of certain polypeptides according to the present invention.
[297] Antibodies of the invention may be described in terms of cross-reactivity. Antibodies that do not specifically bind to any other analog, ortholog or homolog of a polypeptide according to the invention are included in the invention. Less than 95%, less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, less than 50% homology with the polypeptides of the present invention Antibodies that do not bind to a polypeptide having a value measured by a method known in the art are also included in the present invention. In addition, the present invention includes binding antibodies only to polypeptides encoded by polynucleotides that hybridize with the polynucleotides of the present invention under stringent hybridization conditions. Antibodies of the invention may be described in terms of binding affinity. The preferred binding affinity antibodies separation constant (Kd) is 5X10 -6 M, 10 -6 M, 5X10 -7 M, 10 -7 M, 5X10 -8 M, 10 -8 M, 5X10 -9 M, 10 -9 M, 5X10 -10 , 10 -10 M, 5X10 -11 M, 10 -11 M, 5X10 -12 M, 10 -12 M, 5X10 -13 M, 10 -13 M, 5X10 -14 M, 10 -14 M a 5X10 -15 M, 10 -15 M is less than the antibody.
[298] Antibodies of the invention are used in methods known in the art to purify, detect, and target polypeptides according to the invention, including in vitro and in vivo diagnostic and therapeutic methods. For example, antibodies are used in immunoassays to quantitatively and qualitatively measure levels of polypeptides according to the invention in biological samples (Harlow et al., ANTIBODIES: A LABORATORY MANUAL, Cold Spring Harbor Laboratory Press, 2nd ed. 1988).
[299] Antibodies of the invention can be used alone or in combination with other compositions. Antibodies can be recombinantly fused to heterologous polypeptides at the N- or C-terminus or chemically conjugated (including covalent and non-covalent conjugates) to polypeptides or other compositions. For example, antibodies of the invention can be recombinantly fused or conjugated to molecules and effector molecules useful as labels in detection assays, such as heterologous polypeptides, drugs or toxins (WO 92/08495; WO 91/14438; WO 89). / 12624; US Patent 5,314,995; EP 0 396387).
[300] Antibodies of the invention can be made by any suitable method known in the art. For example, the polypeptides of the invention or antigenic fragments thereof can be administered to an animal and used to induce the production of serum containing polyclonal antibodies. "Monoclonal antibodies" are not limited to antibodies made through hybridoma technology. "Antibody" refers to a polypeptide or group of polypeptides comprising one or more binding domains, wherein the antibody binding domain is generated by the folding of various domains in an antibody molecule and is complementary to the antigenic determinant appearance of the antigen and It forms a three-dimensional binding space with a charge distribution, which allows for an immunological reaction with the antigen. "Monoclonal antibody" means an antibody derived from a single clone, including eukaryotic, prokaryotic or phage clones, and does not mean a method of producing it. Monoclonal antibodies can be made using a variety of techniques known in the art, including hybridoma use, recombination, and phage display techniques.
[301] Hybridoma techniques are known in the art (Harlow et al. (1988); Hammerling, et. Al. (1981)). Fab and F (ab ') 2 fragments can be made from hybridoma-generated antibodies by proteolytic cleavage, for example, using enzymes such as papain (producing Fab fragments) or pepsin (producing F (ab') 2 fragments). .
[302] Alternatively, the antibodies of the invention can be made by applying recombinant DNA techniques or synthetic chemistry known in the art. For example, the antibodies of the invention can be made by various phage display methods known in the art. In phage display methods, functional antibody domains are displayed on the surface of phage particles, which phage particles carry a polynucleotide sequence that encodes them. Phage with the desired binding properties are selected from repertoires or combinatorial antibody libraries (eg, human or murine) by direct selection with antigens, in particular antigens that are bound or captured to solid surfaces or beads. Typically, phage used in these methods are fibrous phages including fd and M13 with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to phage gene III or gene VII protein. Phage display methods that can be used to make the antibodies of the invention are described in Brinkman U. et al. (1995); Ames, R.S. et al. (1995); Kettleborough, C. A .. et al. (1994); Persic, L. et al. (1997); Burton, D. R. et al. (1994); PCT / GB91 / 01134; WO 90/02809; WO 91/10737; WO 92/01047; WO 92/18619; WO 93/11236; WO 95/15982; WO 95/20401; US Patents 5,698,426, 5,223,409, 5,403,484, 5,580,717, 5,427,908, 5,750,753, 5,821,047, 5,571,698, 5,427,908, 5,516,637, 5,780,225, 5,658,727, 5,733,743.
[303] As noted in the above references, after phage screening, antibody coding regions are isolated from phage, which are whole antibodies expressed in mammalian cells, insect cells, plant cells, yeast, bacteria, including human antibodies or any other antigen binding fragment. Can be used to create For example, techniques for recombinantly producing Fab, Fab'F (ab) 2, F (ab ') 2 fragments can be utilized by methods known in the art (WO 92/22324; Mullinax, RL et al. (1992). Sawai, H. et al. (1995); Better, M. et al. (1988)).
[304] Techniques that can be used to produce single-chain Fvs and antibodies are described in U.S. Pat. Patents 4,946,778 and 5,258,498; Huston et al. (1991); Shu, L. et al. (1993); Skerra, A. et al. (1988). In some applications, including in vivo use of antibodies in humans and in vitro detection assays, it may be desirable to use chimeric, print or human antibodies (Morrison (1985); Oi et al., (1986); Gillies, SD et). al. (1989); US Patent 5,807,715). Antibodies can be produced in a variety of techniques, for example CDR-tissue transplantation (EP 0 239 400; WO 91/09967; US Patent 5,530,101; 5,585,089), veneering or resurfacing (EP 0 592 106; EP 0 519 596 Padlan EA, (1991); Studnicka GM et al. (1994); Roguska MA et al. (1994)), chain shuffling (US Patent 5,565,332). Human antibodies can be made by a variety of methods known in the art, including the phage display method described above (US Patents 4,444,887, 4,716,111, 5,545,806, 5,814,318; WO 98/46645; WO 98/50433; WO 98/24893; WO 96 / 34096; WO 96/33735; WO 91/10741).
[305] In addition, the present invention includes antibodies that have been recombinantly fused or chemically conjugated (including covalent and non-covalent conjugates) to a polypeptide of the present invention. The antibody may be specific for an antigen other than the polypeptide of the present invention. For example, an antibody can be used to direct a polypeptide of the invention to a particular cell type in vitro or in vivo by fusing or conjugating the polypeptide of the invention to an antibody specific for a particular cell surface receptor. Antibodies fused or conjugated to a polypeptide of the invention can also be used for in vitro immunoassays and purification by methods known in the art (Harbor et al. Supra and WO 93/21232; EP 0 430 095; Naramura M. etal (1994); US Patent 5,474, 981; Gillies, SO et al. (1992); Fell, HP et al. (1991).
[306] The invention also relates to a composition comprising a polypeptide of the invention fused or conjugated to a domain other than a variant region. For example, a polypeptide of the invention can be fused or conjugated to an antibody Fc region or portion thereof. A portion of an antibody fused to a polypeptide of the invention may consist of a hinge region, a CH1 domain, a CH2 domain, a CH3 domain, any combination of whole domains, or portions thereof. Polypeptides of the invention may be fused or conjugated with a portion of an antibody to increase the half-life of the polypeptide or for use in immunoassays using known methods. The polypeptide may also be fused or conjugated with a portion of the antibody to form a multimer. For example, an Fc region fused to a polypeptide of the invention can form a dimer via disulfide bonds between Fc regions. Higher multimeric forms can be made by fusing the polypeptides of the invention to a portion of IgA and IgM. Methods for fusion or conjugation of a polypeptide of the invention to a portion of an antibody are known in the art (US Patents 5,336,603, 5,622,929, 5,359,046, 5,349,053, 5,447,851, 5,112,946; EP 0 307 434, EP 0 367 166; WO 96/04388, WO 91/06570; Ashkenazi A. et al. (1991); Zheng, XX et al. (1995); Vil, H. et al. (1992).
[307] The invention also relates to antibodies which act as agonists or antagonists of the polypeptides according to the invention. For example, the present invention includes antibodies that completely or partially disrupt receptor / ligand interactions with polypeptides according to the present invention. Receptor-specific antibodies and ligand-specific antibodies are also included in the present invention. In addition, the present invention includes receptor-specific antibodies that prevent receptor activation without interfering with ligand binding. Receptor activation (ie signal transduction) can be confirmed by techniques known in the art. The present invention also encompasses receptor-specific antibodies that block both ligand binding and receptor activation. Similarly, the present invention includes neutralizing antibodies that bind to a ligand and block binding of the receptor to the ligand and antibodies that bind to the ligand to block receptor activation but do not block the ligand from binding to the receptor. In addition, the present invention includes antibodies that activate the receptor. These antibodies may also serve as agents for biological activity that are affected by ligand-mediated receptor activation. Such antibodies may be described as agents or antagonists for biological activity, including the specific activities disclosed herein. Such antibody agonists can be made by methods known in the art (WO 96/40281; US Patent 5,811,097; Deng B. et al. (1998); Chen, Z. et al. (1998); Harrop JA et al. (1998); Zhu, Z. et al. (1998); Yoon, DY et al. (1998); Prat, M. et al. (1998); Pitard, V. et al. (1997); Liautard, J et al. (1997); Carlson, NG et al. (1997); Taryman, RE et al. (1995); Muller, YA et al (1998); Bartunek, P. Et al. (1996)).
[308] As noted above, antibodies of polypeptides according to the invention can be used to make anti-idiotype antibodies that "imitate" the polypeptides of the invention by techniques known in the art (Greenspan and Bona, (1989); Nissinoff , (1991)). For example, an antibody that competitively inhibits polypeptide multimerization or binding of a polypeptide and a ligand according to the invention by binding "imitates" a polypeptide multimerization or binding domain and consequently binds to a polypeptide or ligand thereof to neutralize them. Can be used to make anti-idiotype antibodies. Such neutralizing anti-idiotype antibodies can be used to bind to the polypeptides of the invention or their ligand / receptor binding to block their biological activity.
[309] CanIon-Related Double Allele Markers
[310] Advantages of Dual Allele Markers According to the Invention
[311] The CanIon-related double allele markers of the present invention provide a number of important advantages over other genetic markers, such as RFLP (limited fragment length polymorphism) and VNTR (variable total number of serial repeats) markers.
[312] The first generation marker is RFLP, which is a variable that changes the length of the restriction fragment. However, the methods used to identify and classify RFLPs consume a relatively large amount of material, effort, and time. The second generation genetic marker is VNTR, which can be classified as either minisatellite or microsatellite. Minisatellites are serially repeated DNA sequences present in 5-50 repeating units distributed along a region of human chromosomes of 0.1-20 Kilobase length. Because they provide many possible alleles, their information content is valuable. The minisatellite is recorded by performing a Southern blot confirming the total number of serial sequences present in the nucleic acid sample obtained from the test subject. However, only 10 4 potential VNTRs can be classified as Southern blotting. In addition, RELP and VNTR are costly and time consuming to develop and analyze in bulk.
[313] Single nucleotide polymorphisms or dual allelic markers can provide a variety of benefits while being used in the same manner as RELP and VNTR. SNPs are intensively located in the human genome and possible frequent forms of variation. Approximately 10 7 or more sites are dispersed along the 3 × 10 9 base pairs of the human genome. Thus, SNPs occur at a higher frequency and uniformity than RFLP or VNTR markers, meaning that these markers are more likely to be found close to the locus of interest.
[314] In addition, other forms of characterized single nucleotide polymorphisms, such as the dual allelic markers of the invention, are easier to distinguish and can therefore be easily classified on a routine basis. Dual allele markers carry a single nucleotide based allele and only two common alleles, which allow for very parallel detection and automated recording. The dual allele marker of the present invention allows for rapid high performance genotyping of many individuals.
[315] Biallelic markers are concentrated in the genome and are sufficiently informative and can be analyzed in large quantities. Due to the combined effects of these benefits, double allele markers are extremely important for genetic studies. Double allelic markers can be used for chain studies in families, allelic sharing methods, chain imbalance studies in populations and association studies of case-control populations or trait positive and trait negative populations. An important aspect of the present invention is that linkage studies identifying genes involved in complex traits can be carried out via double allele markers. Linkage studies examine the frequency of marker alleles in irrelevant case- and control-populations and are generally used for detection of polygenic or sporadic traits. Association studies can be conducted in the entire population and are not limited to studies conducted on related individuals in affected families (serial studies). Double allele markers in different genes can screen in parallel for direct association with disease or response to treatment. This multigene approach is a powerful tool for human genetic research in that it provides the statistical power needed to examine the synergistic effects of multiple genetic factors on disease states with specific phenotypes, drug responses, sporadic traits, or complex gene pathogenesis. to be.
[316] Candidate Genes of the Invention
[317] Different approaches can be used to implement the association method: genome-wide association studies, candidate region association studies, candidate gene association studies. Genome-wide association studies rely on the screening of genetic markers that are uniformly distributed and cover the entire genome. Candidate genetic approaches are based on the study of genetic markers that are specifically located in genes potentially involved in biological pathways associated with the trait of interest. In the present invention, CanIon is a candidate gene. Such candidate gene analysis provides a shortcut to the identification of genes and polymorphisms associated with specific traits when some information about the ecology of the trait is available. However, all dual allelic markers disclosed herein can be utilized as part of genome-wide association studies or candidate region association studies, which utilization is specifically defined in the present invention and claims.
[318] CanIon-related dual allele markers and related polynucleotides
[319] The present invention also relates to CanIon-related double allele markers. By “CanIon-related double allele marker” is meant herein a group of double allele markers that are in chain imbalance with the CanIon gene. CanIon-related biallelic markers include biallelic markers designated A1 to A17.
[320] Portions of the dual allele markers according to the present invention are disclosed in Table 2. These are described as single base polymorphisms in related 1 to 4 and 6. Primer pairs that allow for amplification of nucleic acids bearing the polymorphic bases of one CanIon allele marker are shown in Table 1 of Example 2.
[321] Seventeen CanIon-related biallelic markers, A1 to A17, are located in the genomic sequence of CanIon. Double allelic markers A12 and A16 are located in the exon of CanIon. Double allele marker A18 flanks the CanIon gene.
[322] The present invention also relates to purified and / or isolated nucleotide sequences comprising the polymorphic bases of the CanIon-related biallelic markers. In suitable embodiments, the dual allele marker is selected from A1 to A18, complements thereof. The sequence is 8 to 1000 nucleotides in length, preferably at least 8, 10, 12, 15, 18, 20, 25, 35, of the nucleotide sequence selected from SEQ IDs 1 to 4 and 6, variants or complementary sequences thereof. , Continuous spans of 40, 50, 60, 70, 80, 100, 250, 500, 1000 nucleotides. These nucleotide sequences comprise polymorphic bases of either allele 1 or 2 of the double allele marker. Optionally, the allele marker may be located within 6, 5, 4, 3, 2, 1 nucleotides from the center of the polynucleotide, or at the center of the polynucleotide. Optionally, the 3 'end of the span may be present at the 3' end of the polynucleotide. Optionally, a dual allele marker can be present at the 3 'end of the polynucleotide. Optionally, the polynucleotide may further comprise a label. Optionally, the polynucleotide can be attached to a solid support. In other embodiments, the aforementioned polynucleotides may be used alone or in any combination.
[323] The invention also relates to 8 to 1000 contiguous nucleotides of a nucleotide sequence selected from SEQ ID 1 to 4 or complementary sequences thereof, preferably at least 8, 10, 12, 15, 18, 20, 25, 35, 40, It relates to purified and / or isolated nucleotide sequences comprising a continuous span of 50, 60, 70, 80, 100, 250, 500, 1000 nucleotides. The 3 'end of the nucleotide is within a CanIon-related bi-allelic marker in the sequence or at least 2, 4, 6, 8, 10, 12, 15, 18, 20, 25, 50, 100, 250, 500 or And may be located upstream of 1000 nucleotides. Optionally, the CanIon-related double allele marker is selected from A1 to A17; Optionally, the 3 'end of the polynucleotide may be located one nucleotide upstream of the CanIon-related double allele marker in the sequence. Optionally, the polynucleotide may further comprise a label. Optionally, the polynucleotide can be attached to a solid support. In other embodiments, the aforementioned polynucleotides may be used alone or in any combination.
[324] In a suitable embodiment, the sequence comprising the polymorphic base of one of the dual allelic markers listed in Table 2 has a continuous span of polynucleotides selected from the nucleic acid, variant or complementary sequence of the amplicon sequence listed in Table 1 Is selected from a nucleotide sequence comprised of, or comprising, such polynucleotides.
[325] The present invention also relates to a nucleic acid encoding a CanIon protein, wherein the nucleic acid comprises a polymorphic base of a double allele marker selected from A12 and A16 and its complement.
[326] The present invention includes any polynucleotide used to identify the identity of one or more nucleotides in a CanIon-related dual allele marker, or the use of such polynucleotides. In addition, the polynucleotides of the present invention used to identify the identity of one or more nucleotides in a CanIon-related double allele marker include the single or combined polynucleotides disclosed herein without further limitation. Optionally the CanIon-related double allele marker is selected from A1 to A18 and its complement, or optionally a dual allele marker that is chain imbalanced thereto; Optionally, the CanIon-associated biallelic marker is selected from A1 to A17 and its complement, or optionally a biallelic marker that is chain imbalanced thereto; Optionally, the CanIon-related double allele marker is selected from A12 and A16 and its complement, or optionally a dual allele marker that is chain imbalanced thereto; Optionally, the polynucleotide comprises a single or combined sequence disclosed herein; Optionally, the polynucleotide consists of any of the polynucleotides disclosed herein; Alternatively, the identification can be performed by hybridization assay, sequencing, microsequence analysis, or enzyme-based mismatch detection assay; Optionally, the polynucleotide can be attached to a solid support, array, or addressable array; Optionally, the polynucleotide can be labeled. Appropriate polynucleotides can be used in hybridization assays to identify the identity of nucleotides in CanIon-related double allele markers. Other suitable polynucleotides can be used for sequencing or microsequence analysis to identify the identity of a nucleotide in a CanIon-related double allele marker. Other suitable polynucleotides can be used in enzyme-based mismatch detection assays to identify the identity of a nucleotide in a CanIon-related double allele marker. Other suitable polynucleotides can be used to amplify segments of polynucleotides that include CanIon-related double allele markers. Optionally, the polynucleotide can be attached to a solid support, array, or addressable array; Optionally, the polynucleotide can be labeled.
[327] The present invention also encompasses any polynucleotide used for amplifying a nucleotide comprising a CanIon-related dual allele marker, or the use of such polynucleotide. In addition, the polynucleotides of the present invention used to amplify segments of nucleotides comprising CanIon-related dual allele markers include the single or combined polynucleotides disclosed herein without further limitation. Optionally the CanIon-related double allele marker is selected from A1 to A18 and its complement, or optionally a dual allele marker that is chain imbalanced thereto; Optionally, the CanIon-associated biallelic marker is selected from A1 to A17 and its complement, or optionally a biallelic marker that is chain imbalanced thereto; Optionally, the CanIon-related double allele marker is selected from A12 and A16 and its complement, or optionally a dual allele marker that is chain imbalanced thereto; Optionally, the polynucleotide comprises a single or combined sequence disclosed herein; Optionally, the polynucleotide consists of any of the polynucleotides disclosed herein; Alternatively, amplification can be by PCR or LCR; Optionally, the polynucleotide can be attached to a solid support, array, or addressable array; Optionally, the polynucleotide can be labeled.
[328] Primers for the amplification or sequencing of polynucleotides comprising the double allele marker of the present invention can be designed from sequences known in the art. The 3 'end of the continuous span homologous to the sequences selected from SEQ IDs 1 to 4 and 6, complementary sequences or variants thereof in the appropriate primer set is present at the 3' end of the primer. This form allows the 3 'end of the primer to hybridize with the selected nucleic acid sequence and dramatically increases the efficacy of the primer for amplification or sequencing. Allele specific primers can be designed such that the polymorphic base of the biallelic marker is located at the 3 'end of the continuous span and the continuous span is at the 3' end of the primer. Such allele specific primers tend to selectively amplify amplification or sequencing reactions when used with a nucleic acid sample containing one of the two alleles present in a double allele marker. The 3 'end of the primer according to the present invention may be at or within the CanIon-related double allele marker or at any other position suitable for use in the sequence or for sequencing, amplification, positioning of a new sequence or marker. 2, 4, 6, 8, 10, 12, 15, 18, 20, 25, 50, 100, 250, 500 or 1000 nucleotides upstream. Thus, another set of suitable amplification primers comprises an isolated polynucleotide consisting of a contiguous span of 8 to 50 nucleotides in a sequence selected from SEQ IDs 1 to 4 and 6, complementary sequences or variants thereof. The 3 'end of the span is located at the 3' end of the polynucleotide, and the 3 'end of the polynucleotide is located upstream of the CanIon-related dialle marker in the sequence. Suitably these amplification primers comprise a sequence selected from sequences B1 to B17 and C1 to C17. Primers positioned 3 ′ upstream of one nucleotide of the canonal allele marker are particularly useful for microsequence analysis. Suitable microsequence primers are described in Table 4. Optionally, the CanIon-associated biallelic marker is selected from A1 to A18 and its complement, or optionally a biallelic marker that is chain imbalanced thereto; Optionally, the CanIon-associated biallelic marker is selected from A1 to A17 and its complement, or optionally a biallelic marker that is chain imbalanced thereto; Optionally, the CanIon-related double allele marker is selected from A12 and A16 and its complement, or optionally a dual allele marker that is chain imbalanced thereto; Optionally, the microbase primer is selected from nucleotide sequences D1 to D18 and E1 to E18.
[329] Probes of the present invention can be designed from methods known in the art, particularly those capable of examining the presence of the markers disclosed herein. Suitable probe sets can be designed for use in the hybridization assays of the present invention in a manner known in the art that selectively binds to one allele of a dual allele marker but does not bind to another allele under any particular assay conditions. . Suitable hybridization probes include polymorphic bases of either allele 1 or 2 of the double allele marker. Optionally, the allele marker may be located within 6, 5, 4, 3, 2, 1 nucleotides from the center of the polynucleotide, or at the center of the polynucleotide. In a suitable embodiment, the probe is selected from each of sequences P1 through P18 and each of its complementary sequences.
[330] The polynucleotides of the present invention are not limited to having the exact flanking sequence surrounding the polymorphic bases specified in the sequence listing. Rather, the flanking sequences surrounding the biallelic marker can be lengthened or shortened for some time depending on the intended use, and the present invention specifically defines such sequences. Lateral areas outside the continuous span should not be homologous to the native side areas that actually occur in human subjects. The addition of any nucleotide sequence that is compatible with the intended nucleotide is specifically incorporated.
[331] Primers and probes can be labeled or immobilized to a solid support as identified in "oligonucleotide probes and primers".
[332] Polynucleotides of the invention attached to a solid support include, without limitation, single or combined polynucleotides disclosed herein: Optionally, the polynucleotides are individually in a single solid support or at least 2, 5 according to the invention. , 8, 10, 12, 15, 20 or 25 different polynucleotides can be attached. Optionally, a polynucleotide other than the polynucleotide according to the present invention may be attached to the same solid support as in the polynucleotide according to the present invention. Optionally, when multiple polynucleotides are attached to a solid support, they can be attached at random positions, or in an aligned array. Optionally, the aligned array may be addressable.
[333] The present invention also includes kits comprising one or a plurality of polynucleotides according to the present invention, along with all or some of the reactants required for genotyping of the test subject by identifying the identity of the nucleotides in the CanIon-related double allele marker. The polynucleotides of the kit may be attached to a solid support, or may be part of an array or addressable array of polynucleotides. The kit can identify the identity of the nucleotide at the marker position by methods known in the art including sequencing methods, microsequencing methods, hybridization assays or enzyme-based mismatch detection assay methods.
[334] Method for De Novo Identification of Double Allele Markers
[335] Genomic fragments of single nucleotide polymorphisms can be screened using a variety of methods, such as hybridization with oligonucleotide probes, direct sequencing of nucleic acids detected or changes in mobility as measured by gel electrophoresis. Suitable methods for identifying double allelic markers include comparative sequencing of genomic DNA fragments from a number of unrelated individuals.
[336] In a first embodiment, DNA samples from unrelated individuals are pooled together and then amplified and sequenced the genomic DNA of interest. The nucleotide sequences thus obtained are analyzed to identify significant polymorphisms. One important advantage of this approach is that pooling DNA samples substantially reduces the recovery of DNA amplification and sequencing reactions. Besides. This method is sensitive enough that the double allele marker thus obtained shows a sufficient frequency of the less common alleles used to conduct the relevant studies.
[337] In a second embodiment, DNA samples are not collected and are therefore amplified and sequenced individually. In general, this method is suitable when it is necessary to identify double allele markers in order to conduct linkage studies in candidate genes. Suitably, highly relevant gene regions, such as promoter regions or exon regions, can be screened for double allele markers. Dual allele markers obtained in this way may exhibit lower levels of informativeness in conducting relevant studies, for example, if the frequency of slightly less frequent alleles is less than 10%. However, these dual allelic markers provide enough information to conduct linkage studies, and including less beneficial markers in the genetic analysis studies of the present invention is in some cases a direct cause of causative mutations that are rare in the frequency of expression. Enable sympathy
[338] The following describes various parameters of a method suitable for use in the identification of a dual allele marker according to the present invention.
[339] Genomic DNA sequence
[340] Genomic DNA samples from which the dual allele markers of the present invention are made are preferably obtained from unrelated individuals corresponding to heterogeneous populations of known ethnic backgrounds. The number of individuals obtaining a DNA sample can vary substantially, preferably about 10 to 1000, more preferably 50 to 200. It is desirable to collect DNA samples from at least 100 individuals in order to identify as many markers as possible in any population and to obtain sufficient polymorphic diversity to produce statistically significant results.
[341] As regards the source of genomic DNA to be analyzed, any test sample can be used without limitation. These test samples contain biological samples that can be tested by the methods of the present invention, including human and animal body fluids such as whole blood, serum, plasma, cerebrospinal fluid, urine, lymph and respiratory, intestinal, urogenital External secretions, tears, saliva, milk, leukocytes, myeloma and the like; Biological fluids such as cell culture supernatants; Fixed tissue specimens such as tumor and non-tumor tissues and lymph node tissues; Bone marrow aspirate and immobilized cell specimens. Suitable sources of genomic DNA for use in the present invention are peripheral venous blood of each donor. Techniques for preparing genomic DNA from biological samples are known to those of skill in the art. Details of suitable embodiments are set forth in Example 1. One skilled in the art can choose whether to amplify the collected or unaggregated sample.
[342] DNA amplification
[343] The identification of double allele markers in samples of genomic DNA can be facilitated by the use of DNA amplification methods. DNA samples may or may not be collected for the amplification step. DNA amplification techniques are known to those skilled in the art.
[344] Amplification techniques that can be used in the present invention include ligase chain reaction (LCR (EP-A-320 308, WO 9320227, EP-A 439 182), polymerase chain reaction (PCR, RT-PCR), nucleic acid sequence based amplification ( Techniques such as NASBA (Guatelli JC, et al. (1990); Compton J. (1991), Q-beta amplification (European patent application No 4544610), strand substitution amplification (Walker et al. (1996); EP A 684 315) target targeted amplification (PCT application WO 9322461), but is not limited to these.
[345] LCR and Gap LCR is an exponential amplification technique that relies on DNA ligase to bind adjacent primers annealed to a DNA molecule. Probe pairs are used in ligase chain reaction (LCR), which includes two primary (first and second) probes and two secondary (third and fourth) probes, all of which are used in excess molar to the target do. The first probe hybridizes with the first segment of the target strand and the second probe hybridizes with the second segment of the target strand, wherein the first and second segments are contiguous, so the primary probe is a 5 'phosphate-3' hydroxyl relationship. And ligase can covalently fusion or ligation of the two probes with the fused product. In addition, the third (secondary) probe hybridizes with the portion of the first probe and the fourth (secondary) probe hybridizes with the portion of the second segment in a similar bordering manner. Of course, if the target is initially double stranded, the secondary probe first hybridizes with the target complement. When the ligation strand of the primary probe is separated from the target strand, it hybridizes with the third and fourth probes, which can be ligated to make complementary secondary ligation products. It is important to recognize that the ligated product is functionally equivalent to the target or its complement. Repeating the hybridization and ligation cycles achieves amplification of the target sequence. Multiple LCR methods have also been reported (WO 9320227). Gap LCR (GLCR) is a modified LCR in which the probe is not contiguous and separated by two to three bases.
[346] For amplification of mRNA in the present invention, a method of reverse transcription of mRNA to cDNA followed by polymerase chain reaction (RT-PCR), a method using a single enzyme in a step (US Pat. No. 5,322,770), or asymmetric Gap LCR (RT-AGLCR) ) (Marsshall et al. (1994)) is used. AGLCR is a modified GLCR capable of amplifying RNA.
[347] PCR techniques are suitable amplification techniques for the present invention. Various PCR techniques are known in the art (White (1997); "PCR Methods and Application", 1991, Cold Spring Harbor laboratory Press). In each of these PCR procedures, PCR primers are added to the nucleic acid sample appropriately prepared with dNTPs and thermostable polymerases such as Taq polymerase, Pfu polymerase or Vent polymerase on both sides of the nucleic acid sequence to be amplified. Hybridized primers are stretched. The cycle of denaturation, hybridization, and extension is then resumed. The cycle repeats several times to produce amplified fragments that retain the nucleic acid sequence between the primer sites. PCR is described in US Pat. No. 4,683,195; 4,683,202; It is described in more detail in several patents, including 4,965,188.
[348] PCR technology is an amplification technology suitable for identifying novel dual allele markers. Typical examples of PCR reactions suitable for the purposes of the present invention are shown in Example 2.
