WO2003089469A2 - Cation channel proteins - Google Patents

Cation channel proteins Download PDF

Info

Publication number
WO2003089469A2
WO2003089469A2 PCT/GB2003/001655 GB0301655W WO03089469A2 WO 2003089469 A2 WO2003089469 A2 WO 2003089469A2 GB 0301655 W GB0301655 W GB 0301655W WO 03089469 A2 WO03089469 A2 WO 03089469A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
polypeptide
disease
nucleic acid
acid molecule
Prior art date
Application number
PCT/GB2003/001655
Other languages
French (fr)
Other versions
WO2003089469A3 (en
Inventor
Anna Elizabeth Lobley
David Michalovich
Kathryn Elizabeth Allen
Lindsey Reynolds
Valerie Nathalie Pierron
Janet Marjorie Allen
Original Assignee
Inpharmatica Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Inpharmatica Limited filed Critical Inpharmatica Limited
Priority to AU2003232308A priority Critical patent/AU2003232308A1/en
Publication of WO2003089469A2 publication Critical patent/WO2003089469A2/en
Publication of WO2003089469A3 publication Critical patent/WO2003089469A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants

Definitions

  • This invention relates to novel proteins, termed INPIONCH03 and INPIONCH04, herein identified as members of the PKD/REJ family of cation channels and to the use of these proteins and nucleic acid sequences from the encoding genes in the diagnosis, prevention and treatment of disease.
  • the PKD/REJ (polycystic kidney disease-receptor for egg jelly) family is small family of modular proteins whose members possess a characteristic combination of structural and functional features.
  • a generic topology for members of the PKD/REJ family of cation channels is shown in Figure 1.
  • the characteristic structural and functional features of the PKD/REJ family include at the N-terminus a REJ (sea urchin receptor for egg jelly)-like domain and a GPS domain (GPCR proteolytic site), followed by eleven transmembrane domains, a cytoplasmic PLAT (or LH2) domain (polycystin-1 lipoxygenase, alpha-toxin or lipoxygenase homology domain; a lipid binding site) and an ion-transfer domain (similar to those found in cationic ion channels).
  • the different members of the family have varied sequences at the extreme N-terminus and C-terminus.
  • the known members of the PKD/REJ family are summarised below in Table 1.
  • Ion channels may be useful for the treatment, prevention and/or diagnosis of medical conditions and diseases which include, but are not limited to, cardiovascular disease, hypertension, heart arrhythmia, angina, myocardial infarction, tachycardia, cerebrovascular disease, thromboembolism, neurological and psychiatric disorders, including stroke, epilepsy, migraine, pain, depression, neuralgia, Alzheimer's disease, brain injury, schizophrenia, Huntington's disease, motor neurone disease, Parkinson's disease, neuropathy, neuromuscular disorder, micturition disorder, impotence, diabetes, dermatitis, pulmonary disease, asthma, cystic f ⁇ brosis, mucous membrane disorders including sinusitis, COPD, rhinitis and cough, leukemia, ocular disease, glaucoma, retinopathy and immune disorders.
  • cardiovascular disease hypertension
  • heart arrhythmia angina
  • myocardial infarction tachycardia
  • cerebrovascular disease thromboembolism
  • ADPKD autosomal dominant polycystic kidney disease
  • the acrosome reaction which occurs as the sperm penetrates the egg during fertilisation and is triggered by the suREJ protein in sea urchins, is characterised by various signal transduction events including Ca 2+ , Na + , H + and K + transportation, cyclic nucleotide changes and protein kinase C activation.
  • the suREJ protein is thought to be involved in the initial Ca 2+ influx that triggers the acrosome reaction.
  • the sequence similarity and expression pattern of the PKDREJ protein suggests it is a mammalian equivalent of the suREJ protein and therefore may have a central role in human fertilisation via an involvement in the acrosome reaction. Since the acrosome reaction is required for animal fertilisation it offers the potential for the development of novel methods of non-hormonal contraception. The precise steps involved in the initiation of the acrosome reaction in animals have not yet been determined (Hughes et al, Hum. Mol. Genet., 1999, 8(3):543-9). Modulation of the acrosome reaction in humans would be of great importance in reproductive health, since it would allow new forms of infertility remedies or contraceptive methods to be devised.
  • PKD2L2 is also believed to play a role in fertilisation because of its specific expression in the testis.
  • PKDILI localises to the Ley dig cells of the testis, a known location of testosterone production, suggesting a possible role in fertility and/or sexual maturation.
  • PKDILI is also expressed in the heart, and although no role for this localised expression of PKDILI has yet been suggested, it may be implicated in cardiac diseases.
  • members of the PKD/REJ family of cation channels may be useful as contraceptive agents or for the treatment, prevention and or diagnosis of medical conditions and diseases which include, but are not limited to, renal disease, polycystic kidney disease, glomerulonephritis, fertility disorders, infertility, reproductive disorders, polycystic ovary disease, polycystic diseases, endocrine diseases, testosterone deficiency, testosterone-related disorder, testicular cancer, prostate cancer, gastrointestinal disorders, developmental disorders, brain tumour, sexual dysfunction