[349] One aspect of the present invention is a method for amplifying a human CanIon gene, particularly the genomic sequence of SEQ ID Nos. 1 to 3 or the cDNA sequence of SEQ ID 4, fragments or variants thereof, preferably using PCR in a test sample. The method consists of the following steps:
[350] a) contacting the test sample with an amplification reaction containing the amplification primer pair described above located on both sides of the polynucleotide region to be amplified;
[351] b) optionally, detecting the amplification product.
[352] The invention also relates to an amplification kit of a human CanIon gene, in particular a genomic sequence of SEQ ID Nos. 1 to 3 or a cDNA sequence of SEQ ID 4, fragments or variants thereof, in a test sample, wherein the kit consists of:
[353] a) a pair of oligonucleotide primers located on both sides of the CanIon region to be amplified;
[354] b) optionally, the reactants required to conduct the amplification reaction.
[355] In one embodiment of the amplification method and kit, the amplification product is detected by hybridization with a labeled probe having a sequence complementary to the amplified region. In another embodiment of the amplification method and kit, the primer comprises a sequence selected from nucleotide sequences of B1 to B17, C1 to C17, D1 to D18, E1 to E18.
[356] In a first embodiment of the invention, dual allele markers are identified using genomic sequence information obtained by the inventors. Sequenced genomic DNA fragments are used to design primers for amplification of 500 bp fragments. These 500 bp fragments amplify from genomic DNA and screen double allele markers. All primers have a common oligonucleotide tail that functions as a sequencing primer, upstream of a particular target base.
[357] Primers suitable for amplification of genomic sequences encoding the CanIon gene concentrate on the promoter, exon, and cleavage sites of the gene. Biele markers are placed in these functional regions of the gene, increasing the likelihood of causing mutations. Suitable amplification primers of the invention carry the nucleotide sequences B1 to B17 and C1 to C17 described in detail in Example 2, Table 1.
[358] Sequencing of Amplified Genomic DNA and Identification of Single Nucleotide Polymorphism
[359] The amplification products made as described above are then sequenced by any method known to those skilled in the art. Methods of sequencing DNA using dideoxy-mediated methods (Sanger method) or Maxam-Gilbert method are known to those skilled in the art. This method is disclosed by way of example in Sambrook et al. (1989). Alternative methods include hybridization to dense DNA arrays as found in Chee et al. (1996).
[360] Suitably, the amplified DNA is measured by an automated dideoxy terminator sequencing reaction using a dye terminator cycle sequencing protocol. Polymorphic search is based on the presence of overlapping peaks in an electrophoretic pattern resulting from different bases occurring at the same location. Because each dideoxy terminator is labeled with a different fluorescent molecule, the two peaks corresponding to the double allele site show differential colors corresponding to two different nucleotides at the same position on the sequence. However, the presence of two peaks may be an artifact due to background noise. To rule out these artifacts, the two DNA strands are sequenced and cross-peak comparisons are made. To be registered as a polymorphic sequence, polymorphism must be detected on both strands.
[361] The above procedure identifies these amplification products with double allele markers. The limit of detection of the frequency of the double allele polymorphism detected by the sequencing pool of 100 individuals is approximately 0.1 in the minor allele, as verified by the sequencing pool of known allele frequency. However, at least 90% of the double allelic polymorphisms detected by the pooling method have a frequency of at least 0.25 for minor alleles. Thus, the dual allele marker selected by the method has a frequency of at least 0.1 in the minor allele and 0.9 or less in the major allele, preferably at least 0.2 in the minor allele and 0.8 or less in the major allele, more preferably. Preferably, they have a frequency of at least 0.3 in the minor allele and 0.7 or less in the major allele and thus have a heterozygosity of at least 0.18, preferably at least 0.32, more preferably at least 0.42.
[362] In another embodiment, the double allele marker is detected by sequencing individual DNA samples, wherein the frequency of the minor allele of this double allele marker is 0.1 or less.
[363] Validation of the Double Allele Marker According to the Invention
[364] Polymorphism assesses utility as a genetic marker by verifying that both alleles are present in a population. Validation of the dual allele marker is accomplished by genotyping a group of individuals by the methods of the invention. Microsequence analysis is a suitable method for genotyping alleles. The validation by genotyping step can be performed on individual samples from each individual in the group or on aggregated samples from one or more individuals. The group may be one individual if one individual is heterozygous for the allele under study. Suitably, the group includes at least three subjects, more preferably at least 5-6 subjects, so that a plurality of dual allele markers can be verified with a single validation test. However, if a validation test is performed in a small group, a sampling error may result in false negatives if the test subject does not carry one of the two alleles. Thus, the verification process is not very effective at demonstrating that certain initial results are artifacts and that the double allele marker actually exists at certain locations on the sequence ( Bona fide ). Alternatively, the genotyping, haplotyping, association, and interaction research methods of the present invention may be performed only with validated dual allele markers.
[365] Frequency assessment of double allele markers according to the present invention
[366] The validated dual allele markers further assess their utility as genetic markers by measuring the frequency of the minimum common allele at the location of the dual allele marker. The more common the less common allele, the greater the utility of the double allele marker in linkage and interaction studies. Measurement of minimal common alleles is accomplished by genotyping a group of individuals by the methods of the invention and demonstrating that both alleles are present. The determination of the frequency by genotyping step can be carried out on individual samples derived from each individual in the group or on aggregated samples derived from one or more individuals. The group should be large enough to represent the entire population. Suitably the group includes at least 20 individuals, more preferably at least 50 individuals, most preferably at least 100 individuals. Of course, the larger the group, the greater the accuracy of the frequency measurement due to the reduced sampling error. Dual allele markers with a less common frequency of allele more than 30% are called "high quality dual allele markers." Optionally, the genotyping, haplotyping, association, and interaction study methods of the present invention may be performed with only high quality double allele markers.
[367] How to genotype an individual for double allele markers
[368] Provided are methods for genotyping a biological sample for one or more dual allele markers of the invention, which methods are performed in vitro. This genotyping method consists of identifying the identity of the nucleotide at the CanIon double allele marker location by methods known in the art. These methods are used to genotype an individual in the detection of an allele of a bi-allele marker known to be associated with a case-controlled population and any trait in a linkage study, wherein both copies of the dual allele marker present in the genome of the individual This is measured, so that an individual can be classified as homozygous or heterozygous for a particular allele.
[369] These genotyping methods can be performed on nucleic acid samples derived from single individuals or aggregated DNA samples.
[370] Genotyping can be carried out using methods similar to those described above for the identification of double allele markers, or using genotyping methods such as those described below. In suitable embodiments, sequence comparisons of genomic fragment fragments amplified from different individuals are used to identify novel dual allele markers, whereas microsequencing analyzes for genotyping known dual allele markers in diagnostic and related studies. Is used.
[371] In one embodiment, the present invention includes genotyping methods for identifying the identity of a nucleotide or complement thereof in a CanIon-related double allele marker in a biological sample; Optionally, the CanIon-associated biallelic marker is selected from A1 to A18 and its complement, or optionally a biallelic marker that is chain imbalanced thereto; Optionally, the CanIon-associated biallelic marker is selected from A1 to A17 and its complement, or optionally a biallelic marker that is chain imbalanced thereto; Optionally, the CanIon-related double allele marker is selected from A12 and A16 and its complement, or optionally a dual allele marker that is chain imbalanced thereto; Optionally, the biological sample is from a single individual; Optionally, said biological sample is from multiple individuals; Optionally, the identity of the nucleotides in the dual allele marker is identified in both copies of the dual allele marker present in the genome of the individual; Optionally, the genotyping methods of the present invention include the methods disclosed herein alone or in combination without any limitation; Optionally, the method is carried out in vitro; Optionally, further comprising amplifying the portion of the sequence bearing the dual allele marker prior to the identifying step. Optionally, the amplification is by PCR, LCR, or replication of a recombinant vector comprising the fragment and the origin of replication in a host cell; Optionally, the identification is performed by hybridization assay, sequencing, microsequence analysis or enzyme-based mismatch detection assay.
[372] Source of Nucleic Acids for Genotyping
[373] Sources of purified or non-purified nucleic acid can be used as starting nucleic acids if they possess or are believed to have the specific nucleic acid sequence desired. DNA or RNA can be extracted from cells, tissues, body fluids and the like as described above. If the nucleic acid used in the genotyping method of the present invention can be derived from any mammalian source, then the test subject and the individual taking the nucleic acid sample are considered human.
[374] Amplification of DNA Fragments Containing Double Allele Markers
[375] Provided are methods and polynucleotides for amplifying nucleotide segments comprising one or more allele markers of the invention. Amplification of DNA fragments containing double allelic markers can be used for a variety of methods and purposes, and is not limited to genotyping. However, most genotyping methods require pre-amplification of the DNA region carrying the double allele marker of interest. This method specifically increases the concentration or the total population of sequences that span or have the biallelic marker and are located far or near there. Diagnostic methods may also rely on the amplification of DNA segments carrying the dual allele markers of the invention. Amplification of DNA can be accomplished by any method known in the art. Amplification techniques are described above in the section "DNA Amplification".
[376] Some of these amplification methods are particularly suitable for the detection of single nucleotide polymorphisms and allow for simultaneous amplification of target sequences and identification of polymorphic nucleotides.
[377] The identification of the dual allele markers described above enables the design of suitable oligonucleotides, which may be used as primers to amplify DNA fragments comprising the dual allele markers of the invention. Amplification can be carried out using the primers used for the first use of a novel allele marker found herein, or a set of primers that allow for amplification of a DNA fragment comprising the dual allele marker of the invention.
[378] In some embodiments, the present invention provides primers for amplifying a DNA fragment carrying one or more dual allele markers of the invention. Suitable amplification primers are described in Example 2. The primers described are by way of example only, and other sets of primers may also be used that produce an amplification product bearing one or more dual allelic markers of the invention.
[379] The spacing of the primers determines the length of the segment to be amplified. In the present invention, the amplified segment carrying the double allele marker is at least 25 bp to 35 bp in size. The amplification fragment is 25-3000 bp, preferably 50-1000 bp, more preferably 100-600 bp. Amplification primers for the biallelic marker can be any sequence that enables specific amplification of the DNA fragment carrying the marker. Amplification primers may be labeled or immobilized to a solid support, as found in “oligonucleotide probes and primers”.
[380] Method of genotyping DNA samples for double allele markers
[381] Methods known in the art can be used to identify nucleotides present in the biallelic marker site. Since the dual allele marker allele to be detected has been identified and embodied in the present invention, one of ordinary skill in the art can readily detect it using a variety of techniques. Many genotyping methods require pre-amplification of the DNA region carrying the double allele marker of interest. Although amplification of the target or signal is presently preferred, supersensitive detection methods that do not require amplification are also included in the genotyping methods of the present invention. Methods known to those skilled in the art that can be used to detect double allelic polymorphisms include conventional dot blot analysis, single stranded polymorphism analysis (SSCP) (Orita et al. (1989)), denaturation gradient gel electrophoresis (DGGE), Heteroduplex analysis, mismatch elimination detection and other conventional techniques (Sheffield et al. (1991), White et al. (1992), Grompe et al. (1989 and 1993)). Another method of identifying the identity of nucleotides present in a particular polymorphic site utilizes specialized exonuclease-resistant nucleotide derivatives as disclosed in US Pat. No. 4,656,127.
[382] Suitable methods include directly measuring the identity of the nucleotides present in the biallelic marker site by sequencing, enzyme-based mismatch detection assays or hybridization assays. The following is a description of some suitable methods. Particularly suitable methods are microsequence analysis techniques. "Base sequencing" as used herein refers to polymerase extension of a double helix primer / template complex, including conventional sequencing and microsequencing.
[383] 1) sequencing
[384] Nucleotide present in the polymorphic site can be confirmed by sequencing. In a suitable embodiment, the DNA sample is PCR amplified prior to the sequencing described above. DNA sequencing methods are described in "Amplification of Amplified Genomic DNA and Identification of Single Nucleotide Polymorphisms".
[385] Suitably, the amplified DNA undergoes an automated dideoxy terminator sequencing reaction using a dye-primer cycle sequencing protocol. Sequencing allows for the identification of bases present at the biallelic marker site.
[386] 2) Microbase Sequence Analysis
[387] In the microsequencing method, nucleotides at the polymorphic sites of the target DNA are detected by single nucleotide primer extension reactions. The method involves a suitable microsequencing primer that hybridizes immediately upstream of the polymorphic base of interest in the target nucleic acid. The polymerase is used to specifically stretch the 3 'end of the primer with one single ddNTP (chain terminator) complementary to the nucleotide at the polymorphic site. The identity of the integrated nucleotides is then identified by any suitable method.
[388] Typically, the microsequence reaction is performed with fluorescence ddNTP and the elongated microsequencing primer is electrophoretically analyzed on an ABI377 sequencing machine to identify the identity of the integrated nucleotides, as identified in EP 412 883. Alternatively, capillary electrophoresis can be used to process a greater number of assays simultaneously. An example of a typical microsequencing procedure that can be used in the present invention is shown in Example 4.
[389] Different approaches can be used for labeling and detecting ddNTPs. Homologous phase detection methods based on fluorescence resonance energy transfer are described in Chen and Kwok (1997) and Chen et al. (1997). In this method, the amplified genomic DNA fragment carrying the polymorphic site is combined with a 5'-fluresin-labeled primer in the presence of the allele dye-labeled dideoxyribonucleoside triphosphate and modified Taq polymerase. Incubate. Dye-labeled primers are stretched monobasic by dye-terminators specific for alleles present in the template. At the end of the genotyping reaction, the fluorescence intensities of the two dyes in the reaction mixture are analyzed directly without separation or purification. All these steps can be performed in the same tube, and fluorescence changes can be monitored in real time. Alternatively, elongated primers can be analyzed by MALDI-TOF mass spectroscopy. Bases at polymorphic sites are identified by mass added to microsequencing primers (Haff and Smirnov, 1997).
[390] Microsequencing can be accomplished with the development of established sequencing methods or methods derived therefrom. Alternative methods include several solid-phase microsequencing techniques. The basic microsequencing protocol is the same as described above, except that the method is carried out with heterogeneous phase analysis in which the primer or target molecule is immobilized or captured on a solid support. To simplify primer separation and termination nucleotide addition assays, oligonucleotides are attached to a solid support or modified in a form that allows for affinity separation and polymerase extension. The 5 'terminus and internal nucleotides of the synthetic oligonucleotides can be modified in a variety of ways, such as biotinylation, to enable different ways of affinity separation. If a single affinity group is used for the oligonucleotide, the oligonucleotide can be separated from the integrated terminator reactant. This eliminates the need for physical or size exclusion. One or more oligonucleotides can be separated from the sampler reactant and analyzed simultaneously using one or more affinity groups. This allows for the analysis of different nucleic acid types or other nucleic acid sequence information per kidney reaction. Affinity groups are not required for the initiator oligonucleotides but may be present in the template. For example, immobilization can be accomplished through interaction between biotinylated DNA and streptavidin-coated microtitration wells or avidin-coated polystyrene particles. In the same way, oligonucleotides or templates can be attached to the solid support in a high density format. In this solid phase microsequence reaction, the integrated ddNTP can be radiolabeled (Syvanen, 1994) or bound to fluresin (Livak and Hainer, 1994). Detection of radiolabeled ddNTPs can be achieved through a scintillation-based technique. Detection of fluresin-bound ddNTPs can be based on the binding of alkaline phosphatase to conjugated antifluresin antibodies and subsequent incubation with a chromogenic substrate (eg p-nitrophenyl phosphate). Other possible reporter-detection pairs are: ddNTP and anti-DNP alkaline phosphatase conjugates bound to dinitrophenyl (DNP) (Harju et al., 1993), or o-phenyl as biotinylated ddNTP and substrate. Horseradish Peroxidase-Conjugated Streptavidin Containing Rendiamine (92/15712). As another alternative solid-phase microsequencing procedure, Nyren et al. (1993) described a method based on the detection of DNA polymerase activity by enzyme luminescent inorganic pyrophosphate detection assay (ELIDA).
[391] Pastinen et al. (1997) describe multiple detection methods for single nucleotide polymorphisms, where the principles of solid phase minisequencing are applied to oligonucleotide array formats. High density arrays of DNA probes attached to solid supports (DNA chips) are further described below.
[392] In one aspect, the invention provides polynucleotides and methods for genotyping one or more dual allele markers of the invention by performing microsequence analysis. Suitable microsequencing primers include nucleotide sequences D1 to D18 and E1 to E18. The microsequencing primers described in Example 4 are by way of example only, and any primer having a 3 'end adjacent to a polymorphic polynucleotide may be used. Similarly, microsequencing can be performed on any dual allele marker or combination of such markers in accordance with the present invention. One aspect of the invention provides one or more microsequencing primers, or at least 8, 12, 15, 20, 25, 30, 40, or 50, as described in Example 4 for identifying the identity of a nucleotide at a biallelic marker site. It is a solid support that retains fragments thereof, including contiguous nucleotides (where the fragment lengths match the primers described above) and has a 3 'end just upstream of the corresponding double allele marker.
[393] 3) Detection of mismatch detection based on polymerase and ligase
[394] In one aspect, the invention provides polynucleotides and methods for identifying alleles of one or more dual allele markers of the invention in a biological sample by polymerase and / or ligase based mismatch detection assays. These assays are based on the specificity of polymerase and ligase. The polymerization reaction requires particularly stringent exact base paring of the amplification primer 3 'end, and the binding of the two oligonucleotides hybridized to the target DNA sequence is very sensitive to mismedium near the ligation site, especially at the 3' end. Do. Methods, primers, and various parameters for amplifying a DNA fragment comprising a biallelic marker of the present invention are described above in "Amplification of a DNA fragment comprising a dual allele marker."
[395] Allele specific amplification primers
[396] The distinction between the two alleles of the double allele marker can also be achieved with allele specific amplification and selection strategies, where one of the alleles is amplified without amplification of the other allele. For allele specific amplification, at least one of the primer pairs is sufficiently complementary to the CanIon gene region comprising the polymorphic base of the dual allele marker according to the present invention to hybridize and induce amplification. Such primers can distinguish two alleles of a double allele marker.
[397] This is accomplished by placing a polymorphic base at the 3 'end of one of the amplification primers. Since elongation is made from the 3 'end of the primer, mismatches near this position show an inhibitory effect on amplification. Thus, under appropriate amplification conditions, these primers induce amplification to these complementary alleles. It is within the common knowledge of sugars to determine the exact location of the mismedia and the corresponding analysis conditions.
[398] Ligation / Amplification-Based Method
[399] "Oligonucleotide ligation analysis" (OLA) uses two oligonucleotides designed to hybridize to adjacent sequences of a single strand of a target molecule. One of the oligonucleotides is biotinylated and the other is detectably labeled. Once the exact complementary sequence is found in the target molecule, the oligonucleotides hybridize to make ends contact and create a capture and detectable ligation substrate. OLA can detect single nucleotide polymorphisms, which is advantageous with parallel PCR as revealed by Nickerson et al. (1990). In this method, PCR is used to achieve exponential amplification of the target DNA, which is then detected by OLA.
[400] Other amplification methods that are particularly suitable for the detection of single nucleotide polymorphisms include LCR (ligase chain reaction), Gap LCR (GLCR) described above in "DNA amplification". LCR uses two pairs of probes to exponentially amplify a particular target. The sequence of each pair of oligonucleotides is selected to allow hybridization with adjacent sequences of the same strand of the target. This hybridization forms a substrate for template-dependent ligase. In accordance with the present invention, LCR can be performed with oligonucleotides having contiguous and centrifugal sequences of the same strand of the double allele marker site. In one embodiment, the oligonucleotides are designed to retain a biallelic marker site. In such embodiments, the reaction conditions are selected such that the oligonucleotides can be ligated with each other only if the target molecule possesses or lacks a specific nucleotide that is complementary to the dual allele marker in the oligonucleotide. In alternative embodiments, oligonucleotides do not carry a dual allele marker, so "blanks" are made when they hybridize with a target molecule (WO 90/01069). This void is then "filled" with complementary dNTPs (mediated by DNA polymerase), or other oligonucleotide pairs. Thus, at the end of each cycle, each single strand has a complement that can function as a target during the next cycle and an exponential allele-specific amplification of the desired sequence is achieved.
[401] Ligase / polymerase-mediated Genetic Bit Analysis is another method of identifying the identity of nucleotides at preselected sites of nucleic acid molecules. Such methods include the step of integrating the nucleoside triphosphate complementary to the primer molecule terminus at the nucleotide present at the preselected site and subsequent ligation to the second oligonucleotide. The reaction is monitored by detecting a specific label attached to the solid phase or by detection in solution.
[402] 4) Hybridization Analysis Method
[403] Suitable methods for identifying the identity of the nucleotides present in the biallelic marker site include nucleic acid hybridization. Hybridization probes that can be conveniently used for such a reaction include probes as defined herein, wherever possible. Any hybridization assay can be used, such as Southern hybridization, northern hybridization, dot blot hybridization, solid phase hybridization (Sambrook et al., 1989).
[404] Hybridization refers to the formation of the double helix structure of two single stranded nucleic acids due to complementary base conjugation. Hybridization can occur precisely between complementary nucleic acid strands or between nucleic acid strands with some mismatched regions. One type of bi-allele marker hybridizes but not the other, so that specific probes can be designed that can distinguish different allelic forms. Allele-specific probes are often used in pairs, where one member of the pair shows a perfect match to the target sequence carrying the circular allele and the other member shows a perfect match to the target sequence carrying the alternative allele. . Hybridization conditions should be sufficiently stringent so that there is a significant difference in hybridization strength between alleles, preferably a binary reaction in which the probe hybridizes to only one of the alleles. Stringent sequence specific hybridization conditions in which the probe hybridizes only to exactly complementary target sequences are known in the art (Sambrook et al., 1989). Stringent conditions are sequence dependent, depending on the individual environment. In general, stringent conditions are selected at about 5 ° C. below the thermal melting point (Tm) for a particular sequence at defined ionic strengths and pH. Although such hybridization can be carried out in solution, it is preferred to use solid phase hybridization assays. Target DNA comprising the double allele marker of the present invention can be amplified prior to the hybridization reaction. The presence of a particular allele in the sample is confirmed by detecting the presence of a stable hybrid double helix formed between the probe and the target DNA. The detection of the hybrid double helix can be carried out in various ways. Various detection assay formats using detectable labels bound to targets or probes to enable detection of hybrid double helices are known in the art. Typically, hybridized double helices are separated from unhybridized nucleic acid and then the label bound to the double helix is detected. As one of ordinary skill in the art will appreciate, washing steps can be utilized to wash away excess DNA or probes and unbound conjugates. In addition, standard heterogeneous assay formats are suitable for detecting hybrids using labels present on primers and probes.
[405] Two recently developed assays are capable of distinguishing hybridized-based alleles without isolation or washing (Landegren U. et al., 1998). TaqMan assays utilize the 5 'nuclease activity of Taq DNA polymerase to cleave DNA probes that are specifically annealed to accumulation amplification products. TaqMan probes are labeled with donor-receptor dye pairs that interact through fluorescence energy transfer. Cleavage of the TaqMan probe by advancing polymerase during amplification separates the donor dye from the quenching receptor dye and significantly increases donor fluorescence. All reactants needed to detect two allelic variants can be aggregated at the beginning of the reaction and the results monitored in real time (Livak et al., 1995). In alternative homologous hybridization based procedures, molecular beacons are used to distinguish alleles. Molecular labels are hairpin-shaped oligonucleotide probes that report the presence of specific nucleic acids in homologous solutions. When they bind to the target, a conformational reorganization proceeds that restores the fluorescence of the internally quenched fluorophore (Tyagi et al., 1998).
[406] The polynucleotides presented herein can be used to produce probes that can be used in hybridization assays for the detection of dual allele markers in biological samples. These probes are complementary enough to hybridize with sequences comprising 8 to 50 nucleotides and comprising the dual allele markers of the invention and are specific enough to distinguish only one nucleotide variation in the target sequence. Is characterized. Particularly suitable probes are 25 nucleotides in length. Suitably, the allele marker is located within 4 nucleotides of the center of the polynucleotide probe. In particularly suitable probes, the allele marker is located at the center of the polynucleotide. Suitable probes include nucleotides selected from the amplicons listed in Table 1, their complementary sequences, or fragments thereof, wherein the fragments comprise at least 8 contiguous nucleotides, preferably 10, 15, 20 contiguous nucleotides, more preferably Contains 25, 30, 40, 47 or 50 consecutive nucleotides and possesses a polymorphic base. Suitable probes include nucleotide sequences selected from P1 to P8 and their complementary sequences. In suitable embodiments, the polymorphic base is located within 5, 4, 3, 2, 1 nucleotides from the center of the polynucleotide, more preferably in the center of the polynucleotide.
[407] Suitably, the probes of the present invention are labeled or immobilized on a solid support. Labels and solid supports are further described in "Oligonucleotide Probes and Primers". Probes may be non-extensionable as found in "oligonucleotide probes and primers".
[408] By analyzing hybridization to allele specific probes, the presence of a double allele marker allele can be detected in a given sample. High performance parallel hybridization in the array format is included in the "hybridization assay" and is detailed below.
[409] 5) Hybridization of Addressable Arrays of Oligonucleotides
[410] Hybridization assays based on oligonucleotide arrays are based on differences in the hybridization stability of short oligonucleotides to fully matched and mismatched target sequence variants. Effective access to polymorphic information is achieved through a basic structure that includes a high density array of oligonucleotide probes (eg chips) attached to the solid support at selected locations. Each DNA chip can have thousands to millions of individual synthetic DNA probes arranged in a lattice-like pattern and scaled down to coin sizes.
[411] Chip technology has been successfully applied in various cases. For example, screening of mutations has been carried out in the BRCA1 gene and the protease gene of HIV-1 virus in S. cerevisiae variant strains (Hacia et al., 1996; Shoemaker et al., 1996; Kozal et al., 1996 ). Chips in a variety of formats used to detect double allelic polymorphism can be custom made by Affymetrix (GeneChip ), Hyseq (HyChip and HyGnostics), Protogene Laboratories.
[412] In general, these methods utilize an array of oligonucleotide probes in which the target sequence is complementary to a target nucleic acid sequence segment derived from an individual carrying a polymorphic marker. EP 785280 describes a tiling strategy for the detection of single nucleotide polymorphisms. In short, an array is where a number of specific polymorphisms are “arranged like a tile”. In general, a “group” is a defined set of oligonucleotide probes consisting of a sequence complementary to a target sequence of interest and a preselected variation of such sequence, eg, a substitution of one or more predetermined positions with one or more members of the base nucleotide. Means synthesis. Tile strategies are described in more detail in PCR Application 95/11955. In certain aspects, the array is arranged in a tile-like manner with a plurality of specific identified double allelic marker sequences. In particular, the array is tiled with multiple probes spanning the sequence segments bearing a particular polymorphism. In order to cover the probes complementary to each allele, the probes are synthesized in pairs that differ in the double allele marker. In addition to probes that differ in polymorphic bases, monosubstituted probes are also tiled in the detection block. These monosubstituted probes carry a constant number of bases, substituted from other polynucleotides (selected from A, T, G, C (or U)) in both directions from polymorphism. Typically, the probes in the detection blocks arranged like tiling retain the substitution of the sequence up to 5 bases from the double allele marker. Monosubstituted probes provide an internal control for the array arranged like a tile to distinguish the actual hybridization from artificial cross-hybridization. Immediately after completion of hybridization with the target sequence and washing of the array, the array scans the location where the target sequence hybridizes. Hybridization data obtained from the scanned array is then analyzed to confirm that the allele of the double allele marker is present in the sample. Hybridization and scanning are described in PCR applications WO 92/10092 and WO 95/11955; It may be carried out as disclosed in US Pat. No. 5,424,186.
[413] Thus, in some embodiments the chip may comprise an array of nucleic acid fragment sequences of approximately 15 nucleotides in length. In another embodiment, the chip may comprise an array having at least one of the sequences selected from the amplicons, complementary sequences thereof or fragments thereof described in Table 1, said fragments comprising at least 8 contiguous nucleotides, preferably It contains 10, 15, 20 contiguous nucleotides, more preferably 25, 30, 40, 47 or 50 contiguous nucleotides and has a polymorphic base. Suitable probes include nucleotide sequences selected from P1 to P8 and their complementary sequences. In suitable embodiments, the polymorphic base is located within 5, 4, 3, 2, 1 nucleotides from the center of the polynucleotide, more preferably in the center of the polynucleotide. In some embodiments, the chip comprises an array consisting of at least 2, 3, 4, 5, 6, 7, 8 or more polynucleotides of the polynucleotides according to the present invention.
[414] The polynucleotides of the invention attached to the solid support and the solid support are further described in "Oligonucleotide Probes and Primers".
[415] 6) integrated system
[416] Other techniques that can be used to analyze polymorphism include multicomponent integration systems that reduce and partition processes such as PCR and capillary electrophoresis reactions in a single functional device. An example of this technique is disclosed in US Pat. No. 5,589,136, which illustrates PCR amplification on a chip. And the integration of capillary electrophoresis.
[417] Integrated systems can be considered primarily when using microfluidic systems. These systems include patterns of microchannels designed in glass, silicon, quartz or plastic wipers that are included in microchips. The movement of the sample is controlled by electrical, electroosmotic or buoyant forces applied to the entire area of the microchip, creating a functional microscopic valve and pump without a moving part.
[418] In genotyping double allele markers, microfluidic systems can incorporate detection methods such as nucleic acid amplification, microsequencing, capillary electrophoresis, and laser-induced fluorescence detection.
[419] Gene analysis method using double allele marker according to the present invention
[420] Several methods are available for genetic analysis of complex traits (Lander and Schork, 1994). The search for disease-vulnerable genes is carried out in two main ways: a chain approach that tracks evidence of cosegregation between loci and putative trait loci using family studies, and traits that induce alleles and alleles. Association method to track evidence of academically significant associations (Khoury et al., 1993). In general, the dual allele markers of the present invention can be used in methods known in the art to illustrate statistically significant correlations between genotypes and phenotypes. Double allele markers can be used in parametric and non-parametric chain analysis methods. Suitably, the dual allele markers of the present invention are used to identify genes associated with detectable traits using chain studies, which do not require the involvement of the affected family and are associated with complex and sporadic traits. Gene identification is possible.
[421] Gene analysis using the double allele marker of the present invention can be carried out on any scale. The entire set of biallelics according to the invention, or any subset of the biele markers according to the invention, corresponding to the candidate genes can be used. It is also possible to use any set of genetic markers carrying the dual allele marker of the invention. A group of double allele polymorphisms that can be used as gene markers with the dual allele markers of the present invention are described in WO 98/20165. As noted above, the dual allele markers of the present invention can be included in a full or partial gene map of the human genome. These separate uses are specifically defined in the present invention and claims.
[422] Chain research
[423] Chain studies are based on establishing a correlation between the delivery of genetic markers and the delivery of specific traits in generations in a family. Therefore, the aim of the chain study is to detect marker loci that show codegregation with the trait of interest in the household.
[424] Parametric method
[425] If data is available from successive generations, one can study the degree of chaining between pairs of loci. Evaluation of the recombinant fraction can arrange the locus and place it on a genetic map. The locus, which is a genetic marker, can be used to establish a gene map, and then calculate the intensity of the link between the marker and the trait, which is used to indicate the relative positions of the marker and the gene that affect these traits (Weir, 1996). The classical method of chain analysis is the algebra of the odd score method (Morton, 1955; Ott, 1991). The calculation of lod scores requires the specification of the genetic mode for the disease (parametric method). In general, the length of candidate regions identified in the chain studies is 2 to 20 Mb. Once the candidate regions have been identified as described above, a detailed description of the candidate regions is possible by analysis of the recombinant individual using additional markers. In general, chain analysis studies are based on the use of up to 5,000 microsatellite markers, so the theoretical maximum possible resolution of the chain analysis is on average 600 kb.
[426] Serial studies map simple genetic traits with clear Mendelian genetic patterns and with high penetrance (ie, the ratio of the population of the transgenic positive carrier of allele a and the total population of a carrier in the population). It is being used successfully. However, parametric chain analysis has many disadvantages. First, this is limited to the dependence on the selection of a genetic model suitable for each study trait. In addition, the resolution achievable using chain analysis as described above is limited, and complementarity studies should refine the analysis of 2Mb to 20Mb initially identified through chain analysis. In addition, parametric chain analysis methods are difficult to apply to complex genetic traits, such as those due to the complex action of multiple genes and / or environmental factors. It is very difficult to properly design these factors in load score analysis. In such cases, too much effort and expense is required to secure the appropriate number of families required to apply chain analysis in these situations (Risch and Merikangas. (1996)).
[427] Non-parametric method
[428] The advantage of the so-called non-parametric methods for chain analysis is that they do not require specification of the genetic mode for the disease and are more suitable for the analysis of complex traits. Non-parametric methods demonstrate that the affected inbreeding inherits the same copy of the chromosomal region more frequently than would be expected by chance, demonstrating that the genetic pattern of the chromosomal region is inconsistent with random Mandelian separation. Affected roots show excessive "allele sharing" even in the presence of incomplete expression and polygenic inheritance. The degree of concordance at the marker locus in two individuals in a non-parametric chain study can be measured by the number of identical (IBS) alleles in the state or the number of identical (IBD) alleles in the generation. Affected sib pair analysis is a special case known and is the simplest of these methods.
[429] The dual allele marker of the present invention can be used for both parametric and non-parametric chain analysis. Suitably, allele markers can be used in non-parametric methods that allow mapping of genes involved in complex traits. The double allele marker of the present invention can be used in IBD- and IBS-methods to map genes that affect complex traits. In this study, several contiguous double allele marker loci can be aggregated into high density double allele markers to achieve the efficiencies achieved by multi-allele markers (Zhao et al., 1998).
[430] Population Association Study
[431] The present invention includes a method for identifying whether the CanIon gene is associated with a detectable trait using the dual allele marker of the present invention. In one embodiment, the present invention includes a method of detecting a double allele marker allele or a double allele marker haplotype and a transgenic association. In addition, the present invention includes methods for identifying double allelic markers and chain imbalanced transgenic alleles of the present invention.
[432] As mentioned above, alternative methods can be used to conduct linkage studies: genome-wide association studies, candidate region association studies, candidate gene association studies. In suitable embodiments, the dual allele marker of the present invention is used to conduct candidate gene association studies. Candidate gene analysis provides a shortcut to the identification of genes and gene polymorphisms associated with specific traits when some information on the ecology of the trait is available. In addition, the dual allelic markers of the present invention can be incorporated into any map of genetic markers in the human genome to conduct genome-wide association studies. A method of making a high density map of a biallelic marker is described in US patent application Ser. The dual allele marker of the present invention may be integrated into any map of a particular candidate region of the genome (eg, a specific chromosome or a specific chromosome segment).
[433] As noted above, linkage studies are not limited to studies conducted on the entire population and on related individuals in the affected families. Association studies are very valuable in that they allow the analysis of sporadic or multiple traits. In addition, linkage studies are a viable method for fine-scale mapping that allows more detailed genetic mapping of transgenic alleles than chain studies. Family based research often precisely narrows the position of the transgenic allele. Thus, association studies using the dual allele markers of the present invention can be used to refine the position of the transgenic allele in candidate regions identified by the chain analysis method. Once the chromosomal segments of interest are identified, the presence of candidate genes, such as candidate genes of the invention, in the region of interest can provide a shortcut to the identification of transgenic alleles. The double allele marker of the present invention can be used to demonstrate that a candidate gene is associated with a trait. Such uses are specifically incorporated herein.
[434] Frequency measurement of double allele marker alleles or double allele marker haplotypes in a population
[435] Association studies explore the correlation between the frequencies of several alleles between loci.
[436] Frequency measurement of alleles in a population
[437] The allele frequency of a dual allele marker in a population can be measured by the method described above in "Methods for Genetic Discrimination of Individuals Against Dual Allele Markers", or by genotyping procedures suitable for this intended purpose. Genotyping of aggregated samples or individual samples can measure the frequency of double allele markers in a population. When using aggregated samples, the main obstacles are the accuracy and repeatability to determine the correct DNA concentration in establishing the pool. Genotyping of individual samples provides higher sensitivity, repeat reproducibility, accuracy, and is a suitable method for use in the present invention. Preferably, each individual is genotyped individually and simple conuting is applied to determine the frequency of alleles or genotypes of the dual allele marker in any population.
[438] The invention also relates to a method for measuring the frequency of alleles in a population, wherein the method consists of: a) genotyping the individual from the population of double allele markers according to the invention; b) measuring the proportional representation of the double allele marker in the population. In addition, methods for measuring the frequency of alleles in a population of the present invention include, without limitation, the methods described herein alone or in combination, wherein the CanIon-related double allelic markers are A1 to A18 and their complements, Or optionally a dual allele marker that is chain imbalanced thereto; Optionally, the CanIon-associated biallelic marker is selected from A1 to A17 and its complement, or optionally a biallelic marker that is chain imbalanced thereto; Optionally, the CanIon-related double allele marker is selected from A12 and A16 and its complement, or optionally a dual allele marker that is chain imbalanced thereto; Optionally, measuring the frequency of a dual allele marker allele in a population identifies the identity of the nucleotides to both copies of the dual allele marker present in the genome of each individual in the population and allows the CanIon-related dual allele marker for that population. Can be achieved by calculating the proportional representation of said nucleotides in; Optionally, confirmation of proportional expression can be achieved by subjecting the representative population in the population or a collection of biological samples derived from each individual to the genotyping method of the present application and calculating the proportional content of the nucleotides compared to the total.
[439] Frequency measurement of haplotypes in a population
[440] If a diploid individual is heterozygous at more than one locus, the haploid spouse phase is unknown. Family information can be used to infer the status of a spouse (Perlin et al., 1994). Different strategies can be used if household information is not available. One possibility is to exclude the multi-site heterozygous diplotype from the analysis and to retain only homozygous and single-site heterozygous individuals, but this approach may lead to underestimation of preconceived and low density haplotypes in the sample composition. Other possibilities are independently single chromosomes, for example by asymmetric PCR amplification (Newton et al, 1989; Wu et al., 1989) or isolation of single chromosomes by restriction dilution followed by PCR amplification (Ruano et al., 1990). To investigate. In addition, samples can be haplotyped to sufficiently close double allele markers by double PCR amplification of specific alleles (Sarkar, G. and Sommer S. S., 1991). These approaches are not entirely satisfactory due to technical complexity, additional costs, lack of generalization on a large scale, or possible prejudices. To overcome this difficulty, an algebra can be used to infer the phase of the PCR-amplified DNA genotype introduced by Clark, A.G. (1990). In short, this principle begins by listing a preliminary list of haplotypes present in the sample by examining distinct individuals, that is, full homozygotes and single-site heterozygotes. Subsequently, another subject in the same sample screens for the likely occurrence of a haplotype previously recognized. For each positive identification, complementary haplotypes are added to the list of recognized haplotypes until the topological information for all individuals is recognized as resolved or unresolved. The method assigns a single haplotype to each multiple heterozygous entity, where multiple haplotypes are possible if more than one heterozygous site is present. Alternatively, a method of measuring haplotype frequency in a population may be used without assigning a haplotype to each individual. Appropriately, the method based on the logarithm of expectation-maximization (EM), which yields a maximum likelihood estimate of haplotype frequency under the assumption of the Hardy-Weinberg ratio (random mating), Used (Exocoffier L. and Slatkin M., 1995). EM algebra is a generalized iterative maximal probabilistic measurement method that is useful when data is unclear / incomplete. EM algebra is used to haplotype heterozygotes. Haplotype measurements are described in detail later in “Statistical Methods”. Other known methods of measuring or evaluating the frequency of haplotypes in a population can be used.
[441] The invention also includes a method of measuring the frequency of haplotypes for a group of double allele markers in a population, the method comprising the following steps: a) in the method of the invention for each individual in the population. According to genotyping at least one CanIon-related double allele marker; b) genotyping the second double allele marker by identifying the identity of the nucleotides in the second double allele marker for both copies of the second double allele marker present in the genome of each individual in the population; c) applying a haplotype identification method to the identity of the nucleotides identified in steps a) and b); d) obtain an estimate of the frequency. In addition, methods for measuring the frequency of haplotypes of the present invention include, without limitation, the methods alone or in combination as disclosed herein, wherein the CanIon-related double allelic markers are A1 to A18 and their complements, or optional. And a double allele marker which is a chain imbalance with this; Optionally, the CanIon-associated biallelic marker is selected from A1 to A17 and its complement, or optionally a biallelic marker that is chain imbalanced thereto; Optionally, the CanIon-related double allele marker is selected from A12 and A16 and its complement, or optionally a dual allele marker that is chain imbalanced thereto; Optionally, haplotype identification methods are performed with asymmetric PCR amplification, double PCR amplification of specific alleles, Clark logarithm, or expected-maximum logarithm.
[442] Chain imbalance analysis
[443] Chain imbalance is a non-random association of alleles in two or more loci, and is a powerful tool for mapping genes involved in disease traits (Ajioka R.S. et al., 1997). Biallelic markers are particularly useful for gene analysis based on chain imbalance because they can be located densely in the human genome and genotyped in greater quantities than other types of genetic markers.
[444] When a disease mutation is first introduced into a population (due to the transfer of a new mutation or mutation carrier), it is placed in the "background" or "ancestor" haplotype of a single chromosome and linked marker. As a result, there is a complete imbalance between these markers and disease mutations: disease mutations are identified only when a group of specific marker alleles are present. In later generations, recombination occurs between disease mutations and these marker polymorphisms, and the disparities gradually disappear. This rate of extinction is a function of recombination frequency, with the marker closest to the disease gene showing a higher level of imbalance than the marker farther away. If not recombinantly disrupted, the "ancestor" haplotypes and linkage imbalances between marker alleles at different loci can be tracked throughout the population as well as in the family. In general, chain imbalance is considered an association between one specific allele at one locus and another specific allele at a second locus.
[445] The pattern or curve of the imbalance between the disease and the marker locus is expected to have a maximum at the disease locus. As a result, the total amount of chain imbalances between genetic markers closely associated with the disease allele can provide valuable information regarding the location of the disease gene. For small scale genetic mapping of disease loci, it is advantageous to have some knowledge regarding the pattern of chain imbalances present between markers in the study area. As mentioned above, the genetic mapping analytical power achieved through chain imbalance analysis is much higher than in the chain study. High-density double allele markers, along with chain imbalance analysis, provide a powerful tool for small-scale genetic mapping. Other methods of measuring chain imbalance are described in detail later in “Statistical Methods”.
[446] Population-Based Case-Control Study of Trait-Marker Association
[447] As mentioned above, the frequency of several pairs of specific alleles at different loci of the same chromosomes is not random, and the deviation from random is called linkage disequilibrium. Association studies focus on population frequency and are based on chain imbalance phenomena. If a particular allele in a given gene is directly involved in causing a particular trait, its frequency will increase statistically in the affected (trait positive) population compared to the frequency in the trait negative or randomized control population. Due to the presence of chain imbalance, the frequency of all other alleles present in haplotypes carrying the transgenic-inducing alleles also increases in transgenic positive individuals as compared to transgenic negative individuals or random controls. Thus, the association between a trait and a trans-induced allele and any allele that is a chain imbalance, in particular a dual allele marker allele, suggests the presence of a trait-associated gene in a particular region. Case-controlled populations can be genotyped for double allele markers to identify associations located in close proximity to the transgenic allele. One constant marker associated with a trait and any marker that is chain imbalance is associated with the trait. Chain imbalances make it possible to analyze relative frequencies in case-controlled populations of defined gene polymorphisms (especially dual allelic markers) as an alternative to the screening of all possible functional polymorphisms to find transgenic-inducing alleles. Linkage studies compare the frequency of marker alleles in irrelevant case-control populations and provide a powerful tool for the precise analysis of complex traits.
[448] Case-Control Population (Industrial Criteria)
[449] Population-based association studies compare the preponderance of a particular genetic marker or group of markers in a case-controlled population, although unrelated to familial inheritance. These are case-control studies based on a comparison of unrelated case (affected or trait positive) individuals with unrelated control (unaffected, trait negative or random) individuals. Suitably, the control consists of unaffected or transgenic negative individuals. In addition, the control group is ethnically matched with the case population. In addition, the control group is matched to the case-individual group for known major confusion factors for the trait under study (eg, age-matched for age-dependent traits). Ideally, individuals in both samples are paired in such a way that they are only distinguished from the disease state. "Trait positive population", "case population", and "affected population" are synonymous.
[450] An important step in the precise analysis of complex traits using linkage studies is the selection of case-control populations (Lander and Schork, 1994). A major step in the selection of case-control populations is the clinical definition of certain traits or phenotypes. Any genetic trait can be carefully selected for inclusion in the trait positive and trait negative groups and analyzed using the association methods presented herein. In general, four criteria are used: clinical phenotype, age of onset, family history, and severity. The screening process for sequential or quantitative traits (eg, blood pressure) involves selecting individuals at both extremes from the phenotype distribution of the trait under study so that individuals with non-overlapping phenotypes are included in the trait positive and trait negative populations do. Suitably, the case-control population consists of homologous populations in the phenotype. The trait positive and trait negative populations are 1 to 98%, preferably 1 to 80%, more preferably 1 to 50%, more preferably 1 to 30%, most preferably of the total population under study, respectively. Is composed of populations homologous in phenotypes of individuals representing 1-20%, and the selected individuals show non-overlapping phenotypes. The clearer the difference between the two trait phenotypes, the higher the probability of detecting association with the double allele marker. Selection of significantly different but relatively homologous phenotypes allows for efficient comparison and detection of significant differences at the genetic level in the study, provided that the sample size of the population under study is large enough.
[451] In a suitable embodiment, the first group consisting of 50 to 300 transfective individuals, preferably approximately 100 individuals, is involved according to the phenotype. A similar number of controls were involved in this study.
[452] Association analysis
[453] The invention also includes a method for detecting association between genotypes and phenotypes, which method comprises the following steps: a) at least one CanIon-related duplex in a transgenic positive population according to the genotyping methods of the invention; Measuring the frequency of allele markers; b) determining the frequency of said CanIon-related double allele markers in a control population according to the genotyping method of the invention; c) Determine if there is a statistically significant correlation between genotypes and phenotypes. In addition, methods for detecting associations between genotypes and phenotypes of the present invention include, without limitation, the methods described herein alone or in combination, wherein the CanIon-related dual allele markers are A1 to A18 and their complements, or Optionally is selected from a double allele marker that is chain imbalanced thereto; Optionally, the CanIon-associated biallelic marker is selected from A1 to A17 and its complement, or optionally a biallelic marker that is chain imbalanced thereto; Optionally, the CanIon-related double allele marker is selected from A12 and A16 and its complement, or optionally a dual allele marker that is chain imbalanced thereto; Optionally, the control population may be a trait negative control, or a random population; Alternatively, genotyping steps a) and b) can be performed on aggregated biological samples from each of said populations; Optionally, the genotyping steps of a) and b) are performed individually on biological samples derived from each individual, or subsample thereof, in said population.
[454] A general strategy for conducting linkage studies using double allele markers derived from regions containing candidate genes is to assess and statistically compare allele frequencies of the double allele markers of the present invention in both groups (cases) Scan the control population).
[455] If a statistically significant association with a trait is identified in one or more dual allele markers analyzed, the associated allele directly induces the trait (ie, the associated allele is the transgenic allele), or the associated allele Can be assumed to be chain imbalanced with the transgenic allele. The characteristic nature of the associated allele with respect to the candidate gene function provides further insight into the correlation (causal or chain imbalance) between the associated allele and the trait. If this evidence suggests that the associated allele in the candidate gene is not a transgenic allele and is in a chain imbalance with the actual transgenic allele, the transgenic allele is sequenced and identified in an iterative manner around the associated marker. Polymorphisms can be discovered by conducting additional association studies.
[456] In general, linkage studies are conducted in two successive steps. In the first step, the frequency of reduced allele markers from candidate genes is measured in the trait positive and control populations. In the second step of the assay, the location of the locus that drives a constant trait is further refined using markers of higher density from the relevant regions. However, if the candidate gene under study is relatively short, as in CanIon, a single step is sufficient to establish a significant association.
[457] Haplotype analysis
[458] As noted above, when a chromosome carrying a disease allele first appears in a population as a result of mutation or migration, the mutant allele is present in a chromosome carrying a group of linked markers: ancestral haplotypes. These haplotypes can be tracked throughout the population and analyzed for statistical association with a given trait. Complementing single point (allele) linkage studies with multi-point linkage studies, called haplotype studies, increases the statistical power of linkage studies. Thus, haplotype association studies can define the frequency and type of ancestor carrier haplotypes. Haplotype analysis is important in that it increases the statistical capability of the assay with individual markers.
[459] In the first step of haplotype frequency analysis, the frequency of possible haplotypes is determined based on various combinations of the identified allele markers of the present invention. Haplotype frequencies are then compared against separate populations of the trait positive and control. The number of transgenic positive individuals to which this analysis is applied in order to obtain statistically significant results is generally from 30 to 300, preferably from 50 to 150. The same considerations apply to the number of unaffected individuals (or randomized controls) who participated in this study. The results of this first analysis give haplotype frequencies in the case-control population, with the p-value and odd ratio calculated at each evaluated haplotype frequency. Once statistically significant associations have been identified, relative risk can be approximated for individuals with certain haplotypes affected by the trait under study.
[460] Another embodiment of the present invention includes a method for detecting an association between a haplotype and a phenotype, which method comprises the following steps: a) transfective according to the method of the present invention for determining the frequency of a haplotype Determining the frequency of at least one haplotype in the population; b) determining the frequency of said haplotype in the control population according to the method of the present invention for determining the frequency of the haplotype; c) Determine if there is a statistically significant association between haplotypes and phenotypes. In addition, methods for detecting the association between haplotypes and phenotypes of the present invention include, without limitation, the methods described herein alone or in combination, wherein the CanIon-related double allelic markers are A1 to A18 and their complements, Or optionally a dual allele marker that is chain imbalanced thereto; Optionally, the CanIon-associated biallelic marker is selected from A1 to A17 and its complement, or optionally a biallelic marker that is chain imbalanced thereto; Optionally, the CanIon-related double allele marker is selected from A12 and A16 and its complement, or optionally a dual allele marker that is chain imbalanced thereto; Optionally, the control population may be a trait negative control, or a random population; Optionally, the control population is a trait negative population, or a random population. Optionally, the method further comprises identifying a phenotype in the trait positive and control population prior to step c).
[461] Interaction analysis
[462] Biallelic markers of the present invention can also be used to identify patterns of biallelic markers associated with detectable traits due to multifactor interactions. Analysis of gene interactions between alleles at loci that do not belong to the same chain group requires individual genotyping using the techniques disclosed herein. Analysis of allele interactions between selected sets of double allele markers with appropriate levels of statistical significance can be considered haplotype analysis. Interaction analysis consists of stratifying case-control populations with respect to a constant haplotype for the first locus and conducting a haplotype analysis with a second locus in each subpopulation.
[463] The statistical methods used in the linkage studies are described in detail below.
[464] Examination of the chain in the presence of association
[465] The dual allele marker of the present invention can also be used for TDT (delivery / imbalance test). TDT testing of chains and associations is not affected by population stratification. TDT requires data on affected individuals and their parents, or data from unrelated relatives instead of parents (Spielmann S. et al., 1993; Schaid DJ et al., 1996, Spielmann S. and Ewens WJ, 1998). This integrated test reduces overall false positives by individual analysis.
[466] Statistical method
[467] In general, any method known in the art can be used to test whether a trait and genotype show a statistically significant correlation.
[468] 1) Method in Chain Analysis
[469] Statistical methods and computer programs useful for chain analysis are known to those skilled in the art (Terwilliger J.D. and Ott J., 1994; Ott J., 1991).
[470] 2) How to measure haplotype frequency in a population
[471] As described above, in the case of scoring genotypes, there are cases where heterozygotes are not distinguished and haplotype frequencies cannot be easily inferred. If the spouse phase is unknown, haplotype frequency can be estimated from the multipositional genotyping data. Any method known to those skilled in the art can be used to assess haplotype frequency (Lange K., 1997; Weir, B.S., 1996). Suitably, the maximal-commune haplotype frequency is expected-maximization (EM) logarithm (Dempster et al., 1977; Exocoffier L. and Slatkin M., 1995). This process is an iterative process to obtain the maximum-communication of haplotype frequency from multi-site genotyping data when the spouse phase is unknown. In general, haplotype assessments are carried out by applying EM algebra using, for example, the EM-HAPLO program (Hawley M. E. et al., 19994) or the Arlequin program (Schneider et al., 1997). The EM logarithm is a generalized iterative maximum probabilistic measurement method, which is briefly described below.
[472] Phenotype in this section, "Method of measuring haplotype frequency in a population" refers to a multi-site genotype with an unknown haplotype phase. Genotype means a multi-site genotype with a known haplotype phase.
[473] Assume a sample of N unrelated individuals of type K marker. The observed data is an unknown phase K -left phenotype that can be classified into F different phenotypes. It is also assumed that H possesses a haplotype (in the case of a K double allele marker, it holds a maximum value of the possible haplotype H = 2 K ).
[474] Phenotype j with C j possible genotypes
[475]
[476] Where P j is the probability of the j th phenotype and P ( h k , h l ) is the probability of the i th genotype consisting of haplotypes h k and h l . Under random confrontation (ie Hardy-Weinberg Equilibrium), P ( h k , h l ) is represented as:
[477]
[478] The EM logarithm consists of the following steps: First, genotype frequency is estimated from a set of initial values of haplotype frequency. These haplotype frequencies are called P 1 (0) , P 2 (0) , P 3 (0) , ..., P H (0) . Initial values for haplotype frequencies can be obtained by random number generators or by other methods known in the art. The second step, called the maximization step, re-calculates haplotype frequency using estimates for genotype frequency. The first iterative haplotype frequency estimates are P 1 (1) , P 2 (1) , P 3 (1) , ..., P H (1) . Overall, the expectation step in the s th iteration calculates the probability of designating each phenotype as a separate possible genotype based on the haplotype frequency of the previous iteration:
[479]
[480] Where n j is the number of individuals with j th phenotype, and P ( h k , h l ) ( s ) is the probability of genotype h k , h i in phenotype j . In the maximizing step corresponding to the gene-counting method (Smith, Ann. Hum. Genet., 21: 254-276, 1957), haplotype frequency is re-evaluated based on genotype estimates:
[481]
[482] Where δ it is an indicator for calculating the number of occurrences of haplotype t present in the l th genotype; it is 0, 1, 2.
[483] EM repetition stops when the following criteria are reached. Using the maximum communicative measurement (MLE) theory, it is assumed that the phenotype j is a multinomial distribution. At each iteration s , the likelihood function L can be calculated. Convergence is achieved if the difference in log-likelihood between two successive iterations is small, preferably 10 −7 .
[484] 3) How to calculate the chain imbalance between markers
[485] Various methods can be used to calculate chain imbalances between any two gene positions. Indeed, chain imbalance is measured by applying statistical correlation tests to haplotype data obtained from a population.
[486] In the marker M i allele (a i / b i) and marker M j in the allele containing at least one of (a j / b j) Double allele marker (M i, M j) of the present invention to hold the The chain imbalance between any pair of allele markers can be calculated for all allele combinations (a i , a j ; a i , b j ; b i , a j ; b i , b j ) according to the Piazza formula:
[487] Δa i a j = √θ4-√ (θ4 + θ3) (θ4 + θ2), where
[488] θ4 = - = alleles at M i a i a does not hold that does not have an allele a j at M j frequency of genotypes;
[489] θ3 = - + = M i allele a i does not hold the frequency of the genotypes for holding allele a j at M j in;
[490] θ2 = + - = holding allele a i at M i and the frequency of the genotypes that do not have an allele at M j a j.
[491] As noted in Weir BS, 1996, allele combinations (a i , a j ; a i , b j ; b i , a j ; b), depending on the maximum communicative value (MLE) for delta (hybrid genotype imbalance coefficient) i , b j ) may also calculate the chain imbalance (LD) between the pair of dual allele markers (M i , M j ). The MLE for hybrid chain imbalance is:
[492] Da i a j = (2n 1 + n 2 + n 3 + n 4/2) / N-2 (pr (a i). Pr (a j))
[493] Where n 1 is the Σ representation (a i / a i , a j / a j ), n 2 is the Σ representation (a i / a i , a j / b j ), and n 3 is the Σ representation (a i / b i , a j / a j ), n 4 is the Σ phenotype (a i / b i , a j / b j ), and N is the number of individuals in the sample.
[494] With this formula, it is possible to assess allele chain imbalances when only genotyping data is available and haplotype data is not available.
[495] Another means of calculating the chain imbalance between markers is as follows. By fitting the Hardy-Weinberg equilibrium for a pair of allele markers, Mi ( a i / b i ) and M j ( a j / b j ), four possible haplotype frequencies in a given population in the manner described above Can be evaluated.
[496] Estimates of spousal imbalance are as follows:
[497] Da i a j = pr (haplotype ( a i , a j ))-pr ( a i ) .pr ( a j ).
[498] Where pr ( a i ) is the probability of allele a i and pr ( a j ) is the probability of allele a j , and pr ( a i ) .pr ( a j ) is estimated as in Equation 3 above.
[499] For a pair of allele markers, only one measure of disequilibrium is needed to explain the association between Mi and M j .
[500] Then, its standardized value is calculated as follows:
[501]
[502] Other chain imbalance calculation methods may be used.
[503] Chain imbalance between a group of double allele markers with an appropriate expected heterozygosity rate can be determined by genotyping between 50 and 1000, preferably 75 and 200, more preferably approximately 100 unrelated individuals. Can be.
[504] 4) Association check
[505] Methods for determining the statistical significance of alleles or haplotypes in these alleles in double allel markers or phenotypes and gene correlations are known in the art and require any acceptable threshold of statistical significance. This can be achieved by statistical verification. The use of certain methods and significant thresholds is known to those skilled in the art.
[506] Association tests measure the frequency of double allele marker alleles in cases and control populations, and statistically compare these frequencies to suggest statistical correlations that suggest the correlation between the traits and the double allele marker alleles they are studying. This step consists of identifying whether there is a significant difference. Similarly, haplotype analysis measures the frequencies of all possible haplotypes for a given set of double allele markers in cases and control populations, and compares these frequencies with statistical validation to compare them with the haplotypes and phenotypes being studied. Identifying whether there is a statistically significant correlation between livers. Any statistical tool useful for verifying statistically significant correlations between genotypes and phenotypes can be used. Suitably, the statistical test used is a chi-square test with one degree of freedom. The P-value is calculated (P-value is the probability that a statistic above the observation occurs by chance).
[507] Statistical significance
[508] In a suitable embodiment of significance for diagnostic purposes as a positive basis for further diagnostic testing or as a preliminary starting point for early prophylactic therapy, the p-value associated with the biallelic marker association is preferably 1 x 10 in a single biele marker analysis. No greater than -2 , more preferably no greater than 1 × 10 −4, and no greater than 1 × 10 −3 , preferably no greater than 1 × 10 −6 , in haplotype assays comprising two or more markers. These values are applicable to any association study involving single or multiple marker combinations.
[509] Those skilled in the art can use the values in the foregoing ranges as starting points for conducting association studies with the dual allele markers of the present invention. By doing so, a significant correlation between the dual allele marker and the trait of the present invention can be identified, which can be used for diagnostic and drug screening purposes.
[510] Phenotype Permutation
[511] In order to test the statistical significance of the one-step haplotype analysis described above, it is desirable to perform further analysis, where genotyping data from case-control individuals is collected and randomized to the trait phenotype. Each individual genotyping data is randomly assigned to two groups with the same number of people as the case-controlled population, and the data obtained in step 1 is edited. Two-stage haplotype analysis is performed in these artificial groups for markers included in the haplotype of the first-stage analysis, where possible, with the highest comparative risk factor. Suitably, this experiment is repeated at least 100-10000 times. This repetition allows the measurement of the probability of accidentally obtaining a haplotype tested.
[512] Evaluation of Statistical Associations
[513] To address the false positive problem, similar analyzes can be performed with the same case-control population in a random genomic region. Results in random and candidate regions are compared as described in the US Division of Application "Methods, software and apparati for identifying genomic regions harboring a gene associated with a dectable trait" (60 / 107,986; filed Nov. 10, 1998).
[514] 5) assessment of risk factors
[515] Associations between risk factors (in genetic toxicity, the presence of specific alleles or haplotypes at the locus of markers) and disease are measured by odds ratio and comparative risk (RR). If P (R +) is the probability that R and disease develop and P (R-) is the probability in individuals without risk factors, the comparative risk is the ratio of these two probabilities:
[516] RR = P (R +) / P (R-)
[517] In case-control studies, a direct measure of comparative risk cannot be obtained due to sampling design. However, the odds ratio allows an effective approximation of the comparative risk for low-incidence diseases, which can be calculated as follows:
[518]
[519] F + is the frequency of exposure to risk factors in the case and F is the frequency of exposure to risk factors in the control. F + and F are calculated using allele or haplotype frequencies in this study, and are further dependent on the underlying genetic model (dominant, recessive, additive ...).
[520] Attribution risk (AR) can be assessed, which refers to the proportion of individuals in a population that are traits due to certain risk factors. Such measurements are important for quantifying the role of certain factors in terms of disease pathology and public health impacts of risk factors. The public health relevance of these measures is to assess the proportion of disease cases in the population that can be prevented in the absence of exposure. AR is measured as follows:
[521] AR = P E (RR-1) / (P E (PR-1) +1)
[522] AR is the contribution risk to the double allele marker allele or the double allele marker haplotype. P E is the frequency of exposure to the allele or haplotype in the entire population; RR is a comparative risk that approximates the odds ratio when the trait under study has a relatively low incidence in the entire population.
[523] Identification of a Dual Allele Marker of Chain Imbalance with the Dual Allele Marker of the Present Invention
[524] Once the first double allele marker is identified in the genomic region of interest, one of ordinary skill in the art can readily identify other double allele markers that are chain imbalanced with the first marker using the technical spirit of the present invention. As mentioned above, any marker that is in chain imbalance with the first marker will be associated with the trait. Thus, if the association is demonstrated between a constant allele marker and a trait, the discovery of additional double allele markers associated with this trait is of great interest to increase the density of the double allele marker in that particular region. Causal genes or mutations are found in the vicinity of a group of markers that show maximum correlation with the trait.
[525] Identification of certain markers and additional markers that are chain imbalance consists of the following steps: (a) amplifying a genomic fragment comprising a first biallelic marker from a plurality of individuals; (b) identifying a second double allele marker in the genomic region containing the first double allele marker; (c) conducting a chain imbalance analysis between the first and second allele markers; (d) Select a second double allele marker that is chain imbalanced with the first marker. Sub-combinations consisting of steps (b) and (c) are also incorporated.
[526] Methods for identifying double allelic markers and performing chain imbalance analysis are described herein and can be performed by those skilled in the art without undue experimentation. The present invention also relates to the dual allele markers A1 to A18, which are in chain imbalance and are expected to exhibit similar properties in terms of individual association with certain traits.
[527] Identification of functional mutations
[528] Mutations in the CanIon gene that drive a detectable phenotype or trait can be identified by comparing the sequence of the CanIon gene from the trait positive and control. If a positive association is identified with the dual allele marker of the present invention, the identified locus can scan for mutations. In suitable embodiments, functional regions such as exons and cleavage sites of the CanIon gene, promoters and other regulatory regions scan for mutations. In a suitable embodiment, the sequence of the CanIon gene is compared in the trait positive and control subjects. Preferably, the trait positive entity has a haplotype that appears to be associated with the trait, and the trait negative entity is a haplotype associated with the trait. Or does not have an allele. Various manifestations of CanIon function modified to detectable traits or phenotypes are included.
[529] The mutation detection process is similar to that used to identify double allele markers. The method used to detect such mutations generally consists of the following steps:
[530] Amplification of the CanIon gene region comprising a double allele marker or a group of allele markers associated with a trait from a DNA sample of a trait positive patient and a trait negative control;
[531] Sequencing of the amplified regions;
[532] Comparison of DNA sequences from the transfective and control individuals;
[533] Identification of mutations specific for trans-positive patients
[534] In one embodiment, the dual allele marker is selected from A1 to A18 and complement thereof. Candidate polymorphism is then validated by screening large populations of cases and controls with the genotyping process disclosed herein, preferably microsequencing techniques in individual test formats. Polymorphisms are considered candidate mutations when present at a frequency consistent with the predicted association outcome in cases and controls. Polymorphisms are considered candidate "trait-induced" mutations if they show a statistically significant correlation with the detectable phenotype.
[535] Dual allele marker of the present invention in a method of gene diagnosis
[536] The CanIon nucleic acid sequence and the double allele of the present invention develop diagnostic tests that can identify individuals who express detectable traits as a result of a particular genotype, or individuals who are at high risk for developing detectable traits in later life due to the genotype. Can also be used to This diagnosis may include staging of various diseases or conditions, including schizophrenia, bipolar disorder, other CNS diseases (e.g. epilepsy), pain diseases, cardiovascular diseases (e.g. heart disease, high blood pressure, arrhythmia) and other diseases and abnormalities, It can be used for monitoring, prediction and / or prophylactic or therapeutic regimens.
[537] The diagnostic techniques of the present invention include a method capable of analyzing individual chromosomes for haplotyping, such as family studies, simple sperm DNA analysis, or somatic hybrids, including dual associated with an increased risk of developing a detectable trait. Various methods can be used to determine whether they have an allele marker pattern or whether an individual suffers from a detectable trait as a result of a particular mutation.
[538] The present invention provides a diagnostic method for identifying whether an individual is at risk of developing an illness or if a disease is caused by mutation or polymorphism in the CanIon gene. The invention also provides a method for ascertaining whether an individual has susceptibility to schizophrenia and bipolar disorder or calcium-channel related abnormalities known or disclosed herein.
[539] These methods include obtaining a nucleic acid sample from an individual and confirming that the nucleic acid sample carries one or more alleles or one or more allele marker haplotypes, the presence of which suggests a risk of expression of the trait. Or to express a trait as a result of an individual carrying a particular CanIon polymorphism or mutation (trait-induced allele).
[540] Suitably, in such a diagnostic method, a nucleic acid sample is obtained from an individual, and the sample is genotyped by the method described above in "Method for Genotyping DNA Samples for Dual Allele Markers". Diagnosis is based on either a single allele marker or a group of double allele markers.
[541] In these methods, nucleic acid samples are obtained from a sample and the pattern of at least one of the allele markers A1 to A18 is identified.
[542] In one embodiment, PCR amplification is performed on a nucleic acid sample to amplify a region in which a polymorphism associated with a detectable phenotype is identified. The amplification product is sequenced to confirm that the individual possesses one or a plurality of CanIon polymorphisms associated with a detectable phenotype. Primers used to make amplification products include the primers listed in Table 1. Alternatively, the nucleic acid sample is subjected to the microsequencing reaction described above to confirm that the individual possesses one or more CanIon polymorphisms associated with a detectable phenotype resulting from mutation or polymorphism in the CanIon gene. Primers used in the microsequencing reaction include the primers listed in Table 4. In other embodiments, the nucleic acid sample is contacted with one or a plurality of allele specific oligonucleotide probes that specifically hybridize to one or more CanIon alleles associated with a detectable phenotype. Probes used in hybridization assays include the primers listed in Table 3. In another embodiment, the nucleic acid sample is contacted with a second CanIon oligonucleotide that can produce an amplification product when used with an allele specific oligonucleotide in an amplification reaction. The presence of the amplification product in the amplification reaction suggests that the individual carries one or more CanIon alleles associated with a detectable phenotype.
[543] In a suitable embodiment, the identity of the nucleotides present in one or more bi-allele markers selected from A1 to A18 and complements thereof is identified, with detectable traits being schizophrenia and bipolar disorder. The diagnostic kit comprises a polynucleotide of the present invention.
[544] These diagnostic methods are very useful in that they can be used to initiate prophylactic treatment in a specific environment or to allow a person with a significant haplotype to recognize warning signs such as minor symptoms.
[545] Diagnosis, which analyzes and predicts the response to drugs or adverse effects on drugs, can be used to determine whether to treat an individual with a particular drug. For example, if the diagnosis indicates that the individual is likely to respond positively to a particular drug treatment, the drug may be administered to the individual. Conversely, if the diagnosis indicates that the individual is likely to respond negatively to certain medications, another course of treatment may be prescribed. Negative responses can be defined as the absence of a beneficial response or the presence of toxic side effects.
[546] The markers of the present invention can also be used for clinical drug experiments. One or more markers in response to an agent that inhibits schizophrenia, bipolar disorder or other calcium-channel related abnormalities, or suggests side effects to agents that inhibit schizophrenia, bipolar disorder or other calcium-channel related abnormalities, may be used in the manner described above. I can sympathize. The potential participants are then screened in clinical trials of these agents to identify those who are most likely to respond positively to the drug and exclude those who are more likely to have side effects. In this way, the effectiveness of treatment can be measured in individuals who respond positively to the drug, without compromising measurement due to the participation of individuals who are less likely to respond positively during the study, or without the risk of unwanted safety issues.
[547] In a particularly suitable embodiment, the trait to be analyzed in this diagnosis is schizophrenia or bipolar disorder. However, the present invention includes methods for preventing, diagnosing, predicting, and treating a dual allele marker of the present invention in a method for preventing, diagnosing, managing, and treating a related disease, particularly a related CNS disease. Examples of related diseases include mental disorders, mood disorders, autism, drug dependence and alcoholism, pain disorders, epilepsy, mental retardation and cognitive disorders, mental disorders, anxiety disorders, eating disorders, impulse control disorders, personality disorders, including personality disorders Diseases are included. Diseases can be defined according to the Diagnosis Statistical Manual of Mental Disorder fourth edition (DSM-IV) classification. Other diseases include cardiovascular diseases such as angina pectoris, hypertension or arrhythmia.
[548] Recombinant vector
[549] By “vector” is meant a circular or linear DNA or RNA molecule, which includes at least one polynucleotide that is double-stranded or single-stranded and delivered to a cell host or to a single cell or multicellular host microorganism.
[550] The present invention includes a group of recombinant vectors comprising regulatory polynucleotides derived from CanIon genomic sequences and / or coding polynucleotides derived from CanIon genomic sequences or cDNA sequences.
[551] In general, recombinant vectors of the invention include polynucleotides of the invention, including regulatory sequences, coding sequences, polynucleotide constructs, and the CanIon primers or probes described above. More specifically, the recombinant vector of the present invention is any of those identified in the "Genome sequence of CanIon gene" section, "CanIon cDNA sequence" section, "coding region" section, "polynucleotide structure" section, "oligonucleotide probe and primer" section. It may comprise a polynucleotide of.
[552] In a suitable first embodiment, the recombinant vector of the present invention comprises an inserted polynucleotide derived from a CanIon genomic sequence of SEQ ID Nos. 1 to 3 or 6 or a CanIon cDNA, eg, cDNA of SEQ ID No 4, in a suitable host cell. Used to amplify, the polynucleotide is amplified each time the recombinant vector replicates.
[553] In a suitable second embodiment, the recombinant vector according to the present invention consists of an expression vector comprising a regulatory polynucleotide, a coding nucleic acid, or both of the present invention. In certain embodiments, an expression vector is used to express a CanIon polypeptide, which may then be used for purification and ligand screening assays or as an immunogen for forming an antibody against the CanIon protein. In other embodiments, the expression vector is used for the generation of transgenic animals and gene therapy. Expression is only possible if an appropriate signal is provided to the vector, which includes a variety of regulatory elements, such as viruses that drive expression of genes in the host cell and enhancers / promoters derived from mammals. The most prevalent drug selection markers for establishing permanent and stable cell clones expressing products are included in the expression vectors of the present invention because they are factors that associate the expression of drug selection markers with the expression of the polypeptide.
[554] More specifically, the present invention relates to an expression vector comprising a CanIon protein, preferably a nucleic acid encoding a CanIon protein, variant or fragment thereof of the SEQ ID No 5 amino acid sequence.
[555] The present invention also relates to recombinant expression vectors useful for the expression of CanIon coding sequences, wherein the vectors comprise SEQ ID No 4 nucleic acids.
[556] Recombinant vectors comprising nucleic acids carrying CanIon-related double allelic markers also belong to the invention. In a suitable embodiment, the double allele marker is selected from A1 to A18 and its complement, or optionally a dual allele marker of chain imbalance.
[557] Some of the elements that can be found in the vectors of the present invention are described in more detail in the following sections.
[558] The invention also provides for a) insertion of an endogenous (heterologous) polynucleotide into an endogenous chromosomal DNA of a target gene, b) an endogenous chromosomal DNA deletion, and / or c) a substituted vertebrate to an exogenous polynucleotide of an endogenous chromosomal DNA. It relates to primary, secondary, perpetuated homologous recombinant host cells of origin, preferably of mammalian origin, in particular of human origin. Insertion, deletion or substitution of a polynucleotide sequence may occur in the coding sequence of the target gene and / or regulatory region, eg, a promoter and enhancer sequence operably linked with the target gene.
[559] The present invention also relates to methods of making homologous recombinant host cells in vitro or in vivo, wherein the expression of target genes that are not normally expressed in the cells is altered. Suitably, such modifications induce the expression of the target gene under normal growth conditions or under conditions suitable for producing the polypeptide encoded by the target gene. The method consists of the following steps: (a) transfecting a cell with a polynucleotide construct in vitro or in vivo, the polynucleotide construct comprising (i) a targeting sequence; (ii) regulatory sequences and / or coding sequences; (iii) a non-conjugated splice donor site, if necessary, to produce transfected cells; (b) Transfected cells in vitro or in vivo are maintained under conditions suitable for homologous recombination.
[560] The present invention also relates to a method of altering the expression of a target gene in a cell in vitro or in vivo, wherein the gene is not normally expressed in the cell and the method consists of the following steps: (a) In vitro Or transfecting a cell with a polynucleotide construct in vivo, the polynucleotide construct comprising (i) a targeting sequence; (ii) regulatory sequences and / or coding sequences; (iii) a non-conjugated splice donor site, if necessary, to produce transfected cells; (b) Transfected cells in vitro or in vivo are maintained under conditions suitable for gene expression.
[561] The invention also relates to a method of making a polypeptide according to the invention by altering the expression of targeted endogenous genes in cells in vitro or in vivo, wherein said genes are not normally expressed in cells and the method comprises the following steps: (A) transfecting a cell with a polynucleotide construct in vitro, said polynucleotide construct comprising (i) a targeting sequence; (ii) regulatory sequences and / or coding sequences; (iii) a non-conjugated splice donor site, if necessary, to produce transfected cells; (b) maintaining the transfected cells in vitro or in vivo under conditions suitable for homologous recombination to produce homologous recombined cells; c) The polypeptide is produced by maintaining the transfected cells in vitro or in vivo under conditions suitable for gene expression.
[562] The invention also relates to polynucleotide constructs that alter the expression of a targeted gene in a cell, wherein the gene is not normally expressed. This occurs when the polynucleotide construct is inserted into the chromosomal DNA of the target cell, wherein the polynucleotide construct is constructed as follows: (i) the targeting sequence; (ii) regulatory sequences and / or coding sequences; (iii) non-conjugated splice-donor sites, if necessary. The invention also includes the polynucleotide constructs described above, wherein the construct further comprises a polynucleotide encoding the polypeptide and is paired with a targeted endogenous gene following homologous recombination with chromosomal DNA.
[563] The compositions can be produced by methods and techniques known in the art (US Pat. WO 94/12650, Scientific Paper Koller et al. Proc, 1994, Natl. Acad. Sci. USA 86: 8932-8935 (1989).
[564] 1.Overall features of expression vectors according to the present invention
[565] Recombinant vectors according to the present invention include YAC (yeast artificial chromosome), BAC (bacterial artificial chromosome), phage, phagemid, cosmid, plasmid, or chromosome, non-chromosome, semi-synthetic, synthetic DNA Linear DNA molecules and the like.
[566] Such recombinant vectors may comprise transcription units comprising the following assemblies:
[567] (1) Genetic elements that play a regulatory role in gene expression, such as promoters or enhancers. Enhancers are cis-acting elements containing DNA on the order of 10 to 300 bp that act on the promoter to increase transcription.
[568] (2) a structure or coding sequence that is transcribed into mRNA and finally translated into a polypeptide, said structure or coding sequence being operably linked to a regulatory element identified in (1).
[569] (3) Appropriate transcription initiation and termination sequences. Structural units used in yeast or eukaryotic expression systems include leader sequences that allow for extracellular release of translated proteins by host cells. Alternatively, N-terminal residues may be included if the recombinant protein is expressed without a leader or delivery sequence. Such residues may or may not be subsequently cleaved from the expressed recombinant protein to provide the final product.
[570] In general, recombinant expression vectors include promoters of origin from replication, selection markers that allow transformation of host cells, and highly expressed genes to induce transcription of downstream structural sequences. Heterologous structural sequences are combined into appropriate initiation and termination sequences and leader sequences that can induce the release of translated proteins into the periplasmic space or extracellular medium. In certain embodiments wherein the vector transfects and expresses the desired sequence in a mammalian host cell, the preferred vector is a starting point of replication, a suitable promoter, an enhancer, any necessary ribosome binding site, a polyadenylation signal, a splice in the desired host. Donor and acceptor sites, transcription termination sequences, 5′-side non-transcription sequences. DNA sequences derived from the SV40 viral genome, such as the SV40 origin, early promoter, enhancer, splice, polyadenylation signal, can be used to provide non-transcribed genetic elements.
[571] In vivo expression of the CanIon polypeptide, fragment or variant thereof of SEQ ID No 5 may be useful for correcting genetic defects associated with expression of native genes or production of biologically inactive CanIon proteins in a host microorganism.
[572] Accordingly, the present invention relates to recombinant expression vectors designed for the in vivo production of CanIon polypeptides, fragments or variants thereof of SEQ ID No 2 by introducing appropriate genetic material into a patient to be treated. The genetic material can be introduced in vitro into cells previously extracted from the patient, after which the modified cells can be reintroduced directly in vivo into the appropriate tissue of the patient.
[573] 2. Adjustable element
[574] Promoter
[575] Promoter regions suitable for use in the expression vector according to the present invention are selected in consideration of the cell host in which the heterologous gene is expressed. Promoters used to regulate the expression of a nucleic acid sequence of interest have little meaning as long as it can induce the expression of the nucleic acid in the target cell. Thus, when targeting human cells, it is desirable to locate nucleic acid coding regions adjacent to or under a promoter that can be expressed in human cells, such as a human or viral promoter.
[576] Suitable promoters may be heterologous to the nucleic acid to control expression or endogenous to a native polynucleotide comprising a coding sequence that is alternatively expressed. In addition, the promoter is heterologous to the recombinant vector sequence into which the structural promoter / coding sequence is inserted.
[577] The promoter region can be selected from any desired gene using, for example, a CAT (chloramphenicol transferase) vector, preferably pKK232-8 and pCM7 vectors.
[578] Preferred bacterial promoters are LacI, LacZ, T3 or T7 bacterial phage RNA polymerase promoters, gpt, lambda PR, PL, trp promoter (EP 0036776), polyhedrin promoter, p10 protein promoter (Kit Novagen) derived from baculovirus ( Smith et al., 1983; O'Reilly et al., 1992), lambda PR promoter or trc promoter.
[579] Eukaryotic promoters include early CMV, HSV thymidine kinase, early and late SV40, LTR derived from retroviruses, and mouse metallothionein-L. Easy selection of vectors and promoters will be apparent to those skilled in the art.
[580] The choice of promoter is apparent to those skilled in the art of genetic engineering. See, for example, the process described in Sambrook et al. (1989) or Fuller et al. (1996).
[581] Other adjusting elements
[582] When using cDNA inserts, it is desirable to include a polaradenylation signal in order to achieve proper polyadenylation of the gene transcript. The nature of the polyadenylation signal does not have a significant meaning in the successful practice of the present invention, and any sequence, such as human growth hormone and SV40 polyadenylation signal, can be used. Terminators are also considered elements of the expression cassette. These elements serve to enhance message levels and minimize readability from cassettes to other sequences.
[583] 3. Selective Markers
[584] Such markers deliver identifiable changes to the cells to allow early identification of the cells containing the expression construct. Selectable marker genes for selection of transformed host cells are preferably dihydrofolate reductase or neomycin resistance in eukaryotic cell culture; TRP1 in S. cerevisiae ; Escherichia coli (E. coli) tetracycline, Levan inde Saqqara claim, the latter marker for in ripam peusin or ampicillin resistance or Mycobacteria (mycobacteria) in is a negative selection marker.
[585] 4. Preferred Vector
[586] Bacteria vector
[587] Expression vectors useful for use in bacteria may include bacterial origins of replication derived from commercially available plasmids containing selectable markers and the pBR322 (ATCC37017) genetic element. Such commercial vectors include, for example, pKK223-2 (Pharmacia, Uppsala, Sweden) and GEM1 (Promega Biotec, Madison, WI, USA).
[588] Many other suitable vectors are known in the art, for example, the following bacterial vectors are commercially available: pQE70, pQE60, pQE-9 (Qiagen), pbs, pD10, phagescript, psiX174 pbluescript SK, pbsks, pNH8A, pNH16A, pNH18A, pNH46A (Stratagene); ptrc99a pKK223-3, pKK233-3, pDR540, pRIT5 from Pharmacia; pWLNEO, pSV2CAT, pOG44, pXT1, pSG (Stratagene); pSVK3 pBPV, pMSG, pSVL from Pharmacia; pQE-30 (QIAexpress).
[589] Bacterial phage vector
[590] P1 bacterial phage vectors may comprise many inserts of 80-100 kb.
[591] The construction of P1 bacterial phage vectors such as p158 or p158 / neo8 is described in Stemberg (1992, 1994). Recombinant P1 clones comprising CanIon nucleotide sequences can be considered for inserting polynucleotides greater than 40 kb (Linton et al., 1993). Suitable protocols for making P1 DNA for transduction experiments are described in McCormick et al. (1994). Briefly, E. coli (preferably, NS3529) carrying the P1 plasmid is incubated overnight in liquid medium containing 25 μg / ml kanamycin. P1 DNA is prepared from E. coli by alkaline lysis using the Qiagen Plasmid Maxi kit (Qiagen, Chatsworth, CA USA). P1 DNA is purified from bacterial lysates using the wash and elution buffers included in the kit on two Qiagen-tip 500 columns. Thereafter, phenol / chloroform extraction is performed, followed by precipitation of DNA with 70% ethanol. After dissolving DNA in TE (10 mM Tris-HCL, pH 7.4, 1 mM EDTA), the concentration of DNA is assessed by spectrophotometry.
[592] When expressing P1 clones carrying CanIon nucleotide sequences in transgenic animals, typically transgenic mice, for example, P1 DNA fragments are cleaved by cleaving P1 DNA at low frequency cleavage sites in the P1 polylinker (SfiI, NotI or SalI). It is preferred to remove the vector sequence from the. The P1 insert is then purified from the vector sequence on a pulse-field agarose gel by the method applied in the separation of DNA from YAC (Schedl et al. 1993a; Peterson et al., 1993). If necessary, purified insert DNA is concentrated in a Millipore Ultrafree-MC Filter Unit (Millipore Bedford, MA USA-30,000 molecular weight limit), followed by 100 mM NaCl, 30 μM in a microdialysis membrane (type VS, 0.025 μm, Millipore). Dialysis is carried out in microinjection buffer (10 mM Tris-HCl, pH 7.4, 250 μm EDTA) containing spermine, 70 μM spremidine. The integrity of the purified P1 DNA insert is assessed by electrophoresis and ethidium bromide staining on a 1% agarose (Sea Kem GTG: FMC Bio-products) pulse-field gel.
[593] Baculovirus vector
[594] Suitable vectors for the CanIon polypeptide, fragment or variant thereof of SEQ ID No 5 are baculovirus vectors capable of propagation in insect cells and insect cell lines. A particularly suitable host vector system is the pVL1392 / 1393 baculovirus delivery vector (Pharmingen) used to transfect SF9 cell line (ATCC N ° CRL 1711) derived from Spodoptera frugiperda .
[595] Other vectors suitable for CanIon polypeptides, fragments or variants thereof of the SEQ ID No 5 in baculovirus expression systems include those identified by Chai et al. (1993), Vlasak et al. (1983), Lenhard et al. (1996). Included.
[596] Virus vector
[597] In certain embodiments, the vector is derived from adenovirus. Suitable adenovirus vectors according to the present invention are those found by Feldman and Steg (1996) or Ohno et al. (1994). Other suitable recombinant adenoviruses according to certain embodiments of the present invention are type 2 or 5 human adenoviruses (Ad2, Ad 5) or adenoviruses of animal origin (French N ° FR-93.05954).
[598] In general, retroviruses and adeno-associated viral vectors are recombinant gene delivery systems selected for delivery of exogenous polynucleotides to mammals, including humans, in vivo. These vectors provide for efficient delivery of genes to cells, and the delivered nucleic acid is stably integrated into the host's chromosomal DNA.
[599] Retroviruses which are particularly suitable for the preparation or construction of retroviral in vitro or in vivo gene transporters according to the present invention are retroviruses selected from Mink-Cell concentrated induction virus, murine sarcoma virus, reticuloendoteliosis virus, Raus sarcoma virus. to be. Particularly preferred murine leukemia viruses include 4070A and 1504A viruses, Abelson (ATCC No VR-999), Friend (ATCC No VR-245), Gross (ATCCNo VR-590), Rauscher (ATCC No VR-998), Moloney murine leukemia virus (ATCC No VR-190; PCT Application WO 94/24298). Particularly preferred Raus sarcoma viruses include Bryan high titer (ATCC Nos VR-334, VR-657, VR-726, VR-659, VR-728). Other suitable retroviral vectors are described in Roth et al. (1996), PCT application WO 93/25234, PCT application WO 94/06920 Roux et al., 1989, Julan et al., 1992, Neda et al., 1991 .
[600] Another viral vector system in the present invention consists of adeno-associated virus (AAV). Adeno-associated viruses are naturally occurring defect viruses that require other viruses, such as adenovirus or herpes virus, as secondary viruses for effective replication and life cycles (Muzyczka et al., 1992). This is one of the few viruses that integrates DNA into non-dividing cells and shows a high frequency of stable integration (Flotte et al., 1992; Samulski et al., 1989; McLaughlin et al., 1989). Another advantage of AAV is due to a decrease in the ability to transduce primary cells associated with the transformed cells.
[601] BAC vector
[602] The bacterial artificial chromosome (BAC) cloning system (Shizuya t al., 1992) was developed to stably maintain large genomic DNA fragments (100-300 kb) in E. coli . Suitable BAC vectors include the pBeloBAC11 vector disclosed by Kim et al. (1996). The BAC library is prepared with the vector using size-selected genomic DNA partially digested with enzymes that allow ligation from the vector to the Bam HI or HindIII site. The sides of these cloning sites are final by RNA transcription or PCR methods. T7 and SP6 RNA polymerase transcription initiation sites that can be used to make probes. After the construction of the BAC library in E. coli , the BAC DNA is purified from the host cell into a spiral helix. Prior to converting these circular molecules linearly, sizing and introduction of BAC into receptor cells proceed. The cloning site is flanked by two NotI sites that allow cleavage of the cloned segment from the vector by NotI cutting. Alternatively, the DNA insert included in the pBeloBAC11 vector can be linearized by treating the BAC vector with a commercially available enzyme lambda terminase that causes cleavage at a single cosN site, but this cleavage method can be linearized with the insert DNA. Results in a full length BAC clone comprising both and BAC sequences.
[603] 5. Delivery of Recombinant Vectors
[604] In order to achieve expression of polynucleotides and polynucleotide constructs according to the invention, these constructs must be delivered to cells. Such delivery can be accomplished in vitro, such as in laboratory procedures for transforming a cell line, or in vivo or as in the treatment of certain diseases.
[605] One mechanism is viral infection, where the expression construct is integrated into infectious viral particles.
[606] The present invention contemplates some non-viral methods for delivering polynucleotides to cultured mammalian cells, including calcium phosphate precipitation (Graham et al., 1973; Chen et al., 1987), DEAE-dextran ( Gopal, 1985), electroporation (Tur-Kaspa et al., 1986; Potter et al., 1984), direct microinjection (Harland et al., 1985), DNA-loaded liposomes (Nicolau et al., 1982). Fraley et al., 1979), receptor-mediated transfection (Wu and Wu, 1987; 1988) and the like. Some of these techniques can be successfully applied for in vivo and external use.
[607] Once the expression polynucleotide is delivered to the cell, it can be stably integrated into the genome of the recipient cell. Such integration can proceed through cognate position and orientation via homologous recombination (gene substitution) or can be integrated into random non-specific positions (gene complement). In other embodiments, the nucleic acid may be stably maintained in the cell as separate episomal DNA segments. Such nucleic acid segments or "episomes" encode sequences that enable replication and maintenance independently of or in conjunction with the host cell cycle.
[608] In certain embodiments, a method of delivering a protein or peptide into a cell of a vertebrate in vivo comprises a formulation of a tissue comprising a cell comprising a physiologically acceptable carrier and a formulation containing a naked polynucleotide that operably encodes the polypeptide of interest. Introducing into the interstitial space, wherein the naked polynucleotide is taken up into the interior of the cell and exhibits a physiological effect. This may apply in particular to in vitro delivery, but may also apply to in vitro delivery.
[609] Compositions used in vitro and in vivo comprising "nacin" polynucleotides are described in PCT application WO 90/11092 (Vical Inc.), PCT application WO 95/11307 (Institut Pasteur, INSERM, Universite d'Ottawa), Tacson et. al. (1996), Huygen et al. (1996).
[610] In another embodiment of the present invention, delivery of the nasine polynucleotides according to the present invention, including the polynucleotide constructs of the present invention, to a cell is carried out by a particle bomb (biological), wherein the particles are DNA-coated at high speed. As microprojectiles, these DNA pass through cell membranes to enter cells (Klein et al. (1987)).
[611] In a suitable embodiment, the polynucleotides of the invention are entrapped in liposomes (Ghosh and Bacchawat, 1991; Wong et al., 1980; Nicolau et al., 1987).
[612] In certain embodiments, the present invention provides a composition for in vivo production of a CanIon protein or polypeptide according to the present invention. The composition consists of a naked polynucleotide that operably encodes such a polypeptide that, when dissolved in a physiologically acceptable carrier and introduced into the tissue, causes cells of the tissue to express the protein or polypeptide.
[613] The amount of vector injected into the desired host microorganism depends on the site of injection. Generally, 0.1 to 100 μg is injected into the animal body, preferably the mammalian body, eg the mouse body.
[614] In another embodiment, the vectors according to the invention can be introduced into host cells, preferably host cells harvested from treated animals, more preferably somatic cells (eg myocytes). In a subsequent step, the cells are transformed with a vector encoding the desired CanIon polypeptide or fragment thereof which reintroduces the animal body to deliver the recombinant protein locally or systemically.
[615] Cell host
[616] Another object of the invention is a host that is transformed or transfected with a polynucleotide of the invention, in particular a CanIon regulatory polynucleotide, or a polynucleotide comprising the coding sequence of a CanIon polypeptide, fragment or variant thereof of SEQ ID Nos. It is concerned with cells. The present invention also includes host cells transformed (prokaryotic) or transfected (eukaryotic) with the aforementioned recombinant vectors. More specifically, the cell host of the present invention is a polynucleotide disclosed in the "Genomic Sequence of the CanIon Gene" section, the "CanIon cDNA Sequence" section, the "coding region" section, the "polynucleotide construct" section, the "oligonucleotide probe and primer" section. Nucleotides may be retained.
[617] Another recombinant host cell according to the present invention comprises a polynucleotide having said double allele marker selected from A1 to A18 and complements thereof.
[618] Other recombinant cell hosts according to the invention may carry any of the vectors identified herein, more specifically any of the vectors identified in the "Recombinant Vectors" section.
[619] Suitable host cells for use as recipients for the expression vectors of the present invention are as follows:
[620] a) Prokaryotic host cells: Escherichia coli (Escherichia coli) (IEDH5-α strain), Bacillus subtilis (Bacillus subtilis), Salmonella typhimurium Moorim (Salmonella typhimurium), and Pseudomonas (Pseudomonas), Streptomyces (Streptomyces), Staphylococcus Strains derived from species such as Staphylococus ;
[621] b) eukaryotic host cells: HeLa cells (ATCC N ° CCL2; N ° CCL2.1; N ° CCL2.2), Cv 1 cells (ATCC N ° CCL70), COS cells (ATCC N ° CRL1650; N ° CRL1651), Sf-9 cells (ATCC ° CRL1711), C127 cells (ATCC N ° CRL-1804); 3T3 (ATCC N ° CRL-6361), CHO (ATCC N ° CCL-61), human height 293 (ATCC N ° 45504; N ° CRL-1573), BHK (ECACC N ° 84100501; N ° 84111301);
[622] c) other mammalian host cells.
[623] CanIon gene expression in mammals, typically human cells, is incomplete, or alternatively proceeds with the insertion of a genomic sequence or cDNA sequence that replaces the CanIon gene counterpart in the genome of the animal cell with the CanIon polynucleotide according to the present invention. Such genetic modifications can be achieved by homologous recombination using the specific DNA constructs described above.
[624] One of the cell hosts that can be used is a mammalian conjugate, for example a murine conjugate. For example, the murine conjugate is 1 ng / ml (BAC insert) in a purified DNA molecule of interest, for example 10 mM Tris-HCl containing 100 mM NaCl 30 µM spermine, 70 µM spermidine, pH 7.4, 250 µm EDTA. Purified DNA molecules adjusted to a range from 3 ng / μl (P1 bacteriophage insert) concentration can be microinjected. If the microinjected DNA is large, polyamines and high salt concentrations can be used to avoid physical destruction of the DNA (Schedl et al. (1993b)).
[625] Polynucleotides according to the present invention, including the DNA constructs disclosed herein, can be introduced into embryonic stem (ES) cell lines, preferably mouse ES cell lines. ES cell lines are derived from omnipotent, non-committed cells of the inner cell mass of preimplantation blastocysts. Suitable ES cell lines are as follows: ES-E14TG2a (ATCC n ° CRL-1821), ES-D3 (ATCC n ° CRL1934, n ° CRL-11632), YS001 (ATCC n ° CRL-11776), 36.5 (ATCC n ° CRL-11116). ES cells are cultured in the presence of stunt inhibited feeder cells that provide a suitable signal to preserve the embryo phenotype and serve as a matrix for ES cell adhesion, in order to remain uncommitted. Appropriate feeder cells are sustained in culture as found in Abbondanzo et al. (1993) and almost inhibited in growth by the presence of inhibitory concentrations of LIF as indicated by UV or Pease and Williams (1990) as shown by Robertson (1987). Primitive embryonic fibroblasts established from the tissues of 13-14 day old embryos of all mouse strains.
[626] Constructs in host cells can be used in conventional methods of producing gene products encoded by recombinant sequences.
[627] Following transformation of the appropriate host and incubation of the host to an appropriate cell density, the selection promoter is induced by suitable means, such as temperature change or chemical induction, and the cells are further cultured for a period of time.
[628] Typically, cells are harvested by centrifugation and destroyed by physical or chemical means, and the resulting crude extract is stored for further purification.
[629] Microbial cells used for expression of proteins can be destroyed by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption or the use of cytolytic agents. Such methods are known in the art.
[630] Transgenic animal
[631] As used herein, "transgenic animal" or "host animal" refers to an animal whose genome is genetically and artificially engineered to comprise one of the nucleic acids according to the present invention. Preferred animals are non-human animals, in which the genome is artificially and genetically modified by insertion of a nucleic acid according to the invention and selected from Mus (eg mice), Rattus (eg mice), Oryctogalus (eg rabbits) Included are animals belonging to the genus. In one embodiment, the present invention includes human human host mammals and animals carrying a CanIon gene that has been destroyed by homologous recombination with a recombinant vector according to the present invention or a hit vector of the present invention.
[632] The transgenic animal of the present invention comprises a recombinant or synthetic DNA sequence cloned into a plurality of cells, more specifically a DNA sequence encoding a CanIon coding sequence, a CanIon regulatory polynucleotide, a polynucleotide construct or an antisense polynucleotide as disclosed herein. Retain the isolated or purified nucleic acid.
[633] In general, transgenic animals according to the present invention possess the polynucleotides, recombinant vectors, and cellular hosts disclosed herein. In particular, the transgenic animals of the present invention comprise a "genome sequence of CanIon gene" section, a "CanIon cDNA sequence" section, a "coding region" section, a "polynucleotide construct" section, an "oligonucleotide probe and primer" section, a "recombinant vector" And the polynucleotides identified in the "section," cell host "section.
[634] Another transgenic animal according to the present invention comprises a polynucleotide having said double allele marker selected from A1 to A18 and complement thereof in somatic and / or germ cells.
[635] In a suitable first embodiment, these transgenic animals, particularly transgenic animals having a genome in which one or several copies of a polynucleotide encoding a native CanIon protein or alternatively a mutant CanIon protein, are involved in various It can be a good experimental model for studying pathology.
[636] In a suitable second embodiment, these transgenic animals express desired polypeptides under the control of regulatory polynucleotides of the CanIon gene, ultimately achieving high yields of the protein synthesis and tissue specific expression of such proteins.
[637] The design of the transgenic animal according to the present invention can be made by conventional techniques known in the art. US Pat. No. 4,873,191 (Oct. 10, 1989) for the production of transgenic animals, in particular transgenic mice; 5,464,764 (Nov 7, 1995); 5,789,215 (Aug 4, 1998).
[638] The transgenic animal of the present invention can be made into a process that results in an animal having a genome incorporating exogenous genetic material. This process includes obtaining a genetic material encoding a CanIon coding sequence or a portion thereof, a CanIon regulatory polynucleotide, or a DNA sequence encoding a CanIon antisense polynucleotide as disclosed herein.
[639] Recombinant polynucleotides of the invention are inserted into an embryo or ES stem cell line. This insertion is done by electroporation (Thomas et al. (1987)). The cells to be electroporated are screened (selection via selectable marker, PCR or Southern blot analysis) to find positive cells incorporating exogenous recombinant polynucleotides into their genome, preferably by homologous recombination. Positive-negative selection procedures that can be used in accordance with the present invention are described in Mansour et al. (1988).
[640] Positive cells are then isolated, cloned and injected into 3.5 day blastocysts obtained from mice (Bradley (1987)). The blastocyst is then inserted into a female host animal and grown for a period of time.
[641] Alternatively, positive ES cells are contacted with 2.5-day old 8-16 cell stage embryos (lost embryos) (Wood et al. (1993), Nagy et al. (1993)) and ES cells are absorbed and developed into germ cells. Form extensive colonies of blastocysts, including cells.
[642] Descendants of female hosts are examined to identify wild-type animals and animals that have been transgenic, ie, have inserted exogenous DNA.
[643] Accordingly, the present invention relates to transgenic animals carrying nucleic acids, recombinant expression vectors or recombinant host cells according to the invention.
[644] Recombinant cell line derived from the transgenic animal of the present invention
[645] Another object of the invention relates to recombinant host cells obtained from the transgenic animals disclosed herein. In one embodiment, the invention includes cells derived from human host mammals and animals other than humans carrying a CanIon gene that has been destroyed by homologous recombination with a recombinant vector or hit vector according to the invention.
[646] Recombinant cell lines can be established by transfection of primitive cell cultures with vectors expressing onc -genes, such as, for example, the SV40 large T antigen, in vitro from cells obtained from any tissue of another transgenic animal. Chou (1989), Shay et al. (1991))
[647] Screening method of CanIon modulators and interacting compounds
[648] In various embodiments, the present invention provides compounds that interact with, bind to, activate, or inhibit their expression or activity with CanIon polypeptides, channels, polynucleotides. Such compounds may be organic or inorganic, including polypeptides, polynucleotides, lipids, carbohydrates, nucleotides, amino acids or small molecule inhibitors or actives. As found herein, such compounds are useful for the treatment or prevention of various diseases or conditions. Suitably, inhibitors of CanIon activity or expression are used to treat or prevent mental disorders such as schizophrenia or bipolar disorder.
[649] How to Screen CanIon Channel Modulators
[650] Compounds that can bind CanIon and compounds that modulate CanIon function are important for the treatment of diseases. Voltage-gated ion channels are generally established drug targets in that they are pharmacologically accessible, encoded by various genes, and move to multiple bond protein assemblies, thus resulting in high functional and anatomical specificity. In addition, because ion channel opening and closing, which affects the movement of charged voltage sensitive amino acids, induces a change in conformation, the ion channel binds only to channels in the conducting (activated) or non-conductive (inactivated) state, for example. It enables the design of dependent molecules.
[651] In addition, a number of calcium channel modulators have been proven effective in the treatment or prevention of various diseases and conditions. For example, calcium channel inhibitors include a variety of heart diseases and abnormalities (e.g. angina pectoris, arrhythmia, hypertension) and CNS and neurological diseases (e.g. migraines, neurological effects of seizures, manic, neuroleptic-induced delayed dyskinesia, bipolar Disorders, pain, epilepsy, etc.). In addition, calcium channel agonists have been found to be effective in reducing and diluting the duration of various uses, such as local anesthetics. Antagonists and agonists of CasIon channels are similarly effective in the treatment or prevention of these diseases and disorders. For example, CanIon antagonists are effective in the treatment or prevention of schizophrenia and bipolar disorder.
[652] Since voltage-gated ion channels do not require agonist binding for activation, the compounds screen for functional CanIon channels. Assays include functional radioligand binding assays applied to cells (vesicles or membranes) that express native or cloned channels, or whole cells. Functional charter cell analysis utilizes electrophysical techniques such as patch clamping. The analysis relates to voltage-gated channel types, preferably L, N, T type channels. Dynamic ion flow through the channel can be measured, for example, by fluorescence, end-point radiotracer, and cell viability techniques.
[653] In addition, the assay may use a variety of toxins, venoms, or compounds that bind to and open channels (Denyer et al., Drug Disc. Today 3 (7): 323-332 (1998). Uses a number of known calcium channel agonists and antagonists as positive or negative controls, for example Examples of suitable known calcium channel antagonists include phenylalkylamines (eg verapamil), benzothiazepines (eg diltia). Gems), dihydropyridine (e.g. nifedipine); calcium channel agonists include FPL-64176 and BAYK 8644; sodium channel agonists include betrachotoxin; sodium channel antagonists include spiradoline, Mexyltin, U-54494A ((+/-)-cis-3,4-dichloro-N-methyl-N- [2- (1-pyrrolidinyl) -cyclohexyl] -benzamide) Compounds Induce Specific Binding to or Modulating CanIon Channels It may be used as a "lead" compound from the molecular role in the design or discovery of molecules.
[654] In a suitable embodiment, the assay of the present invention comprises a method of screening candidate substances, which method consists of the following steps:
[655] a) a recombinant host cell expressing a polynucleotide encoding (i) a sample or host cell carrying a polypeptide comprising a CanIon protein or fragment thereof, or (ii) a polypeptide comprising a CanIon protein or fragment thereof; ;
[656] b) obtaining a candidate substance;
[657] c) contacting said candidate substance with a host cell;
[658] d) determine the effect of the candidate on CanIon activity.
[659] Measurement of candidate substance effects on CanIon activity can be accomplished by methods known in the art. Suitably, the effect of the candidate on CanIon activity is an agonist or antagonist effect. In general, the compound inhibits CanIon if the ability to transfer ions (eg, Ca 2+ or Na +) decreases. As the ability to transfer ions increases, the compound promotes CanIon.
[660] CanIon activity can be detected by any suitable means. In a suitable example, CanIon activity is detected by measuring signaling phenomena. Signaling phenomena may include appropriate changes in the molecular characteristics or parameters of a cell. Unlimited examples of signaling phenomena include changes in ion flow, such as changes or occurrences of Ca2 + or Na +, or K + flow or enzyme activation. do.
[661] In one aspect, ion flow can be monitored by measuring the electrophysical properties of the CanIon channel using, for example, techniques to measure total cell current in single cell or membrane patches. In another example, fluorescent or radioactive labels can be used to detect known CanIon-binding compound migration, or ion (eg, labeled Ca 2+ or Na +) flow in cells. Indicators for the physiological parameters of the cells can be used, for example fluorescence indicators for cell survival. In another example, a change in the physical location of the indicator can be identified, for example, by the use of fluorescent activated cell sorting to confirm the release or uptake of the physiological indicator.
[662] Samples for use in the assays of the invention are i) a host cell expressing a polypeptide comprising a CanIon protein or fragment thereof, or ii) a recombinant host cell expressing a polynucleotide encoding a polypeptide comprising a CanIon protein or fragment thereof. It contains. Suitably, the CanIon assay of the present invention includes the use of recombinant host cells expressing a functional CanIon polypeptide. The host cell may express or comprise a functional alpha subunit of the CanIon channel, one or more additional ion channel subunits, or an ion channel complex comprising CanIon. Suitably, host cells are used that possess low endogenous ion channel expression or have low background ions, in particular Ca 2+ and / or Na + conductivity.
[663] Radioligand Bond
[664] In one aspect, the CanIon channel can be screened by detecting a high affinity ligand that binds to the site of interest of CanIon and preferably possesses the desired regulatory effect and detects the ability of the test compound to substitute for the labeled ligand. A list of toxicological / pharmacological agents used for voltage switching (Ca2 +, Na +, K +) is provided by Denyer et al. (Supra). In general, these methods are suitable for detecting compounds that bind to the same site or are allosterically bound to the site with labeled ligands but do not provide information relating to the agonist or antagonist properties.
[665] Cell-based Fluorescence and Radiotracer Analysis
[666] In another assay, CanIon function can be monitored by measuring changes in intracellular concentrations of permeable ions with fluorescent-ion indicators or radiolabeled ions.
[667] Typically, ion channels, such as Na + channels, are inactivated within seconds after voltage stimulation. Ca2 + channels show little inactivation and can be opened with high K + depolarization. In cell-based fluorescence and radiotracer assays, CanIon channels can be activated with toxins or any test compound or high K + depolarization to open for extended periods (maximum minutes).
[668] In fluorescence based assays, fluorescent Ca2 + dyes (Fluo-3, Calcium Green-1, Molecular Probes, OR, USA) are available. Ca2 + channels can be activated by depolarizing the membrane with isotonic solution or by depolarizing the Na + channel with toxin or other compound, where the transient movement of fluorescence in the cell is measured for 20 to 60 seconds. Fluorescence measurement systems and devices are described in detail in Denyer et al. (Supra). Radiotracers 22Na + and 14C-guanidine are commonly used for Na + channel analysis and 45Ca2 + is used for Ca2 + channel analysis. In a suitable embodiment identified by Denyer et al. (Supra), Cytostar- T scintillation microplates (Amersham International, UK) are used for high performance CanIon cell based assays.
[669] In further analysis, the Ca2 + function of the ion channel is monitored by measuring the membrane potential with the membrane potential indicator. The high electrical resistance of biological membranes allows for less ion current to pass through the plasma membrane and induce many changes in membrane potential. Thus, voltage analysis can be readily used to detect overall ion flow through the membrane. In general, cell lines are chosen such that the effects are minimized from endogenous ion channels. Various dyes are available as membrane potential indicator dyes, which are subdivided into fast and slow reaction dyes and FRET-based voltage sensor dyes (Auora Biosciences, CA, USA; Gonzalez et al., Drug Disc. Today 4 (9)). : 431-439 (1999).
[670] Cell viability
[671] In cell viability assays, ion channel activity and ion flow are directly related to cell viability. Yeast and mammalian cell systems are available for testing of ion channel targets. For example, a yeast system using ion-specific K + uptake deficient Saccharomyces cerevisiae cell lines is used, where functional K + channels are expressed in the cell lines, thus restoring K + uptake and promoting cell survival (Anderson et al. al., Symp. Soc. Exp. Biol. 48: 85-97 (1994)). This assay for Ca2 + or Na + channels can be used to identify compounds that can block CanIon function. Na + channel analysis using mammalian neuroblastoma cells in mammalian cell systems, such as colorimetric cell viability readout, is also available. Cells treated with open material and Na + / K + pump inhibitors with Na + channels promote lethal intracellular Na + overload. Treatment with test compounds that can block channels improves cell viability, while compounds that enhance channel opening further promote cell death (Manger et al., Anal. Biochem. 214: 190-194 (1993)).
[672] Electrophysiology
[673] Electrophysiological voltage-clamping techniques include the measurement of ion current flow through one or more channels. Membrane voltage is controlled with a single microelectrode and current is measured through a single cell or membrane patch (Hamil, Pfugers Arch. 391, 85-100 (1981)). Thus, the ion current can be measured in the presence of the test compound of interest. Large-scale compound screening systems have been designed (Neurosearch A / S, Glostrup, Denmark; Olesen et al., Voltage gated ion channel modulators, 7-8 December, Philadelphia PA, USA (1995); Denyer et al., Supra).
[674] Screening Methods for Substances Interacting with CanIon Polypeptides
[675] In the present invention, a ligand refers to a molecule such as a protein, peptide, antibody or any chemical synthetic compound capable of binding to a CanIon protein, fragment or variant thereof, or modulating the expression of a polynucleotide encoding CanIon, fragment or variant thereof. it means.
[676] In the ligand screening method according to the present invention, a biological sample or defined molecule to be tested with the putative ligand of the CanIon protein is contacted with a corresponding purified CanIon protein, for example a corresponding purified recombinant CanIon protein produced by a recombinant cell host. To form the putative ligand intermolecular complex to be tested with the protein.
[677] For example, CanIon comprising at least 6 amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably at least 12, 15, 20, 25, 30, 40, 50 or 100 consecutive amino acids Proteins or fragments thereof and drugs or small molecules such as combinatorial chemistry, microdialysis (Wang et al. (1997)) in conjunction with HPLC or affinity capillary electrophoresis methods (Bush et al. (1997)) Form a complex of molecules.
[678] In another method, at least 6 amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably at least 12, 15, 20, 25, 30, 40, 50 or 100 consecutive amino acids Peptides, drugs, fatty acids, lipoproteins or small molecules that interact with CanIon proteins or fragments thereof can be identified by the following assays. The molecule to be tested for binding is labeled with a detectable label such as a fluorescent, radioactive or enzymatic tag and contacted with the immobilized CanIon protein or fragment thereof under conditions capable of specific binding. After removal of the non-specifically bound molecule, the bound molecule is detected by appropriate means.
[679] Another object of the present invention is directed to methods and kits for screening candidate materials for interacting with CanIon polypeptides.
[680] The present invention provides a method for screening a substance that interacts with a CanIon protein, fragment or variant thereof. Due to their ability to covalently or non-covalently bind CanIon proteins, fragments or variants thereof, these materials can be used effectively in vitro and in vivo.
[681] In vitro, such interacting molecules can be used as detection means to confirm the presence of CanIon protein in a sample, preferably a biological sample.
[682] The method for screening candidate materials consists of the following steps.
[683] a) CanIon comprising at least 6 amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably at least 12, 15, 20, 25, 30, 40, 50 or 100 contiguous amino acids Providing a polypeptide comprising or comprising a protein or fragment thereof;
[684] b) obtaining a candidate substance;
[685] c) contacting said polypeptide with a candidate substance;
[686] d) detecting the complex formed between the polypeptide and the candidate substance.
[687] In addition, the present invention relates to a kit for screening a candidate substance that interacts with a CanIon polypeptide, wherein the kit is configured as follows.
[688] a) CanIon comprising at least 6 amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably at least 12, 15, 20, 25, 30, 40, 50 or 100 contiguous amino acids Proteins or fragments thereof;
[689] b) optionally, means useful for detecting the complex formed between the CanIon protein, fragment or variant thereof and the candidate.
[690] In suitable embodiments of the aforementioned kits, the means for detection comprises monoclonal or polyclonal antibodies directed against the CanIon protein, fragments or variants thereof.
[691] Various candidates or molecules can analyze the interaction with the CanIon polypeptide. These substances or molecules include, but are not limited to, native or synthetic organic compounds or molecules of biological origin, such as polypeptides. If the candidate substance or molecule comprises a polypeptide, the polypeptide is an expression product of a phage clone belonging to a phage-based random peptide library or alternatively an expression product of a cDNA library cloned into a vector suitable for conducting a dual-hybrid screening assay Can be.
[692] The present invention also relates to kits useful for carrying out the screening methods described above. Suitably, such kits consist of means useful for detecting CanIon polypeptides, fragments or variants thereof and optionally complexes formed between CanIon proteins, fragments or variants thereof and candidates. In a suitable embodiment, the detection means consists of monoclonal or polyclonal antibodies directed against the CanIon protein, fragments or variants thereof.
[693] A. Candidate Ligands Obtained from Random Peptide Libraries
[694] In certain embodiments of the screening methods, the putative ligand is the expression product of the DNA insert included in the phage vector (Parmley and Smith, 1988). In particular, random peptide phage libraries are used. Random DNA inserts encode peptides of 8 to 20 amino acids in length (Oldenburg KR et al., 1992; Valadon P., et al., 1996; Lucas AH, 1994; Westerink MAJ, 1995; Felici F. et. al., 1991). According to this particular embodiment, the recombinant phage expressing the protein that binds to the immobilized CanIon protein is maintained, and the complex formed between the CanIon protein and the recombinant phage can be immunoprecipitated with polyclonal or monoclonal antibodies against the CanIon protein.
[695] Once the ligand library is constructed from recombinant phage, the phage population is contacted with the immobilized CanIon protein. The complex is then washed to remove non-specifically bound recombinant phage. Phage that specifically binds to the CanIon protein is then eluted with buffer (acidic pH) or immunoprecipitated with monoclonal antibodies made by hybridoma anti-CanIon, and such phage populations are bacteria (eg, E. coli ( E. amplification by over-infection of coli )). The selection step is repeated several times, preferably two to four times, to select more specific recombinant phage clones. The final step is to characterize the peptides produced by the selected recombinant phage clones by expression and isolation in infected bacteria, to express the phage inserts in other host-vector systems or to sequence the inserts included in the selected recombinant phages. It consists of the steps of analysis.
[696] B. Candidate Ligands Obtained by Competition Experiments
[697] Alternatively, at least 6 amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably at least 12, 15, 20, 25, 30, 40, 50 or 100 contiguous amino acids Peptides, drugs or small molecules that bind to CanIon proteins or fragments thereof can be identified by competition experiments. In this assay, the CanIon protein or fragment thereof is immobilized on a surface, for example a plastic plate. Peptides, drugs or small molecules are contacted with an immobilized CanIon protein or fragment thereof with a gradual increase in content in the presence of a detectable label known as CanIon protein ligand. For example, CanIon ligands can be detectably labeled with fluorescent, radioactive or enzymatic tags. The ability of a test molecule to bind a CanIon protein or fragment thereof is confirmed by measuring the content of detectably labeled ligand bound in the presence of the test molecule. When the test molecule is present, the content of known ligands that bind to the CanIon protein or fragment thereof is reduced, suggesting that the test molecule can bind to the CanIon protein or fragment thereof.
[698] C. Candidate Ligands Obtained by Affinity Chromatography
[699] A CanIon protein comprising at least 6 amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably at least 12, 15, 20, 25, 30, 40, 50 or 100 contiguous amino acids Proteins or other molecules that interact with fragments thereof can be found using an affinity column containing the CanIon protein or fragments thereof. The CanIon protein or fragment thereof may be attached to the column by chemical bonding to a suitable column matrix, such as conventional techniques such as Agarose Affi Gel® or other matrices known to those skilled in the art. In some embodiments of the method, the affinity column contains a chimeric protein in which the CanIon protein or fragment thereof is fused with glutathione S transferase (GST). A mixture of cellular proteins or a pool of expressed proteins described above is added to an affinity column. The protein or other molecule that interacts with the CanIon protein or fragment thereof attached to the column can then be isolated and analyzed on a 2-D electrophoretic gel as revealed by Ramunsen et al. (1997). Alternatively, the peptide remaining in the affinity column can be purified and sequenced by electrophoretic based methods. The method can be utilized to isolate antibodies, screen phage display products, or screen phage display human antibodies.
[700] D. Candidate Ligands Obtained by the Optical Biosensor Method
[701] A CanIon protein comprising at least 6 amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably at least 12, 15, 20, 25, 30, 40, 50 or 100 contiguous amino acids Proteins that interact with their fragments can also be screened with optical biosensors (Edwards and Leatherbarrow (1997), Szabo et al. (1995)). This technique enables the detection of intermolecular interactions in real time and does not require labeled labels. This technique is based on surface plasmon resonance (SPR) phenomena. In short, candidate ligand molecules to be tested are attached to a surface (eg, carboxymethyl dextran matrix). The light beam is directed to the side of the surface containing no test sample and reflected by the surface. SPR phenomena reduce the density of reflected light specifically in relation to angle and wavelength. Binding of the candidate ligand material results in a change in refractive index at the surface, which change is perceived as a change in the SPR signal. In the screening of candidate ligand molecules or substances that can interact with the CanIon protein or fragment thereof, the CanIon protein or fragment thereof is immobilized on the surface. The surface consists of cells through which the candidate material to be analyzed passes. The binding of the candidate to the CanIon protein or fragment thereof is detected as a change in the SPR signal. Candidates tested can be proteins, peptides, carbohydrates, lipids or small molecules made with combinatorial chemistry. This technique can also be carried out by immobilizing eukaryotic, prokaryotic or lipid vesicles showing endogenous or recombinantly expressed CanIon proteins.
[702] The greatest advantage of the method is that it is possible to measure the CanIon protein and the intermolecular binding rate that interacts with the CanIon protein. Thus, it is possible to specifically select ligand molecules that interact with CanIon proteins or fragments thereof through strong or relatively weak binding constants.
[703] E. Candidate Ligands Obtained Through Double-Hybrid Screening Assay
[704] The dual-hybrid system is for studying protein-protein interactions in vivo (Fields and Song 1989) and relies on the fusion of bait proteins to the DNA binding domain of the yeast Gal4 protein. This technique is also described in US Pat. Nos. 5,667,973 and 5,283,173 (Fields et al.).
[705] The overall procedure for library screening by double-hybrid analysis can be carried out by the procedure described by Harper et al. (1993), Cho et al. (1998) or Fromont-Racine et al. (1997).
[706] The bait protein or polypeptide may comprise at least 6 amino acids of SEQ ID No 5, preferably at least 8 to 10 amino acids, more preferably at least 12, 15, 20, 25, 30, 40, 50 or 100 consecutive amino acids. Consists of or comprises a CanIon protein or fragment thereof.
[707] More specifically, the nucleotide sequence encoding the CanIon protein, fragment or variant thereof is fused to a polynucleotide encoding the DNA binding domain of the GAL4 protein, wherein the nucleotide sequence is fused to a suitable expression vector, eg pAS2 or pM3. Insert in
[708] A human cDNA library is then constructed in a specially designed vector such that the human cDNA insert is fused with the nucleotide sequence on the vector encoding the transcriptional domain of the GAL4 protein. Suitably the vector used is a pACT vector. Polypeptides encoded by nucleotide inserts of the human cDNA library are referred to as "feed" polypeptides.
[709] The third vector carries a beta galactosidase gene or CAT gene located under the control of a regulatory sequence that responds to the binding of a complete Gal4 protein comprising a detectable marker gene, eg, a transcriptional active domain and a DNA binding domain. . For example, the vector pG5EC can be used.
[710] In addition, two different yeast strains are used. By way of example, two different yeast strains are:
[711] Y190, its phenotype is (MATa, Leu2-3, 112 ura3-12, trpl-901 his3-D200 ade2-101, gal4Dgall80D URA3 GAL-LacZ, LYS GAL-HIS3, cyh);
[712] Y187, its phenotype (MATa gal4 gal80 his3 trpl-901 ade2-101 ura3-52 leu2-3, -112 URA3 GAL-lacZmet), is the opposite mating type of Y190.
[713] In short, 20 μg pAS2 / CanIon and 20 μg pACT-cDNA library co-transform to yeast strain Y190. Transformants are screened for growth in minimal medium containing histidine, leucine, tryptophan but containing histidine synthesis inhibitor 3-AT (50 mM). Positive colonies are screened for beta galactosidase by filter lift assay. Subsequently, double positive colonies ( His + , beta-gal + ) were cultured on a plate lacking histidine and leucine but lacking tryptophan and cyclohexamide (10 mg / ml), without the pAS2 / CanIon plasmid, but the pACT-cDNA library plasmid Select colonies to keep. The resulting Y190 strain was a Gal4 fusion with CanIon or irrelevant control protein; For example, hybridized with Y187 strain expressing cyclophilin B, lamin or SNF1 (Harper et al. (1993), Bram et al. (Bram RJ et al., 1993)) and beta by filter lift analysis. Selected against galactosidase. Beta-gal - in yeast clones after crossing with control Gal4 fusions are false positives.
[714] In another embodiment of the dual-hybrid method according to the invention, the interaction between CanIon, fragment or variant thereof and cellular protein can be assessed using Matchmaker Hybrid System 2 (Catalog No. K1604-1, Clontech). As indicated in the manual accompanying Matchmaker Two Hybrid System 2 (Catalog No. K1604-1, Clotech), nucleic acids encoding CanIon proteins or portions thereof can be inserted into expression vectors to encode the DNA of the yeast transcriptional activator GAL4. Pair with DNA encoding the region. Desired cDNAs, preferably human cDNAs, are inserted into a second expression vector so that they are paired with DNA encoding the activation domain of GAL4. Double expression plasmids are transformed with yeast, and yeast are plated in a selection medium that selects the expression of a selection marker in each expression vector as well as GAL4 dependent expression of the HIS3 gene. Transformants capable of growing in histidine deficient media are screened for GAL4 dependent lacZ expression. Cells positive in both histidine selection and lacZ analysis retain the interaction between CanIon and a protein or peptide encoded by the initially selected cDNA insert.
[715] How to screen for substances that interact with CanIon regulatory sequences
[716] The present invention also relates to a method for screening substances or molecules that can interact with regulatory sequences of the CanIon gene, such as promoter or enhancer sequences.
[717] Can interact with a regulatory sequence of the CanIon gene, more specifically with a nucleotide sequence selected from polynucleotides of 5 'and 3' regulatory regions, fragments or variants thereof, preferably one of the dual allele markers of the invention Nucleic acids encoding proteins present can be identified as single-hybrid systems, as described in the booklet included in the Matchmaker One-Hybrid System kit (Catalog Ref. N ° K1603-1, Clontech). In brief, the target nucleotide sequence is cloned upstream of the selectable reporter sequence and the resulting DNA construct is integrated into the Saccharomyces cerevisiae genome. Then, the yeast cell comprising the reporter sequence in the genome is a library containing an intersequence fusion molecule encoding the activator domain of the yeast transcription factor such as cDNA and GAL4, which encodes a candidate protein that binds to the regulatory sequence of the CanIon gene. Transform. Recombinant yeast cells are plated in liquid medium to select for cells expressing the reporter sequence. The recombinant yeast cells thus selected carry a fusion protein capable of binding to the regulatory sequence of the CanIon gene. The cDNA encoding the fusion protein can then be sequenced and cloned into expression or transcription vectors in vitro. The binding of the encoded polypeptide to the target regulatory sequence of the CanIon gene can be confirmed by techniques known to those skilled in the art, such as gel retardation analysis or DNAse protection analysis.
[718] Gel retardation analysis can be performed independently to screen candidate molecules that can interact with regulatory sequences of the CanIon gene (Fried and Crothers (1981); Garner and Revzin (1981); Dent and Latchman (1993)). These techniques are based on the principle that DNA fragments that bind proteins move slower than the same DNA fragments that do not bind. In short, the target nucleotide sequence is labeled. The labeled nucleotide sequence is then contacted with the entire nuclear extract of the cell containing the transcription factor or with a different candidate to be tested. Interactions between the target regulatory sequences of the CanIon gene and candidates or transcription factors are detected as migration retardation after gel or capillary electrophoresis.
[719] How to screen for ligands that regulate expression of the CanIon gene
[720] Another object of the present invention is a method for screening molecules that regulate the expression of CanIon protein. This screening method consists of the following steps:
[721] a) culturing prokaryotic or eukaryotic cells transfected with a nucleotide sequence encoding a CanIon protein, fragment or variant thereof under the control of its own promoter;
[722] b) contacting the cultured cells with a molecule to be tested;
[723] c) Quantify the expression of CanIon protein, fragment or variant thereof.
[724] In one embodiment, the nucleotide sequence encoding the CanIon protein, fragment or variant thereof comprises at least one allele of the dual allele marker A12 or A16 and its complement.
[725] Using DNA recombination techniques known to those skilled in the art, the CanIon protein encoding the DNA sequence is inserted downstream from the promoter sequence in the expression vector. For example, the promoter sequence of the CanIon gene is included in the nucleic acid of the 5 'regulatory region.
[726] Expression quantitation of CanIon protein can be achieved at the mRNA level or at the protein level. In the latter case, for example, polyclonal or monoclonal antibodies can be used to quantify the content of CanIon protein produced in an ELISA or RIA assay.
[727] In a suitable embodiment, quantification of CanIon mRNA is achieved by quantitative PCR amplification of cDNA obtained by reverse transcription of total mRNA of cultured CanIon-transfected host cells with primers specific for CanIon.
[728] The present invention also relates to methods for screening substances or molecules that can increase or decrease the expression level of the CanIon gene. In this way, those skilled in the art can select useful substances as active ingredients contained in pharmaceutical compositions that have a modulating effect on the expression level of the CanIon gene and treat patients suffering from the diseases described above.
[729] Accordingly, the present invention includes a method for screening candidate substances or molecules for controlling the expression of the CanIon gene, which comprises the following steps:
[730] Providing a recombinant cell host carrying nucleic acid, wherein said nucleic acid comprises a nucleotide sequence of a 5 'regulatory region, a biologically active fragment or variant thereof, located upstream of a polypeptide encoding a detectable protein;
[731] Obtaining a candidate substance;
[732] Identify the ability of the candidate substance to modulate the expression level of a polynucleotide encoding a detectable protein.
[733] In another embodiment, a nucleic acid comprising a nucleotide sequence of a 5 ′ regulatory region, a biologically active fragment or variant thereof, comprises a 5′UTR region of a CanIon cDNA of SEQ ID No 4, a biologically active fragment or variant thereof.
[734] Suitable polynucleotides encoding detectable proteins include polynucleotides encoding beta galactosidase, green fluorescent protein (GFP), chloramphenicol acetyl transferase (CAT).
[735] The present invention also relates to kits useful for carrying out the screening methods described above. Suitably, such kits express the expression of a nucleotide sequence of a 5 ′ regulatory region, biologically active fragment or variant thereof, located upstream of and operably linked to a detectable protein or CanIon protein, a fragment thereof or variant thereof. It consists of recombinant vectors that make it possible.
[736] In another embodiment of the method for screening candidate substances or molecules for controlling the expression of the CanIon gene, the method consists of the following steps:
[737] a) providing a recombinant cell host carrying a nucleic acid, wherein said nucleic acid comprises a 5'UTR sequence of a CanIon cDNA of SEQ ID No 4, a biologically active fragment or variant thereof, and the 5'UTR sequence, a biologically active fragment thereof Or the variant is operably linked to a polypeptide that encodes a detectable protein;
[738] b) obtaining a candidate substance;
[739] c) Identify the candidate's ability to modulate the expression level of a polynucleotide encoding a detectable protein.
[740] In certain embodiments of the above screening methods, a nucleic acid comprising a 5'UTR sequence of CanIon cDNA of SEQ ID No 4, a nucleotide sequence selected from a biologically active fragment or variant thereof, comprises a CanIon 5'UTR sequence described above and an endogenous promoter sequence. Holds.
[741] In another specific embodiment of the above screening method, a nucleic acid comprising a 5'UTR sequence of CanIon cDNA of SEQ ID No 4, a nucleotide sequence selected from a biologically active fragment or variant thereof, is derived from the exogenous promoter of the aforementioned CanIon 5'UTR sequence. Holds the sequence.
[742] In another suitable embodiment, the nucleic acid comprising the 5′UTR sequence of the CanIon cDNA of SEQ ID No 4 or a biologically active fragment thereof carries a dual allele marker selected from A12 or A16 or a complement thereof.
[743] The present invention also relates to a kit for screening a candidate substance that modulates the expression of the CanIon gene, wherein the nucleic acid contains a 5'UTR sequence of CanIon cDNA of SEQ ID No 4, a biologically active fragment or variant thereof. Comprising a recombinant vector comprising a nucleic acid, said 5'UTR sequence, a biologically active fragment or variant thereof, is operably linked to a polypeptide encoding a detectable protein.
[744] The expression level and pattern of CanIon can be analyzed by solution hybridization with long probes found in international patent application WO 97/05277. In brief, the aforementioned CanIon cDNA or CanIon genomic DNA or fragments thereof can be inserted at a downstream cloning site very close to the bacteriophage (T3, T7 or SP6) RNA polymerase promoter to produce antisense RNA. Suitably, the CanIon insert consists of at least 100 contiguous nucleotides of a genomic DNA sequence or cDNA sequence. The plasmid is linearized and transcribed in the presence of ribonucleotides comprising modified ribonucleotides (ie biotin-UTP and DIG-UTP). Excess double labeled RNA hybridizes in solution with mRNA isolated from the cells or tissues of interest. Hybridization is carried out under average stringent conditions (40-50 ° C., 16 hours, 80% formamide, 0.4 M NaCl buffer pH 7.8). Unhybridized probes are removed by cleavage with ribonucleases specific for single-chain RNA (ie, RNases CL3, T1, Phy M, U2 or A). The presence of the biotin-UTP modifications allows the capture of hybrids in microtiter plates coated with streptavidin. The presence of the DIG modifications makes it possible to detect and quantify hybrids by ELISA using anti-DIG antibodies bound to alkaline phosphatase.
[745] Quantitative analysis of CanIon gene expression can also be performed using arrays. Arrays herein refer to one-dimensional, two-dimensional or multidimensional alignments comprising nucleic acids of sufficient length to specifically detect mRNA expression. For example, the array may comprise a plurality of nucleic acids derived from a gene whose expression level should be evaluated. The array may comprise a CanIon genomic DNA, a CanIon cDNA sequence, a complementary sequence or fragment thereof, in particular a sequence comprising at least one of the allele markers according to the invention, preferably at least one of the alleles A1 to A17. have. Preferably the fragment is at least 15 nucleotides. In another embodiment, the fragment is at least 25 nucleotides. In some embodiments, the fragment is at least 50 nucleotides. More suitably, the fragment is at least 100 nucleotides. In another suitable embodiment, the fragment is at least 100 nucleotides. In some embodiments, the fragment is at least 500 nucleotides.
[746] For example, quantitative analysis of CanIon gene expression can be performed with complementary DNA microarrays (Schena et al. (1995 and 1996)). Full length CanIon cDNAs or fragments thereof are amplified by PCR and aligned on silylate microscope slides from 96-well microtiter plates using high speed robotics. The printed array is incubated in a humid chamber to hydrate the array elements and washed once in 0.2% SDS for 1 minute, twice in water for 1 minute, and once in sodium borohydride solution for 5 minutes. The array is soaked in water for 2 minutes at 95 ° C, transferred to 0.2% SDS for 1 minute, washed twice with water, air dried and stored in the dark at 25 ° C.
[747] Cell or tissue mRNA is isolated or purchased commercially and probes are prepared in one reverse transcription. The probes are hybridized for 6-12 hours in a 1 cm 2 microarray under 14 × 14 mm glass curveslip at 60 ° C. The array is washed for 5 minutes in low stringency wash buffer (1 × SSC / 0.2% SDS) at 25 ° C. and then for 10 minutes in high stringency wash buffer (0.1 × SSC / 0.2% SDS) at room temperature. The array is scanned at 0.1 x SSC with fluorescence laser scanning with a custom filter set. Accurate specific expression measurements are obtained by averaging the percentage of independent hybridizations.
[748] Quantitative analysis of CanIon gene expression can also be performed with full length CanIon cDNA or fragments thereof in complementary DNA arrays (Pietu et al. (1996)). Full length CanIon cDNA or fragments thereof are PCR amplified and spotted on the membrane. Thereafter, mRNAs from various tissues or cells are labeled with radionucleotides. After hybridization and washing under controlled conditions, hybridized mRNA is detected by phosphoimaging or autoradiography. Duplicate experiments are then performed, followed by quantitative analysis of the differently expressed mRNAs.
[749] Alternatively, expression analysis using CanIon genomic DNA, CanIon cDNA or fragments thereof can be performed by high density nucleotide analysis (Lockhrt et al. (1996); Sosnowsky et al. (1997)). CanIon genomic DNA or CanIon cDNA sequence, in particular a sequence comprising at least one of the dual allele markers according to the invention, preferably a sequence comprising at least one of the dual allele markers selected from A1 to A17, or complementary thereof Oligonucleotides comprising 15-50 nucleotides derived from the sequence are synthesized directly on the chip (Lockhart et al. Supra), or synthesized and subsequently addressed on the chip (Sosnowski et al., Supra). Suitably, the oligonucleotide is approximately 20 nucleotides.
[750] Suitable compounds such as CanIon cDNA probes labeled with biotin, digoxigenin or fluorescent dyes are synthesized from suitable mRNA populations and then randomly fragmented to an average of 50 to 100 nucleotides in size. The probe then hybridizes to the chip. After washing (Lockhart et al., Supra) and application of different electric fields (Sosnowsky et al., 1997), dyes or labeled compounds are detected and quantified. Perform redundant hybridization Comparative intensity analysis of signals originating from cDNA probes on the same target oligonucleotide on different cDNA samples suggests different expression of CanIon mRNA.
[751] How to inhibit the expression of the CanIon gene
[752] Other therapeutic compositions according to the present invention contain oligonucleotide fragments, antisenses or triple helices of the CanIon nucleotide sequence that inhibit the expression of the corresponding CanIon gene. Suitable fragments of the CanIon nucleic acid sequence include alleles of at least one of the allele markers A1 to A17.
[753] Antisense
[754] A suitable method for using antisense polynucleotides according to the present invention is the procedure found in Sczakiel et al. (1995).
[755] Suitably, antisense is selected from polynucleotides (15-200 bp) complementary to the 5 'end of CanIon mRNA. In other embodiments, a combination of distinct antisense polynucleotides that are complementary to different portions of the desired target gene is used.
[756] Suitable antisense polynucleotides according to the present invention are complementary to the mRNA initiation of CanIon having translation initiation codon ATG or truncation donor or acceptor sites.
[757] Antisense nucleic acids should have a length and melting point to enable the formation of intracellular double helix with sufficient stability to doublely inhibit the expression of CanIon mRNA. Strategies for designing antisense nucleic acids suitable for use in gene therapy are described in Green et al., (1986); Izant and Weintraub, (1984).
[758] In some strategies, antisense molecules are obtained by transcribing strands opposite to those normally transcribed in cells by reversing the orientation of the CanIon coding region in relation to the promoter. Antisense molecules can be transcribed into an in vitro transcription system, for example, a transcription system using T7 or SP6 polymerase to obtain a transcript. Another approach is to transcribe CanIon antisense nucleic acids in vivo by operably linking the DNA carrying the antisense sequence to the antisense sequence in a suitable expression vector.
[759] Other suitable antisense strategies are described in Rossi et al. (1991); International patent applications WO 94/23026, WO 95/04141, WO 92/18522; It is described in European patent application EP 0 572 287 A2.
[760] An alternative to the antisense technology utilized in the present invention is the use of ribozymes (ie, "hammerhead ribozymes") that cleave the corresponding RNA by binding to the target sequence through complementary polynucleotides and hydrolyzing the target site. In short, the cycle of hammerhead ribozyme consists of: (1) sequence specific binding to target RNA via complementary antisense sequences; (2) site-specific hydrolysis of the cleavage motif of the target strand; (3) release of the cleavage product, which leads to another catalytic cycle. Indeed, ribozymes with long chain antisense polynucleotides (at least 30 bases) or long antisense arms are effective. Delivery systems suitable for antisense ribozymes are achieved by covalently binding these antisense ribozymes to lipoaffinity groups or by using liposomes as vectors. Prepare as shown in a suitable antisense ribozyme Sczakiel et al. (1995) according to the present invention.
[761] Triple helix method
[762] CanIon genomic DNA can also be used to inhibit the expression of the CanIon gene based on intracellular triple helix formation.
[763] Triple helix oligonucleotides are used to inhibit transcription from the genome. They are particularly useful for studying changes in cell activity with respect to specific genes.
[764] Similarly, portions of the CanIon genomic DNA can be used to study the effects of inhibiting CanIon transcription in cells. Traditionally, homopurine sequences have been thought to be best suited for triple naked strategy. However, homopyrimidine sequences can also inhibit gene expression. Such homopyrimidine oligonucleotides bind to major grooves in the homopurine: homopyrimidine sequence. Thus, both types of sequences derived from CanIon genomic DNA are included within the scope of the present invention.
[765] To implement gene therapy strategies using triple helices, the CanIon genomic DNA sequence can first be scanned and a 10-mer to 20-mer homopyrimidine or homopurine stretch that can be used in a triple-helix based strategy that inhibits CanIon expression. I sympathize. After identification of candidate homopyrimidine or homopurine stretch, their efficacy of inhibiting CanIon expression is assessed by introducing various amounts of oligonucleotides carrying candidate sequences into tissue culture cells expressing the CanIon gene.
[766] The oligonucleotides can be introduced into cells by a variety of methods known to those skilled in the art, such as calcium phosphate precipitation, DEAE-dextran, electroporation, liposome-mediated transfection, or natural uptake.
[767] Treated cells monitor altered cellular function or reduced CanIon expression using techniques such as Northern blotting, RNase protection assays, or PCR based strategies to monitor the transcription level of the CanIon gene in cells treated with oligonucleotides.
[768] Oligonucleotides that are effective at inhibiting gene expression in tissue culture cells can then be introduced in vivo utilizing the techniques described above at doses calculated based on in vitro results as revealed by antisense.
[769] In some embodiments, native (beta) anomers of oligonucleotide units can be substituted with alpha anomers to make oligonucleotides more resistant to nucleases. In addition, an insert, such as ethidium bromide, can be attached to the 3 'end of the alpha oligonucleotide to stabilize the triple helix. See Griffin et al. (1989) for information on the generation of oligonucleotides suitable for triple helix formation.
[770] Pharmaceutical Compositions and Formulations
[771] CanIon-Regulating Compounds
[772] The methods disclosed herein can be utilized to identify CanIon agonists or antagonists that selectively modulate CanIon activity in vitro and in vivo. Accordingly, the present invention includes methods of treating schizophrenia, bipolar disorder, or other diseases or conditions described above in a patient, the method comprising administering an effective amount of a CanIon-modulating compound. Suitably, the compound is a selective CanIon modulating compound. Compounds identified by the methods of the present invention include antibodies having binding specificities for human CanIon polypeptides. It is also expected that analogs of CanIon will be effective in modulating CanIon-mediated activity and associated physiological abnormalities associated with schizophrenia or bipolar disorder. In general, it is anticipated that the analytical methods of the present invention based on the role of CanIon in central nervous system disease may be utilized to identify compounds that may intervene in the analytical cascade of the present invention. In a suitable embodiment, the patient suffering from schizophrenia or bipolar disorder is treated by administering to the patient a pharmaceutical composition containing a therapeutically effective amount of a CanIon antagonist.
[773] symptom
[774] Since CanIon has been associated with schizophrenia and bipolar disorder, signs associated with CanIon may include central nervous system disease. Nervous system diseases are expected to have a complex genetic basis and share certain symptoms. In particular, as indicated herein, signs include schizophrenia and other mental disorders, neurodegenerative disorders, mood disorders, autism, drug dependence and alcoholism, epilepsy, pain disorders, mental retardation and cognitive disorders, mental disorders, anxiety disorders, eating disorders And other psychiatric disorders, including impulse control disorders, personality disorders [Diagnosis and Statistical Manual of Mental Disorders (DSM-IV)]. In addition, many cardiovascular diseases, including angina, hypertension, arrhythmia, can also be treated with CanIon modulators, preferably antagonists.
[775] Pharmaceutical Formulations and Routes of Administration
[776] Compounds identified using the methods of the present invention are mammals, including human patients, in the form of pharmaceutical compositions alone or in admixture with suitable carriers or excipients, in therapeutically effective amounts to treat or alleviate schizophrenia or bipolar disorder related diseases. Can be administered. By therapeutically effective amount is meant a compound content sufficient to result in alleviation of symptoms by the methods disclosed herein. Suitably, the therapeutically effective amount is suitable for sustained periodic dosing or administration. Techniques for the formulation and administration of compounds according to the invention can be found in the latest edition of "Remington's Pharmaceutical Sciences," (Mack Publishing Co. Easton, Pa.).
[777] Route of administration
[778] Suitable routes of administration include oral, rectal, transmucosal or intestinal administration, including subarachnoid, intraventricular, intravenous, intraperitoneal, intranasal or intraocular injections; Extra-intestinal delivery, including intramuscular, subcutaneous and intramedullary injections. Particularly suitable methods for administering drugs to treat central nervous system disease include the surgery of devices that deliver compounds for extended periods of time, such as subarachnoid delivery, including infusion into the spinal fluid via implanted pumps (Medtronic, Inc., Minneapolis, MN). Enemy transplants are included.
[779] Composition / Formulation
[780] Pharmaceutical compositions and drugs for use in the present invention may be made in a conventional manner using one or a plurality of physiologically acceptable carriers including excipients and adjuvants.
[781] For injection, the drugs of the invention may be prepared in the form of an aqueous solution, preferably a physiologically compatible buffer (eg Hank's solution, Ringer's solution) or a physiological saline buffer (phosphate or bicarbonate buffer). For transmucosal administration, penetrants suitable for the barrier to penetrate are used in the formulation. Such penetration penetrants are known in the art.
[782] Pharmaceutical formulations that can be taken orally include push-fit capsules made of gelatin, light sealed capsules made of gelatin and plasticizers such as glycerol or sorbitol. Push-fit capsules may contain an active ingredient mixed with a filler such as lactose, a binder such as starch and / or a lubricant such as talc or magnesium stearate, optionally a stabilizer. In soft capsules, the active ingredient may be dissolved or suspended in suitable liquids, such as fatty acids, liquid para or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration are provided in dosages suitable for such administration.
[783] For buccal administration, the compositions may take the form of tablets or lozenges made in a conventional manner.
[784] For inhalation administration, the composition according to the invention is delivered in the form of an aerosol spray from a pressurized pack or inhalation therapy using a suitable gas propellant, for example carbon dioxide. As an example for use in an inhaler or blower, capsules and cartridges of gelatin can be made to contain a powder mixture of the compound and a suitable powder base such as lactose or starch.
[785] The compounds may be made for extra-intestinal administration by injection, for example by cyclic injection or continuous infusion. Injectable formulations may be presented in unit dosage form, eg, in ampoules or in multi-use containers, with preservatives added. Such compositions may take the form of emulsions dissolved in suspensions, solutions or aqueous vesicles, and may contain formulated agents such as suspensions, stabilizers and / or dispersants.
[786] Pharmaceutical formulations for extra enteral administration include aqueous solutions of the active compounds in hydrophobic form. Aqueous suspensions may contain substances which increase the viscosity, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may contain suitable stabilizers or drugs that increase the solubility of the compounds that allow for the preparation of highly concentrated solutions.
[787] Alternatively, the active ingredient may be in lyophilized form or powder that is reconstituted with suitable vesicles, eg, pyrogen-free sterile water, prior to use.
[788] In addition to the formulations described above, the compounds may also be made into reservoir formulations. Such organ formulations may be administered by implantation (eg, subcutaneously or intramuscularly) or by intramuscular injection. Thus, such compounds can be mixed with polymerizable or hydrophobic materials (eg in emulsions dissolved in oil) or ion exchange resins, or made into nearly insoluble derivatives such as almost insoluble salts.
[789] The compounds can also be delivered using sustained-release systems, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic drug. Various sustained release materials are known in the art. Sustained-release capsules may release the compound for weeks to up to 100 days, depending on the chemical nature.
[790] Depending on the chemical properties and biological stability of the therapeutic drug, additional strategies for protein stabilization may be devised.
[791] The pharmaceutical composition may contain a suitable solid or gel carrier or excipient. Examples of such carriers or excipients include, but are not limited to, polymers such as calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, polyethylene glycols.
[792] Effective amount
[793] Pharmaceutical compositions suitable for use in the present invention include compositions containing an effective amount of the active ingredient in order to achieve the desired purpose. More specifically, a therapeutically effective amount means an amount effective to prevent or alleviate the occurrence of existing symptoms of the treated individual. Determination of the effective amount belongs to the ability of those skilled in the art.
[794] In the compounds used in the methods of the invention, the therapeutically effective amount can be estimated initially from cell culture assays, and a single dose can be administered to the animal model. This information can be used to determine the correct dose in humans.
[795] By therapeutically effective amount is meant the amount of the compound that results in relief of symptoms in the patient. Toxicity and therapeutic efficacy of these compounds can be determined by standard pharmaceutical procedures in cell culture or experimental animals, for example, LD50 (the amount that kills 50% of the test population) and ED50 (the amount that is effective for 50% of the population). . The fractional ratio between toxicity and therapeutic efficacy is the therapeutic index and can be expressed as the ratio between LD50 and ED50. Preferred are compounds that exhibit a high therapeutic index.
[796] Data from these cell culture assays and animal studies can be used to confirm the range of doses used in humans. Suitably, the dose of such compounds is within a range of circulating concentrations, including ED50, with little side effects. Dosages of such compounds may vary within this range depending upon the route of administration under the dosage form employed. The exact formulation, route of administration, and dose can be chosen by the individual physician in light of the patient's conditions (Fingl et al., 1975, in "The Pharmacological Basis of Therapeutics", Ch.1).
[797] Computer-Related Embodiments
[798] A "nucleic acid code of the invention" includes a) at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, of SEQ ID Nos. Continuous span of 500, 1000, 2000, 3000, 4000, 5000 or 10000 nucleotides; b) at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 500, 1000, 2000, 3000 of SEQ ID No 4 or its complement , Continuous span of 4000, 5000 or 10000 nucleotides; c) at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 500, 1000, 2000, 3000, of SEQ ID Nos. Contiguous span of 4000, 5000 or 10000 nucleotides, wherein the contiguous span comprises a biallelic marker selected from A1 to A17; d) a continuous span of at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300 or 400 nucleotides of SEQ ID No 6; e) a continuous span of at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300 or 400 nucleotides of SEQ ID No 6, wherein The continuous span comprises a biallelic marker A18; f) a nucleotide sequence complementary to one of the nucleotide sequences described above.
[799] “Nucleic Acid Codes of the Invention” also include a) at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, of SEQ ID Nos. Continuous span of 200, 500, 1000, 2000, 3000, 4000, 5000 or 10000 nucleotides; b) at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 500, 1000, 2000, 3000, 4000, of SEQ ID No 4 Consecutive spans of 5000 or 10000 nucleotides; c) a continuous span of at least 12, 15, 18, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300 or 400 nucleotides of SEQ ID No 6; f) nucleotide sequences complementary to nucleotide sequences complementary to one of the nucleotide sequences described above. Homologous sequences are meant sequences having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, 80% or 75% homology with these consecutive spans. Homology can be measured by any method disclosed herein, including BLASTN with default or modified parameters. Homologous sequences also include RNA sequences in which uridine replaces thymidine in the nucleic acid code of the invention. Nucleic acid code of the invention may be represented in the traditional single character format (Stryer, Lubert. Biochemistry, 3 rd edition), or any other form or code to record on the nucleotide sequence.
[800] "Polypeptide code of the invention" includes at least 6, 8, 10, 12, 15, 20, 25, 30, 40, 50, 100, 150, 200, 300, 400, 500, 700, 1000, Consecutive spans of 1200, 1400, 1600 or 1700 amino acids are included. In a suitable embodiment, the continuous span is amino acid positions 277, 338, 574, 678, 680, 683, 691, 692, 695, 696, 894, 1480, 1481, 1483, 1484, 1485, 1630, of SEQ ID No 5 At least 1, 2, 3, 5 or 10 of 1631, 1632, 1636, 1660, 1667, 1707, 1709. Polypeptide code of the present invention may be represented in the traditional single character format or three letter format (Stryer, Lubert. Biochemistry, 3 rd edition), or any other form or code to record on the polypeptide sequence.
[801] As will be appreciated by those skilled in the art, the nucleic acid codes and polypeptide codes of the present invention may be stored, recorded, and processed in any computer readable and accessible medium. As used herein, "recording" and "storage" means the process of storing information on a computer medium. One skilled in the art can readily adopt one of the known methods of recording information on a computer readable medium to yield a product comprising one or more nucleic acids or one or more polypeptides of the present invention. Another aspect of the invention is a computer readable medium having at least 2, 5, 10, 15, 20, 25, 30 or 50 nucleic acid codes of the invention recorded thereon. Another aspect of the invention is a computer readable medium having at least 2, 5, 10, 15, 20, 25, 30 or 50 polypeptide codes of the invention recorded thereon.
[802] Computer readable media includes magnetic readable media, optical readable media, electrically readable media, magnetic / optical media. For example, computer readable media are hard disks, floppy disks, magnetic tapes, CD-ROMs, digital versatile disks (DVD), Random Access Memory (RAM), Read Only Memory (ROM) and other types of media known to those skilled in the art. .
[803] Embodiments of the invention include systems for storing and processing sequence information as disclosed herein, in particular computer systems. One example of computer system 100 is illustrated in the block diagram of FIG. 2. "Computer system" as used herein means a hardware component, software component, data storage component used to analyze the nucleotide sequence of a nucleic acid code according to the invention or the amino acid sequence of a polypeptide code according to the invention. In one embodiment, computer system 100 is a Sun Enterprise 1000 server (Sun Microsystems, Palo Alto, Calif.). Suitably, computer system 100 maintains processes for processing, accessing, and manipulating sequence data. Process 105 can be any known central processing unit, such as Pentium III (Intel Corporation), or a similar process (Sun, Motorola, Compaq or International Business Machines).
[804] Suitably, computer system 100 comprises a general purpose system consisting of a process 105, one or more internal data storage components 110 for storing data, and one or more data retrieval devices for retrieving data stored in the data storage components. to be. One of the currently available computer systems is suitable.
[805] In certain embodiments, computer system 100 includes a process 105 connected to a bus coupled to main memory 115 (preferably RAM) and one or more internal data storage devices 110, such as hard drives and the like. And / or computer readable media for recording the data. In some embodiments, computer system 100 further includes one or more data retrieval devices 118 for reading data stored in data storage device 110.
[806] The data recovery device 118 may be, for example, a floppy disk, a compact disk drive, a magnetic tape drive, or the like. In some embodiments, internal data storage device 110 is a removable computer readable medium, such as a floppy disk, a compact disk, a magnetic tape, or the like, that holds the control logic and / or data written thereto. Computer system 100 may be programmed by suitable software that, when inserted into a data recovery device, reads control logic and / or data from data storage components.
[807] Computer system 100 includes an output device 120 that is used to display results to a computer user. Computer system 100 may connect with other computer systems 125a-c in a network or wide area network to provide centralized access to computer system 100.
[808] Software that accesses and processes the nucleotide sequence of the nucleic acid code according to the present invention or the amino acid sequence of the polypeptide code according to the present invention (eg, a search tool, a comparison tool, a modeling tool, etc.) may be present in main memory 115 during execution. have.
[809] In some embodiments, computer system 100 may further comprise a sequence comparator that compares a reference nucleotide or polypeptide sequence stored on a computer readable medium with the aforementioned nucleic acid code or polypeptide code of the invention stored on a computer readable medium. have. A “sequence comparator” is a computer system 100 that compares a nucleotide or polypeptide sequence with other nucleotide or polypeptide sequences and / or compounds, including but not limited to peptides, peptidomimetics, chemicals stored in data storage means. ) Means one or more programs to run. For example, a sequence comparator compares the nucleotide sequence of a nucleic acid code according to the present invention stored in a computer readable medium or the amino acid sequence of the polypeptide code according to the present invention with a reference sequence stored in a computer readable medium to participate in homology, biological function. Motif, or structural motifs can be identified. Various sequence comparison programs of the present invention are specifically related to the present invention.
[810] FIG. 3 is a flow diagram illustrating an embodiment of a process 200 for comparing sequences of new nucleotides or proteins with sequences on a database to determine levels of homology between new nucleotides and sequences on a database. The database of sequences may be a personal database stored in computer system 100 or a public database such as GENBANK, PIR or SWISSPROT available via the Internet.
[811] Process 200 begins at start step 201 and proceeds to step 202, where the new sequences being compared are stored in the storage of computer system 100. As mentioned above, the storage device can be any type of storage device including RAM or internal storage.
[812] Process 200 then proceeds to step 204, where the first sequence stored in the database is read into the storage of the computer. Next, a comparison is made at step 210 to confirm that the first and second sequences are identical. Methods of comparing two nucleotide or protein sequences even if they are not identical are known to those skilled in the art. For example, blanks may be introduced in one sequence to increase the level of homology between the two test sequences. Parameters that control whether blanks or other features are introduced into the sequence during the comparison are typically entered by the user of the computer system.
[813] If a comparison of the two sequences is performed at step 210, a determination is made at decision 210 whether the two sequences are identical. Of course, the meaning of "identical" is not limited to absolutely identical sequences. The sequences within the homology parameters entered by the user are marked as “identical” in process 200.
[814] If a determination is made that the two sequences are the same, process 200 proceeds to step 214, where the name of the sequence is displayed to the user from the database. This step informs the user that the sequence with the displayed name meets the entered homology limit. Once the name of the stored sequence is displayed to the user, process 200 proceeds to decision step 218 where a determination is made as to whether more sequences are present in the database. If no more sequences are present in the database, process 200 terminates at final step 220. However, if more sequences are present in the database, process 200 proceeds to step 224 where the pointer moves to the next sequence in the database and compares it with the sequence. In this way, the new sequence is aligned and compared with all sequences in the database.
[815] If a decision is made at 212 that the sequences are not homologous, then process 200 proceeds immediately to decision 218 to ascertain if any other sequences are available in the comparison database.
[816] Thus, one aspect of the present invention relates to a process, a data storage device having stored therein a nucleic acid code or polypeptide code, a data storage device having a recoverable reference nucleotide sequence or polypeptide sequence compared with a nucleic acid code or polypeptide code of the invention, A computer system comprising a sequence comparator for performing a comparison. Sequence comparators indicate the level of homology between sequences compared, identify motifs involved in biological function and structural motifs in nucleic acid codes and polypeptides of the invention, or identify structural motifs in sequences compared with these nucleic acid codes and polypeptide codes. I can sympathize. In some embodiments, the data storage device may store at least 2, 5, 10, 15, 20, 25, 30 or 50 sequences of the nucleic acid code or polypeptide code of the invention.
[817] Another aspect of the present invention is a method of determining the level of homology between a nucleic acid code of the present invention and a reference nucleotide sequence, wherein the method reads the nucleic acid code and the reference nucleotide sequence through the use of a computer program for determining the level of homology. A computer program consists of measuring homology between the nucleic acid code and the reference nucleotide sequence. The computer program can be one of a number of computer programs that measure homology levels, including programs specified herein, such as BLAST2N with default or modified parameters. The method can be implemented using the computer system described above. In addition, the method reads at least two, five, ten, fifteen, twenty, twenty, thirty, or fifty of the aforementioned nucleic acid codes of the present invention through the use of a computer program to determine homology between the nucleic acid code and the reference nucleotide sequence Can be run.
[818] 4 is a flow diagram illustrating an embodiment of a process 250 for identifying whether two sequences are homologous. Process 250 begins at start step 252 and proceeds to step 254 where the first sequence being compared is stored in memory. The second sequence to be compared is then stored in memory at step 256. Process 250 then proceeds to step 260 where the first letter in the first sequence is read and then proceeds to step 262 where the first letter in the second sequence is read. If the sequence is a nucleotide sequence, the letters are A, T, C, G (or U). If the sequence is a protein sequence, it is a single letter amino acid code, so that the first and second sequences can be easily compared.
[819] In decision step 264 a determination is made as to whether two characters are identical. If they are the same, process 250 proceeds to step 268, where the next letter is read in the first and second sequences. A determination is made as to whether the next character is the same. If they are the same, process 250 continues this loop until the two characters are not identical. If a determination is made that the next two letters are not the same, process 250 proceeds to decision step 274 and checks whether there are more letters or sequences to read.
[820] If there are no more characters to read, process 250 proceeds to step 276, where the level of homology between the first sequence and the second sequence is displayed to the user. Homology levels are determined by calculating the ratio of letters between identical sequences of the entire sequence in the first sequence. Thus, if all letters in the first 100 nucleotide sequences are aligned with all letters in the second sequence, the homology level is 100%.
[821] Alternatively, the computer program may be a computer program that compares the nucleotide sequence of the nucleic acid code according to the present invention with the reference nucleotide sequence to identify whether the nucleic acid code of the present invention is distinguished from the reference nucleic acid sequence at one or more positions. Optionally, such a program records the length and identity of the deleted or substituted nucleotides relative to the sequence of the reference polynucleotide or to the nucleic acid code of the invention. In one embodiment, the computer program may be a program that identifies whether the nucleotide sequence of the nucleic acid code according to the present invention possesses one or more single nucleotide polymorphisms (SNPs) as compared to the reference nucleotide sequence. These single nucleotide polymorphisms may each comprise a single base substitution, insertion or deletion.
[822] Another aspect of the invention is a method of determining the level of homology between a polypeptide code of the present invention and a reference polypeptide sequence, which method utilizes a computer program to measure the level of homology to determine the polypeptide code of the present invention and the reference polypeptide sequence. Reading and measuring homology between the polypeptide code and the reference polypeptide sequence with the computer program.
[823] Accordingly, another aspect of the present invention is a method of identifying whether a nucleic acid code of the present invention is distinguished from a reference nucleotide sequence in one or more nucleotides, which method is characterized by the use of a computer program to identify differences between nucleic acid sequences. Reading the reference nucleotide sequence and identifying the difference between the nucleic acid code and the reference nucleotide sequence with the computer program. In some embodiments, the computer program is a program for identifying single nucleotide polymorphisms. The method can be implemented by the computer system described above and the method illustrated in FIG. The method also reads at least two, five, ten, fifteen, twenty, twenty, thirty or fifty, and reference nucleotide sequences of the foregoing nucleic acid codes of the present invention through the use of a computer program and the Differences between reference nucleotide sequences can be identified and performed.
[824] In another embodiment, the computer based system further comprises an identifier that identifies a feature in the nucleotide sequence of the nucleic acid code according to the invention or the amino acid sequence of the polypeptide code according to the invention.
[825] "Identifier" means one or more programs that identify topologies within the nucleotide sequence of the nucleic acid code according to the present invention or the amino acid sequence of the polypeptide code according to the present invention. In one embodiment, the identifier may comprise a program for identifying an open reading frame in the cDNA code of the present invention.
[826] 5 is a flow diagram illustrating an embodiment of an identifier process 300 for detecting the presence of a feature in a sequence. Process 300 begins at start step 300 and proceeds to step 304, where the first sequence whose geometry has been examined is stored in storage 115 in computer system 100. Process 300 then proceeds to step 306, where the database of sequence topography is opened. This database contains a list of names and attributes for each terrain. For example, the terrain name may be "Initiation Codon" and the attribute may be "ATG". In another example, the terrain name is "TAATAA Box" and the attribute is "TAATAA". An example of such a database is created by the University of Winsconsin Genetics Computer Group (www.gcg.com).
[827] If the database of terrain is opened in step 306, process 300 proceeds to step 308, where the first terrain is read from the database. Thereafter, the comparison of the properties of the first terrain with the first sequence proceeds to step 310. Then, in decision step 316, a determination is made as to whether the feature of the terrain is found in the first sequence. If the attribute is found, process 300 proceeds to step 318 where the name of the terrain is displayed to the user.
[828] Process 300 then proceeds to decision step 320 where a determination is made as to whether a move feature exists in the database. If there is no more terrain then process 300 ends at final step 324. However, if there are more terrain in the database, process 300 reads the next sequence terrain in step 236 and returns to step 310, where the attributes of the next terrain are compared with the first sequence.
[829] If the terrain attribute is not found in the first sequence in decision step 316, process 300 proceeds directly to decision step 320 and checks whether more terrain exists in the database.
[830] In another embodiment, the identifier may comprise a molecular modeling program for measuring the three dimensional structure of the polypeptide code according to the invention. In some embodiments, the molecular modeling program identifies target sequences that best match a profile representing the structural environment of the residues in known three-dimensional protein structures (US Pat. No. 5,436,850). In another technique, the known three-dimensional structure of a protein in a given family overlaps, thus defining a structurally conserved region in that family. This protein modeling technique also employs known three-dimensional structures of homologous proteins that approximate the structure of the polypeptide code according to the present invention (US Pat. No. 5,557,535). Traditional homology modeling techniques are commonly used to establish models of proteases and antibodies (Sowdhamini et al., (1997)). In addition, when the protein of interest has poor sequence homology with the template protein, a three-dimensional protein model can be developed in a comparative manner. In some cases, proteins fold into similar three-dimensional structures despite very weak sequence homology. For example, the three-dimensional structure of many helix cytokines folds into a similar three-dimensional topology despite weak sequence homology.
[831] Recent developments in threading methods have enabled the identification of similar folding patterns in many cases where no structural relatedness between target and template is detected at the sequence level. In the hybrid method of fold recognition with multiple sequence threading (MST), structural equivalents are inferred from the threading output using the distance geometry program DRAGON, which constitutes a low analytical model. It is constructed using a molecular modeling package, for example QUANTA.
[832] According to the three-step approach, candidate templates are first identified with the new fold recognition program MST, which can perform simultaneous threading of multiple aligned sequences in one or more 3-D structures. In the second step, the structural equivalent obtained from the MST output is converted into an interresidue distance restraint and put into the distance geometry program DRAGON along with additional information obtained from the secondary structure prediction. The program incorporates the constraints in an equitable manner and yields a number of low analytical model validations. In the third step, these low analytical model validations are converted to full-atomic models and energy minimized using the molecular modeling package QUANTA (Aszodi et al., (1997)).
[833] The results of the molecular modeling assays can then be utilized in rational drug design techniques to identify agents that modulate the activity of the polypeptide code according to the invention.
[834] Accordingly, another aspect of the invention is a method of identifying a feature within a nucleic acid code or polypeptide code of the invention, wherein the method reads the nucleic acid code or polypeptide code through the use of a computer program that identifies the feature and A computer program consists of identifying topography in nucleic acid code and polypeptide code. In one embodiment, the computer program is a computer program that identifies an open reading frame. In other embodiments, the computer program identifies structural motifs in the polypeptide sequence. In another embodiment, the computer program is a molecular modeling program. The method reads a single sequence or at least two, five, ten, fifteen, twenty, twenty, thirty, fifty, or 50 of the above-described nucleic acid code or polypeptide code of the present invention through the use of a computer program and the nucleic acid code into And topography can be identified and executed within the polypeptide code.
[835] The nucleic acid code or polypeptide code of the present invention can be stored and manipulated in various formats in various data processor programs. For example, they can be stored as document files in a word processor, for example Microsoft Word or WordPerfect, or as ASCII files in various programs familiar to those skilled in the art, for example DB2, SYBASE or ORACLE. In addition, many computer programs and databases can be used as sequence comparators, identifiers, or sources of reference nucleotide or polypeptide sequences compared to nucleic acid codes or polypeptide codes according to the present invention. The following list provides guidance to the programs and databases useful for nucleic acid codes or polypeptide codes according to the present invention, but does not limit the present invention: MacPattern (EMBL), DiscoveryBase (Molecular Applications Group), GeneMine (Molecular Applications Group), Look (Molecular Applications Group), MacLook (Molecular Applications Group), BLAST and BLAST2 (NCBI), BLASTN and BLASTX (Altschul et al, 1990), FASTA (Pearson and Lipman, 1988), FASTDB (Brutlag et al., 1990) , Catalyst (Molecular Simulations Inc.), Catalyst / SHAPE (Molecular Simulations Inc.), Cerius 2 .DBAccess (Molecular Simulations Inc.), HypoGen (Molecular Simulations Inc.), Insight II (Molecular Simulations Inc.), Discover (Molecular Simulations Inc.), CHARMm (Molecular Simulations Inc.), Felix (Molecular Simulations Inc.), DelPhi (Molecular Simulations Inc.), QuanteMM (Molecular Simulations Inc.), Homology (Molecular Simulations Inc.), Modeler (Molecular Simulations Inc. ), ISIS (Molecular S imulations Inc.), Quanta / Protein Design (Molecular Simulations Inc.), WebLab (Molecular Simulations Inc.), WebLab Diversity Explorer (Molecular Simulations Inc.), Gene Explorer (Molecular Simulations Inc.), SeqFold (Molecular Simulations Inc.) , the EMBL / Swissprotein database, MDL Available Chemicals Directory database, MDL Drug Data Report database, Comprehensive Medicinal Chemistry database, Derwents's World Drug Index database, BioByteMasterFile database, Genbank database, Genseqn database. In view of the disclosure herein, many other programs and databases are apparent to those skilled in the art.
[836] The program detectable motifs include leucine zippers, helix-rotating-helix motifs, glycosylation sites, ubiquitination sites, alpha helices, sequences encoding beta sheets; Signal sequences encoding signal peptides directing secretion of the encoded protein; Sequences involved in transcriptional regulation such as homeobox, acidic stretch, enzyme active site, substrate binding site, enzyme cleavage site, are included.
[837] Herein, various publications, documents, and published patent applications are mentioned. These publications, patents, and published patent applications are hereby incorporated by reference in their entirety.
[838] Example 1 Identification of Double Allele Markers-DNA Extraction
[839] The donor was irrelevant and healthy. They provided enough diversity to represent heterogeneous populations in France. DNA was extracted from 100 individuals and tested for detection of the double allele marker.
[840] 30 ml peripheral venous blood is drawn from each donor in the presence of EDTA. Cells (pellets) are harvested after centrifugation at 2000 rpm for 10 minutes. Erythrocyte cells are lysed with lysis solution (50 mL final volume: 10 mM Tris pH 7.6; 5 mM MgCl 2 ; 10 mM NaCl). After resuspension of the pellet in the lysis solution, the solution is centrifuged (10 min, 2000 rpm) for the number of times necessary to remove residual red blood cells present in the supernatant.
[841] Pellets of leukocytes were lysed overnight at 42 ° C. in a 3.7 ml lysis solution consisting of:
[842] 3 ml TE 10-2 (Tris-HCl 10 mM, EDTA 2 mM) / NaCl 0.4 M
[843] 200 μl SDS 10%
[844] 500 μl K-proteinase (2 mg K-proteinase / TE 10-2 / NaCl 0.4 M)
[845] For protein extraction, 1 ml saturated NaCl (6M) (1 / 3.5 v / v) is added. After vigorous stirring, the solution is centrifuged at 10000 rpm for 20 minutes.
[846] For precipitation of DNA, 2 to 100% ethanol is added to the supernatant and the solution is centrifuged at 2000 rpm for 30 minutes. The DNA solution is washed three times with 70% ethanol to remove salts and centrifuged for 20 minutes at 2000 rpm. The pellet is dried at 37 ° C. and resuspended in 1 ml TE 10-1 or 1 ml water. DNA concentration is assessed by measuring OD at 260 nm (1 unit OD = 50 μg / ml DNA).
[847] To confirm the presence of the protein in the DNA solution, the OD 260 / OD 280 ratio is measured. Only DNA formulations with an OD 260 / OD 280 ratio of 1.8 to 2 are used in the examples below.
[848] A pool consists of a mixture of equal amounts of DNA from each individual.
[849] Example 2 Identification of Double Allele Markers: Amplification of Genomic DNA by PCR
[850] Amplification of specific genomic sequences in the DNA samples of Example 1 was performed in the DNA pool obtained previously. In addition, 50 individual samples were similarly amplified.
[851] PCR analysis is performed using the following protocol:
[852] 25 μl final volume
[853] DNA 2 ng / μl
[854] MgCl2 2 mM
[855] dNTP (each) 200 μM
[856] Primer (each) 2.9 ng / μl
[857] Ampli Taq Gold DNA Polymerase 0.05 unit / μl
[858] PCR buffer (10x = 0.1 M TrisHCl pH 8.3 0.5 M KCl) 1x
[859] Each pair of first primers was designed using the sequence information of the CanIon gene disclosed herein and the OSP software (Hilier & Green, 1991). The first pair of primers are approximately 20 nucleotides in length and have the sequences disclosed in the columns classified as PU and RP in Table 1.
[860] Table 1:
[861]
[862] Suitably the primers are used for sequencing and have a common oligonucleotide tail upstream of the particular base targeted for amplification.
[863] Primer PU has the following additional PU 5 'sequence: TGTAAAACGACGGCCAGT; Primer RP has the following RP 5 'sequence: CAGGAAACAGCTATGACC. Primers carrying additional PU 5 'sequences are described in SEQ ID No 7. Primers carrying additional RP 5 'sequences are described in SEQ ID No 8.
[864] The synthesis of these primers is carried out according to the phosphoramidite method in the following GENSET UFPS 24.1 sequencer.
[865] DNA amplification is carried out in a Genius II sequence amplifier. After heating at 95 ° C. for 10 minutes, 40 runs are performed. Each cycle consists of: 30 seconds at 95 ° C., 1 minute at 54 ° C., 30 seconds at 72 ° C. For final extension, amplification is terminated at 72 ° C. for 10 minutes. The content of amplification products obtained is measured on 96-well microtiter plates using a fluorometer and an intercalant agent Picogreen (Molecular Probes).
[866] Example 3 Identification of Dual Allele Markers—Sequencing and Polymorphism of Amplified Genomic DNA
[867] Sequencing of the DNA amplified in Example 2 is carried out in an ABI sequencer. Sequences of amplification products are determined by automated dideoxy terminator sequencing using a dye terminator cycle sequencing protocol. The product of this sequencing reaction is transferred on a sequencing gel, and the sequence is confirmed by gel image analysis (ABI Prism DNA sequencing software (2.1.2)).
[868] Sequence data is further assessed to detect the presence of a dual allele marker in the amplified fragment. Polymorphic search is based on the presence of overlapping peaks in the electrophoretic pattern due to different bases occurring at the same location as described above.
[869] In 17 amplified fragments, 18 double allele markers were detected. The locations of these dual allele markers are shown in Table 2.
[870] Table 2:
[871]
[872] BM means "double allele marker". all1 and all2 mean allele 1 and allele 2 of the double allele marker, respectively.
[873] Table 3:
[874]
[875] Example 4 Verification of Polymorphism by Microsequence Analysis
[876] The double allele markers identified in Example 3 are further identified and the frequency of each of them is determined by microsequencing. Microsequencing is performed on individual DNA samples identified in Example 1.
[877] Amplification from the genomic DNA of the individual is carried out by PCR as described above in the detection of the double allele marker using the same set of PCR primers (Table 1).
[878] Primers suitable for microsequencing are approximately 19 nucleotides in length and hybridize directly upstream of the polymorphic bases considered. Primers used for microsequencing according to the present invention are described in detail in Table 4.
[879] Table 4:
[880]
[881] Mis1 and Mis2 refer to microsequencing primers that hybridize with the non-coding strand of the CanIon gene or the coding strand of the CanIon gene, respectively.
[882] The microbase sequence reaction is carried out as follows:
[883] After purification of the amplification product, the microsequencing reaction mixture was prepared according to the manufacturer's recommendations, 10 pmol sequencing oligonucleotide, 1U Thermosequenase (Amersham E79000G), 1.25 μl Thermosequenase buffer (260 mM Tris HCl pH 9.5, 65 mM MgCl 2 ) And two suitable fluorescence ddNTPs (Perkin Elmer, Dye Terminator Set 401095) complementary to nucleotides at the polymorphic position of each biele marker used are added in a 20 μl final volume. After 4 minutes at 94 ° C., 20 PCR cycles of 15 seconds at 55 ° C., 5 seconds at 72 ° C., and 10 seconds at 94 ° C. were performed on a Tetrad PTC-225 sequence amplifier (MJ Research). The unintegrated dye terminator is then removed by ethanol precipitation. Samples are finally resuspended in formamide-EDTA dropping buffer, heated at 95 ° C. for 2 minutes and then loaded onto polyacrylamide sequencing gel. Data is collected with ABI PRISM 377 DNA Sequencer and processed with CanIonScan software (Perkin Elmer).
[884] After gel analysis, data is automatically processed with software that allows for allele identification of the double allele marker present in each amplified fragment.
[885] The software evaluates factors such as whether the signal strength resulting from the microsequencing process is weak, normal or full, or whether such a signal is unclear. In addition, the software identifies significant peaks (according to shape and height criteria). Among the prominent peaks, the peaks corresponding to the target sites are identified based on location. If two prominent peaks are detected at the same location, each sample is classified into homogeneous or heterogeneous type based on height ratio.
[886] Example 5
[887] Preparation of Antibody Compositions Against CanIon Proteins
[888] Substantially pure proteins or peptides are isolated from transfected or transformed cells comprising an expression vector encoding a CanIon protein or portion thereof. The concentration of protein in the final formulation is adjusted to levels of several mg / ml, for example by concentration in an Amicon filter device. Then, monoclonal or polyclonal antibodies to the protein can be made as follows.
[889] A. Monoclonal Antibody Production by Hybridoma Fusion
[890] Monoclonal antibodies against the CanIon protein or portions thereof can be made from murine hybridomas according to the typical methods of Kohler, G. and Milstein, C., (1975) or modifications thereof (Harlow, E. and D. Lane. 1988).
[891] In brief, mice are repeatedly inoculated with several mg of CanIon protein or portions thereof for several weeks. The mice are then killed and the antibody producing cells of the spleen are isolated. Splenocytes are fused with myeloma cells with polyethylene glycol, and excess unfused cells are grown and destroyed in selection medium (HAT medium) containing aminopterin. Successfully fused cells are diluted and a portion of the dilution is plated into the wells of the microtiter plate, where the growth of the culture is continued. Antibody-producing clones are identified by detection of antibodies in the supernatant of the wells by immunoassay procedures, such as ELISA (Engvall, (1980)) and variants thereof. Selected positive clones are amplified and their monoclonal antibody products are harvested. Detailed procedures for monoclonal antibody production are described in Davis, L. et al. Basic Methods in Molecular Biology Elsevier, New York. It is described in Section 21-2.
[892] B. Polyclonal Antibody Production by Immunization
[893] Polyclonal antisera that contain an antibody to a heterologous epitope in the CanIon protein or portion thereof can be made by immunizing a suitable human exclusion animal with the CanIon protein or portion thereof, which may or may not be altered to enhance immunogenicity. have. Suitably, the non-human animal is a non-human mammal selected from mice, rats, rabbits, goats or horses. Alternatively, a crude CanIon crude formulation may be used to make the antibody. Such proteins, fragments or formulations are introduced into non-human mammals in the presence of suitable adjuvants (eg, aluminum hydroxide, RIBI, etc.) known in the art. In addition, such proteins, fragments or formulations may be pretreated with agents that increase antigenicity, which are known to those of skill in the art, including methylated bovine serum albumin (mBSA), bovine serum albumin (BSA), Hepatitis B surface antigen, keyhole limpet hemocyanin (KLH), and the like. Serum from immunized animals is collected, processed and tested according to known procedures. If such serum contains polyclonal antibodies against unwanted epitopes, the polyclonal antibodies can be purified by immunoaffinity chromatography.
[894] Effective polyclonal antibody production is affected by a number of factors related to both antigen and host type. In addition, host animals exhibit different responses to the site and dose of inoculation, with insufficient or excessive doses of antigen resulting in low titer antisera. Small amounts (ng levels) of antigen administered to many intradermal sites are considered to be the safest. Techniques for producing and processing polyclonal antisera are known in the art (Mayer and Walker (1987)). Effective immunization protocols for rabbits can be found in Vaitukaitis, J. et al. (1971).
[895] Efficacy booster injections are given at regular intervals and antiserum is collected when antibody titers begin to decrease when measured semi-quantitatively, for example, by double immunodiffusion in agar against known concentrations of antigen (Ouchterlony, O. et. al. (1973)). The plateau phase concentration of the antibody is generally from 0.1 to 0.2 mg (approximately 12 μΜ) per ml serum. The affinity of antiserum for antigen is confirmed by creating a competitive binding curve (Fisher, D., (1980)).
[896] Antibody formulations made according to monoclonal or polyclonal protocols are useful for quantitative immunoassays to measure the concentration of antigen-bearing material in biological samples; They are also used to qualitatively or quantitatively confirm the presence of antigens in biological samples. These antibodies can also be used in therapeutic compositions that kill cells that express the protein or reduce the level of the protein in the body.
[897] While suitable embodiments of the invention have been illustrated, various modifications may be made by those skilled in the art without departing from the spirit and scope of the invention.
[898] references
[899] Abbondanzo SJ et al., 1993, Methods in Enzymology, Academic Press, New York, pp 803-823
[900] Ajioka RS et al., Am. J. Hum. genet ., 60: 1439-1447, 1997
[901] Altschul et al., 1990, J. Mol. Biol. 215 (3): 403-410
[902] Altschul et al., 1993, Nature genetics 3: 266-272
[903] Altschul et al., 1997, Nuc. Acids Res. 25: 3389-3402
[904] Ames, R.S. et al. J. Immunol. Methods 184: 177-186 (1995)
[905] anderson et al., Symp. Soc. Exp. Biol. 48: 85-97 (1994)
[906] Anton M. et al., 1995, J. Virol., 69: 4600-4606
[907] Araki K et al. (1995) Proc. Natl. Acad. Sci. USA . 92 (1): 160-4.
[908] Ashkenazi, A. et al. PNAS 88: 10535-10539 (1991)
[909] Asz di et al., Proteins: Structure, Function, and genetics, Supplement 1: 38-42 (1997)
[910] Ausubel et al. (1989) Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N.Y.
[911] Bartunek, P. et al. Cytokine 8 (1): 14-20 (1996)
[912] Baubonis W. (1993) Nucleic Acids Res . 21 (9): 2025-9.
[913] Beaucage et al., Tetrahedron Lett 1981, 22: 1859-1862
[914] Better, M. et al. Science 240: 1041-1043 (1988)
[915] Blouin JL et al. Nature genetics, 20: 70-73 (1998)
[916] Bradley A., 1987, Production and analysis of chimaeric mice. In : EJ Robertson (Ed.), Teratocarcinomas and embryonic stem cells: A practical approach. IRL Press, Oxford, pp. 113.
[917] Bram RJ et al., 1993, Mol. Cell Biol., 13: 4760-4769
[918] Brinkman U. et al. J. Immunol. Methods 182: 41-50 (1995)
[919] Brown EL, Belagaje R, Ryan MJ, Khorana HG, Methods Enzymol 1979; 68: 109-151
[920] Brutlag et al. Comp. App. Biosci. 6: 237-245, 1990
[921] Brzustowicz et al., Am. J. Hum. genet. 65: 1096-1103 (1999)
[922] Bulman et al., Hum. Mol. Gen. 6 (10) 1679-1685 (1997)
[923] Burton, D. R. et al. Advances in Immunology 57: 191-280 (1994)
[924] Bush et al., 1997, J. Chromatogr., 777: 311-328.
[925] Carlson, N.G. et al. J. Biol. Chem. 272 (17): 11295-11301 (1997)
[926] Chai H. et al. (1993) Biotechnol. Appl. Biochem . 18: 259-273.
[927] Chee et al. (1996) Science. 274: 610-614.
[928] Chen and Kwok Nucleic Acids Research 25: 347-353 1997
[929] Chen et al. (1987) Mol. Cell. Biol. 7: 2745-2752.
[930] Chen et al. Proc. Natl. Acad. Sci. USA 94/20 10756-10761,1997
[931] Chen, Z. et al. Cancer Res. 58 (16): 3668-3678 (1998)
[932] Cho RJ et al., 1998, Proc. Natl. Acad. Sci. USA, 95 (7): 3752-3757.
[933] Chou J.Y., 1989, Mol. Endocrinol., 3: 1511-1514.
[934] Clark AG (1990) Mol. Biol. Evol. 7: 111-122.
[935] Coles R, Caswell R, Rubinsztein DC, Hum Mol genet 1998; 7: 791-800
[936] Compton J. (1991) Nature . 350 (6313): 91-92.
[937] Davis L.G., M.D. Dibner, and J.F. Battey, Basic Methods in Molecular Biology, ed., Elsevier Press, NY, 1986
[938] Dempster et al., (1977) JR Stat. Soc., 39B: 1-38.
[939] Deng, B. et al. Blood 92 (6): 1981-1988 (1998)
[940] Dent DS & Latchman DS (1993) The DNA mobility shift assay. In: Transcription Factors: A Practical Approach (Latchman DS, ed.) Pp1-26. Oxford: IRL Press
[941] Denyer et al., Drug Disc. Today 3 (7): 323-332 (1998
[942] Eckner R. et al. (1991) EMBO J. 10: 3513-3522.
[943] Edwards et Leatherbarrow, Analytical Biochemistry , 246, 1-6 (1997)
[944] Engvall, E., Meth. Enzymol. 70: 419 (1980)
[945] Excoffier L. and Slatkin M. (1995) Mol. Biol. Evol., 12 (5): 921-927.
[946] Feldman and Steg, 1996, Medecine / Sciences, synthese, 12: 47-55
[947] Felici F., 1991, J. Mol. Biol., Vol. 222: 301-310
[948] Fell, H.P. et al. J. Immunol. 146: 2446-2452 (1991)
[949] Fields and Song, 1989, Nature, 340: 245-246
[950] Fisher, D., Chap. 42 in: Manual of Clinical Immunology, 2d Ed. (Rose and Friedman, Eds.) Amer. Soc. For Microbiol., Washington, D.C. (1980)
[951] Flotte et al. (1992) Am. J. Respir. Cell Mol. Biol . 7: 349-356.
[952] Fodor et al. (1991) Science 251: 767-777.
[953] Fraley et al. (1979) Proc. Natl. Acad. Sci. USA. 76: 3348-3352.
[954] Fried M, Crothers DM, Nucleic Acids Res 1981; 9: 6505-6525
[955] Fromont-Racine M. et al., 1997, Nature genetics, 16 (3): 277-282.
[956] Fuller SA et al. (1996) Immunology in Current Protocols in Molecular Biology , Ausubel et al. Eds, John Wiley & Sons, Inc., USA.
[957] Furth PA et al. (1994) Proc. Natl. Acad. Sci USA. 91: 9302-9306.
[958] Garner MM, Revzin A, Nucleic Acids Res 1981; 9: 3047-3060
[959] Geysen H. Mario et al. 1984. Proc. Natl. Acad. Sci. U.S.A. 81: 3998-4002
[960] Ghosh and Bacchawat, 1991, Targeting of liposomes to hepatocytes , IN: Liver Diseases, Targeted diagnosis and therapy using specific receptors and ligands . Wu et al. Eds., Marcel Dekeker, New York, pp. 87-104.
[961] Gillies, S.D. et al. J. Immunol. Methods 125: 191-202 (1989)
[962] Gillies, S.O. et al. PNAS 89: 1428-1432 (1992)
[963] Gonnet et al., 1992, Science 256: 1443-1445
[964] Gonzalez et al., Drug Disc. Today 4 (9): 431: 439 (1999)
[965] Gopal (1985) Mol. Cell. Biol. , 5: 1188-1190.
[966] Gossen M. et al. (1992) Proc. Natl. Acad. Sci. USA. 89: 5547-5551.
[967] Gossen M. et al. (1995) Science . 268: 1766-1769.
[968] Graham et al. (1973) Virology 52: 456-457.
[969] Green et al., Ann. Rev. Biochem . 55: 569-597 (1986)
[970] Greenspan and Bona, FASEB J. 7 (5): 437-444 (1989)
[971] Griffin et al. Science 245: 967-971 (1989)
[972] Grompe, M. (1993) Nature genetics. 5: 111-117.
[973] Grompe, M. et al. (1989) Proc. Natl. Acad. Sci. USA 86: 5855-5892.
[974] Gu H. et al. (1993) Cell 73: 1155-1164.
[975] Gu H. et al. (1994) Science 265: 103-106.
[976] Guatelli JC et al. Proc. Natl. Acad. Sci. USA. 35: 273-286.
[977] Hacia JG, Brody LC, Chee MS, Fodor SP, Collins FS, Nat genet 1996; 14 (4): 441-447
[978] Hall LA and Smirnov IP (1997) Genome Research , 7: 378-388.
[979] Hames BD and Higgins SJ (1985 ) Nucleic Acid hybridization: A Practical Approach . Hames and Hamill, Pfugers Arch. 391, 85-100 (1981).
[980] Hammerling, et al., In: MONOCLONAL ANTIBODIES and TCELL HYBRIDOMAS 563681 (Elsevier, N.Y., 1981)
[981] Harju L, Weber T, Alexandrova L, Lukin M, Ranki M, Jalanko A, Clin Chem 1993; 39 (11 Pt 1): 2282-2287
[982] Harland et al. (1985) J. Cell. Biol. 101: 1094-1095.
[983] Harlow, E., and D. Lane. 1988. Antibodies A Laboratory Manual. Cold Spring Harbor Laboratory. pp. 53-242
[984] Harper JW et al., 1993, Cell, 75: 805-816
[985] Harrop, J.A. et al. J. Immunol. 161 (4): 1786-1794 (1998)
[986] Hawley ME et al. (1994) Am. J. Phys. Anthropol. 18: 104.
[987] Hell et al., Annals N.Y. Acad. Sci. 747: 282-293 (1994)
[988] Henikoff and Henikoff, 1993, Proteins 17: 49-61
[989] Higgins et al., 1996, Methods Enzymol. 266: 383-402
[990] Higgins Ed., IRL Press, Oxford.
[991] Hillier L. and Green P. Methods Appl., 1991, 1: 124-8.
[992] Hoess et al. (1986) Nucleic Acids Res . 14: 2287-2300.
[993] Huang L. et al. (1996) Cancer Res 56 (5): 1137-1141.
[994] Huston et al. Methods in Enzymology 203: 46-88 (1991)
[995] Huygen et al. (1996) Nature Medicine. 2 (8): 893-898.
[996] Izant JG, Weintraub H, Cell 1984 Apr; 36 (4): 1007-15
[997] Julan et al. (1992) J. Gen. Virol . 73: 3251-3255.
[998] Kanegae Y. et al., Nucl. Acids Res. 23: 3816-3821 (1995).
[999] Karlin and Altschul, 1990, Proc. Natl. Acad. Sci. USA 87: 2267-2268
[1000] Kettleborough, C. A. et al. Eur. J. Immunol. 24: 952-958 (1994)
[1001] Khoury J. et al ., Fundamentals of genetic Epidemiology, Oxford University Press, NY , 1993
[1002] Kim UJ. et al. (1996) genomics 34: 213-218.
[1003] Klein et al. (1987) Nature . 327: 70-73.
[1004] Koch, et al., J. Biol. Chem. 265 (29): 17786-17791 (1990)
[1005] Kohler, G. and Milstein, C., Nature 256: 495 (1975)
[1006] Koller et al. (1992) Annu. Rev. Immunol . 10: 705-730.
[1007] Kozal MJ, Shah N, Shen N, Yang R, Fucini R, Merigan TC, Richman DD, Morris D, Hubbell E, Chee M, Gingeras TR, Nat Med 1996; 2 (7): 753-759
[1008] Lander and Schork, Science, 265, 2037-2048, 1994
[1009] Landegren U. et al. (1998) Genome Research , 8: 769-776.
[1010] Lange K. (1997) Mathematical and Statistical Methods for genetic Analysis . Springer, New York.
[1011] Lee et al., FEBS Lett. 445 231: 236 (1999).
[1012] Lenhard T. et al. (1996) gene. 169: 187-190.
[1013] Liautard, J. et al. Cytokinde 9 (4): 233-241 (1997)
[1014] Lin MW et al. Hum. genet., 99 (3): 417-420 (1997)
[1015] Linton MF et al. (1993) J. Clin. Invest. 92: 3029-3037.
[1016] Liu Z. et al. (1994) Proc. Natl. Acad. Sci. USA. 91: 4528-4262.
[1017] Livak et al., Nature genetics , 9: 341-342, 1995
[1018] Livak KJ, Hainer JW, Hum Mutat 1994; 3 (4): 379-385
[1019] Lockhart et al. Nature Biotechnology 14: 1675-1680, 1996
[1020] Lucas A. H., 1994, In: Development and Clinical Uses of Haempophilus b Conjugate;
[1021] Manger et al., Anal. Biochem. 214: 190-194 (1993).
[1022] Mansour SL et al. (1988) Nature. 336: 348-352.
[1023] Marshall RL et al. (1994) PCR Methods and Applications. 4: 80-84.
[1024] McCormick et al. (1994) genet. Anal. Tech. Appl. 11: 158-164.
[1025] McLaughlin BA et al. (1996) Am. J. Hum. genet . 59: 561-569.
[1026] Morena et al., Annals NY Acad. Sci. 102-117 (1999)
[1027] Mori, et al., Nature 350: 398-402 (1991)
[1028] Morrison, Science 229: 1202 (1985)
[1029] Morton NE, Am . J. Hum . Genet ., 7: 277-318, 1955
[1030] Muller, Y.A. et al. Structure 6 (9): 1153-1167 (1998)
[1031] Mullinax, R.L. et al. BioTechniques 12 (6): 864-869 (1992)
[1032] Muzyczka et al. (1992) Curr. Topics in Micro. and Immunol . 158: 97-129.
[1033] Nada S. et al. (1993) Cell 73: 1125-1135.
[1034] Nagy A. et al., 1993, Proc. Natl. Acad. Sci. USA, 90: 8424-8428.
[1035] Naramura, M. et al. Immunol. Lett. 39: 91-99 (1994)
[1036] Narang SA, Hsiung HM, Brousseau R, Methods Enzymol 1979; 68: 90-98
[1037] Neda et al. (1991) J. Biol. Chem . 266: 14143-14146.
[1038] Newton et al. (1989) Nucleic Acids Res. 17: 2503-2516.
[1039] Nickerson DA et al. (1990) Proc. Natl. Acad. Sci. USA 87: 8923-8927.
[1040] Nicolau C. et al., 1987, Methods Enzymol., 149: 157-76.
[1041] Nicolau et al. (1982) Biochim. Biophys. Acta . 721: 185-190.
[1042] Nissinoff, J. Immunol. 147 (8): 2429-2438 (1991).
[1043] Nyren P, Pettersson B, Uhlen M, Anal Biochem 1993; 208 (1): 171-175
[1044] O'Reilly et al. (1992) Baculovirus Expression Vectors: A Laboratory Manual . WH Freeman and Co., New York.
[1045] Ohno et al. (1994) Science. 265: 781-784.
[1046] Oi et al., BioTechniques 4: 214 (1986)
[1047] Oldenburg K.R. et al., 1992, Proc. Natl. Acad. Sci., 89: 5393-5397.
[1048] Olesen et al., Voltage gated ion channel modulators , 7-8 December, Philadelphia PA, USA (1995)
[1049] Orita et al. (1989) Proc. Natl. Acad. Sci. USA 86: 2776-2770.
[1050] Ott J., Analysis of Human genetic Linkage , John Hopkins University Press , Baltimore, 1991
[1051] Ouchterlony, O. et al., Chap. 19 in: Handbook of Experimental Immunology D. Wier (ed) Blackwell (1973)
[1052] Padlan E.A., Molecular Immunology 28 (4/5): 489-498 (1991)
[1053] Parmley and Smith, gene, 1988, 73: 305-318
[1054] Pastinen et al., Genome Research 1997; 7: 606-614
[1055] Pearson and Lipman, 1988, Proc. Natl. Acad. Sci. USA 85 (8): 2444-2448
[1056] Pease S. ans William R. S., 1990, Exp. Cell. Res., 190: 209-211.
[1057] Perlin et al. (1994) Am. J. Hum. genet. 55: 777-787.
[1058] Persic, L. et al. gene 187 9-18 (1997)
[1059] Peterson et al., 1993, Proc. Natl. Acad. Sci. USA, 90: 7593-7597.
[1060] Pietu et al. Genome Research 6: 492-503, 1996
[1061] Pitard, V. et al. J. Immunol. Methods 205 (2): 177-190 (1997)
[1062] Potter et al. (1984) Proc. Natl. Acad. Sci. USA 81 (22): 7161-7165.
[1063] Prat, M. et al. J. Cell. Sci. 111 (Pt2): 237-247 (1998)
[1064] Ramunsen et al., 1997, Electrophoresis, 18: 588-598.
[1065] Reid LH et al. (1990) Proc. Natl. Acad. Sci. USA 87: 4299-4303.
[1066] Risch, N. and Merikangas, K. ( Science , 273: 1516-1517, 1996
[1067] Robertson E., 1987, Embryo-derived stem cell lines. In: EJ Robertson Ed. Teratocarcinomas and embrionic stem cells: a practical approach . IRL Press, Oxford, pp. 71.
[1068] Roguska M.A. et al. PNAS 91: 969-973), (1994)
[1069] Rossi et al., Pharmacol. Ther . 50: 245-254, (1991)
[1070] Roth JA et al. (1996) Nature Medicine. 2 (9): 985-991.
[1071] Roux et al. (1989) Proc. Natl. Acad. Sci. USA 86: 9079-9083.
[1072] Ruano et al. (1990) Proc. Natl. Acad. Sci. USA 87: 6296-6300.
[1073] Sambrook, J., Fritsch, EF, and T. Maniatis. (1989) Molecular Cloning: A Laboratory Manual. 2ed. Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.
[1074] Samson M, et al. (1996) Nature , 382 (6593): 722-725.
[1075] Samulski et al. (1989) J. Virol . 63: 3822-3828.
[1076] Sanchez-Pescador R. (1988) J. Clin. Microbiol . 26 (10): 1934-1938.
[1077] Sarkar, G. and Sommer SS (1991) Biotechniques .
[1078] Sauer B. et al. (1988) Proc. Natl. Acad. Sci. USA 85: 5166-5170.
[1079] Sawai, H. et al. AJRI 34: 26-34 (1995)
[1080] Schedl A. et al., 1993a, Nature, 362: 258-261.
[1081] Schedl et al., 1993b, Nucleic Acids Res., 21: 4783-4787.
[1082] Schena et al. Science 270: 467-470, 1995
[1083] Schena et al., 1996, Proc Natl Acad Sci U S A ,. 93 (20): 10614-10619.
[1084] Schneider et al. (1997) Arlequin: A Software For Population genetics Data Analysis. University of geneva.
[1085] Schwartz and Dayhoff, eds., 1978, Matrices for Detecting Distance Relationships: Atlas of Protein sequence and Structure, Washington: National Biomedical Research Foundation
[1086] Sczakiel G. et al. (1995) Trends Microbiol . 3 (6): 213-217.
[1087] Shay J.W. et al., 1991, Biochem. Biophys. Acta, 1072: 1-7.
[1088] Sheffield, VC et al. (1991) Proc. Natl. Acad. Sci. USA 49: 699-706.
[1089] Shizuya et al. (1992) Proc. Natl. Acad. Sci. USA 89: 8794-8797.
[1090] Shoemaker DD, et al., Nat genet 1996; 14 (4): 450-456
[1091] Shu, L. et al. PNAS 90: 7995-7999 (1993)
[1092] Skerra, A. et al. Science 240: 1038-1040. (1988)
[1093] Smith (1957) Ann. Hum. genet. 21: 254-276.
[1094] Smith et al. (1983) Mol. Cell. Biol . 3: 2156-2165.
[1095] Sosnowski RG, et al., Proc Natl Acad Sci USA 1997; 94: 1119-1123
[1096] Sowdhamini et al., Protein Engineering 10: 207, 215 (1997)
[1097] Spielmann S. and Ewens WJ, Am. J. Hum. genet ., 62: 450-458, 1998
[1098] Spielmann S. et al., Am. J. Hum. genet ., 52: 506-516, 1993
[1099] Sternberg N.L. (1992) Trends genet. 8: 1-16.
[1100] Sternberg NL (1994) Mamm. Genome. 5: 397-404.
[1101] Stryer, L., Biochemistry, 4th edition, 1995
[1102] Studnicka G.M. et al. Protein Engineering 7 (6): 805-814 (1994)
[1103] Syvanen AC, Clin Chim Acta 1994; 226 (2): 225-236
[1104] Szabo A. et al. Curr Opin Struct Biol 5, 699-705 (1995)
[1105] Tacson et al. (1996) Nature Medicine . 2 (8): 888-892.
[1106] Taryman, RE et al. Neuron 14 (4): 755-762 (1995) Schaid DJ et al., Genet . Epidemiol ., 13: 423-450, 1996
[1107] Te Riele et al. (1990) Nature. 348: 649-651.
[1108] Terlau et al., Naturwissenschaften 85: 437-444 (1998)
[1109] Terwilliger JD and Ott J., Handbook of Human genetic Linkage , John Hopkins University Press , London , 1994
[1110] Thomas KR et al. (1986) Cell . 44: 419-428.
[1111] Thomas KR et al. (1987) Cell . 51: 503-512.
[1112] Thompson et al., 1994, Nucleic Acids Res. 22 (2): 4673-4680
[1113] Tur-Kaspa et al. (1986) Mol. Cell. Biol. 6: 716-718.
[1114] Tyagi et al. (1998) Nature Biotechnology. 16: 49-53.
[1115] Urdea MS (1988) Nucleic Acids Research. 11: 4937-4957.
[1116] Urdea MS et al. (1991) Nucleic Acids Symp. Ser . 24: 197-200.
[1117] Vaitukaitis, J. et al. J. Clin. Endocrinol. Metab. 33: 988-991 (1971)
[1118] Valadon P., et al., 1996, J. Mol. Biol., 261: 11-22.
[1119] Van der Lugt et al. (1991) gene . 105: 263-267.
[1120] Vil, H. et al. PNAS 89: 11337-11341 (1992)
[1121] Vlasak R. et al. (1983) Eur. J. Biochem . 135: 123-126.
[1122] Wabiko et al. (1986) DNA. 5 (4): 305-314.
[1123] Walker et al. (1996) Clin. Chem. 42: 9-13.
[1124] Wang et al., 1997, Chromatographia, 44: 205-208.
[1125] Weir, BS (1996) genetic data Analysis II: Methods for Discrete population genetic Data , Sinauer Assoc., Inc., Sunderl, and MA, USA .
[1126] Westerink M.A.J., 1995, Proc. Natl. Acad. Sci., 92: 4021-4025
[1127] White, MB et al. (1992) genomics. 12: 301-306.
[1128] White, MB et al. (1997) genomics. 12: 301-306.
[1129] Williams, et al. Science 257: 389-395 (1992)
[1130] Wong et al. gene . 10: 87-94 (1980)
[1131] Wood S.A. et al., 1993, Proc. Natl. Acad. Sci. USA, 90: 4582-4585.
[1132] Wu and Wu (1987) J. Biol. Chem. 262: 4429-4432.
[1133] Wu and Wu (1988) Biochemistry . 27: 887-892.
[1134] Wu et al. (1989) Proc. Natl. Acad. Sci. USA 86: 2757.
[1135] Yagi T. et al. (1990) Proc. Natl. Acad. Sci. USA 87: 9918-9922.
[1136] Yoon, D.Y. et al. J. Immunol. 160 (7): 3170-3179 (1998)
[1137] Zhang et al., Neuropharmacology 32 (11): 1075-1088 (1993)
[1138] Zhao et al., Am. J. Hum. genet ., 63: 225-240, 1998
[1139] Zheng, X.X. et al. J. Immunol. 154: 5590-5600 (1995)
[1140] Zhu, Z. et al. Cancer Res. 58 (15): 3209-3214 (1998)
[1141] Zou YR et al. (1994) Curr. Biol. 4: 1099-1103.
权利要求:
Claims (23)
[1" claim-type="Currently amended] An isolated, purified or recombinant polynucleotide comprising one of the nucleotide sequences shown in SEQ ID NOs 1 to 4 or 6, or sequences complementary to these sequences.
[2" claim-type="Currently amended] The isolated, purified or recombinant polynucleotide comprising a continuous span of at least 50 nucleotides of SEQ ID NO 4, wherein the polynucleotide encodes a biologically active CanIon polypeptide.
[3" claim-type="Currently amended] An isolated, purified or recombinant polynucleotide encoding a human CanIon polypeptide comprising the amino acid sequence of SEQ ID NO 5, or a biologically active fragment thereof.
[4" claim-type="Currently amended] The polynucleotide according to any one of claims 1 to 3, which is attached to a solid support.
[5" claim-type="Currently amended] An array of polynucleotides comprising at least one polynucleotide according to claim 4.
[6" claim-type="Currently amended] 6. The array of claim 5, wherein the array is addressable.
[7" claim-type="Currently amended] The polynucleotide according to any one of claims 1 to 4, further comprising a label.
[8" claim-type="Currently amended] Recombinant vector comprising the polynucleotide of any one of claims 1 to 3.
[9" claim-type="Currently amended] A recombinant vector comprising a polynucleotide of claim 2 or 3 operably linked to a promoter.
[10" claim-type="Currently amended] A host cell comprising the recombinant vector of claim 8.
[11" claim-type="Currently amended] A human exclusion host animal or mammal comprising the recombinant vector of claim 8.
[12" claim-type="Currently amended] Mammalian host cells or human exclusion host mammals comprising the CanIon gene disrupted by homologous recombination with a knock out vector.
[13" claim-type="Currently amended] An isolated, purified or recombinant polypeptide comprising the amino acid sequence shown in SEQ ID NO 5, or a biologically active fragment thereof.
[14" claim-type="Currently amended] A method for producing a polypeptide, wherein the method consists of the following steps:
a) providing a cell population comprising a polynucleotide encoding the polypeptide of claim 13 operably linked to a promoter;
b) culturing the cell population under conditions conducive to the production of said polypeptide in cells;
c) Purify the polypeptide from the cell population.
[15" claim-type="Currently amended] 14. A method of binding an anti-CanIon antibody to a polypeptide of claim 13, comprising contacting the antibody with the polypeptide under conditions such that the antibody and polypeptide can specifically bind.
[16" claim-type="Currently amended] A method for detecting the expression of the CanIon gene in a cell, the method comprising the following steps:
i) a polynucleotide which hybridizes with the polynucleotide of claims 1, 2 or 3 under stringent conditions; Or ii) contacting the cell or cell extract with a polypeptide that specifically binds to the polypeptide of claim 13;
b) identifying the presence of hybridization between polynucleotide and RNA species in said cell or cell extract, or the presence of polypeptide and protein binding in said cell or cell extract;
Here, the presence of hybridization or binding suggests that the CanIon gene is expressed intracellularly.
[17" claim-type="Currently amended] The method of claim 16, wherein the polynucleotide is an oligonucleotide primer and hybridization is confirmed by detecting the presence of an amplification product comprising the primer sequence.
[18" claim-type="Currently amended] The method of claim 16, wherein the polypeptide is an anti-CanIon antibody.
[19" claim-type="Currently amended] A method of identifying a candidate modulator of a CanIon polypeptide, comprising the following steps:
a) contacting the polypeptide of claim 13 with the test compound;
b) confirming that said compound specifically binds said polypeptide;
Here, the confirmation that the compound specifically binds to the polypeptide suggests that the compound is a candidate modulator of the CanIon polypeptide.
[20" claim-type="Currently amended] 20. The method of claim 19, further comprising measuring the bioactivity of the CanIon polypeptide in the presence of the candidate modulator, wherein the change in bioactivity of the CanIon polypeptide in the presence of the candidate modulator is compared to the activity in the absence of the candidate modulator. A method characterized by suggesting that the substance is a modulator of the CanIon polypeptide.
[21" claim-type="Currently amended] A method for identifying a modulator of a CanIon polypeptide, comprising the following steps:
a) contacting the polypeptide of claim 13 with the test compound;
b) confirming the activity of said polypeptide in the absence of said compound;
Here, the identification of the difference in activity in the presence of the compound compared to the activity in the absence of the compound suggests that the compound is a modulator of the CanIon polypeptide.
[22" claim-type="Currently amended] 22. The method of claim 20 or 21, wherein the polypeptide is present in the cell or cell membrane and the bioactivity is voltage-gated ion channel activity.
[23" claim-type="Currently amended] A process for preparing a pharmaceutical composition, comprising the following steps:
a) identifying modulators of the CanIon polypeptide using the method of any one of claims 19-22;
b) mixing the modulator with a physiologically acceptable carrier.
类似技术:
公开号 | 公开日 | 专利标题
Loftus et al.1997|Murine model of Niemann-Pick C disease: mutation in a cholesterol homeostasis gene
Ferguson et al.1997|Cloning of Tabby, the murine homolog of the human EDA gene: evidence for a membrane-associated protein with a short collagenous domain
Basson et al.1997|Mutations in human cause limb and cardiac malformation in Holt-Oram syndrome
DeSilva et al.1997|The human reelin gene: isolation, sequencing, and mapping on chromosome 7.
Gennarelli et al.1995|Survival motor-neuron gene transcript analysis in muscles from spinal muscular-atrophy patients
Yasunaga et al.2000|OTOF encodes multiple long and short isoforms: genetic evidence that the long ones underlie recessive deafness DFNB9
Zhang et al.1994|Positional cloning of the mouse obese gene and its human homologue
US6228594B1|2001-05-08|Method for determining the presence or absence of a hereditary hemochromatosis gene mutation
Herzing et al.2001|The human aminophospholipid-transporting ATPase gene ATP10C maps adjacent to UBE3A and exhibits similar imprinted expression
Onuchic et al.2002|PKHD1, the polycystic kidney and hepatic disease 1 gene, encodes a novel large protein containing multiple immunoglobulin-like plexin-transcription–factor domains and parallel beta-helix 1 repeats
Lin et al.1999|The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin | receptor 2 gene
DiDonato et al.1997|Cloning, characterization, and copy number of the murine survival motor neuron gene: homolog of the spinal muscular atrophy-determining gene
US6521747B2|2003-02-18|Haplotypes of the AGTR1 gene
Kestilä et al.1998|Positionally cloned gene for a novel glomerular protein—nephrin—is mutated in congenital nephrotic syndrome
Dietz et al.1993|Four novel FBN1 mutations: significance for mutant transcript level and EGF-like domain calcium binding in the pathogenesis of Marfan syndrome
AU749198B2|2002-06-20|A novel human G-protein coupled receptor
Mochizuki et al.1998|Human PON2 gene at 7q21. 3: cloning, multiple mRNA forms, and missense polymorphisms in the coding sequence
Dgany et al.2002|Congenital dyserythropoietic anemia type I is caused by mutations in codanin-1
Noben-Trauth et al.1996|A candidate gene for the mouse mutation tubby
US6265546B1|2001-07-24|Prostate cancer gene
US7045675B2|2006-05-16|Genes for Niemann-Pick type C disease
Bernal et al.1998|Insulin receptor substrate-2 amino acid polymorphisms are not associated with random type 2 diabetes among Caucasians.
EP0705903B2|2009-08-12|Mutations in the 17q-linked breast and ovarian cancer susceptibility gene
Sahoo et al.1999|Mutations in the gene encoding KRIT1, a Krev-1/rap1a binding protein, cause cerebral cavernous malformations |
EP0915973B1|2009-07-01|Gene involved in cadasil, method of diagnosis and therapeutic application
同族专利:
公开号 | 公开日
EA006367B1|2005-12-29|
EA200500974A1|2006-02-24|
EA200300642A1|2003-12-25|
DE60123109T2|2007-05-16|
CA2430712A1|2002-06-13|
UA79927C2|2007-08-10|
AT339500T|2006-10-15|
AU3357302A|2002-06-18|
EA008252B1|2007-04-27|
ES2271092T3|2007-04-16|
CN1777676A|2006-05-24|
DE60123109D1|2006-10-26|
AU2002233573B2|2007-03-01|
EP1339840B1|2006-09-13|
JP2004525614A|2004-08-26|
US20040091497A1|2004-05-13|
IL156073D0|2003-12-23|
US20080184381A1|2008-07-31|
WO2002046404A2|2002-06-13|
BR0115937A|2003-09-30|
EP1339840A2|2003-09-03|
MXPA03005069A|2003-09-05|
WO2002046404A3|2002-12-12|
WO2002046404A9|2002-08-15|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题
法律状态:
2000-12-05|Priority to US25131700P
2000-12-05|Priority to US60/251,317
2001-12-04|Application filed by 쟝세
2001-12-04|Priority to PCT/IB2001/002798
2003-09-19|Publication of KR20030074643A
优先权:
申请号 | 申请日 | 专利标题
US25131700P| true| 2000-12-05|2000-12-05|
US60/251,317|2000-12-05|
PCT/IB2001/002798|WO2002046404A2|2000-12-05|2001-12-04|Schizophrenia-related voltage-gated ion channel gene and protein|
[返回顶部]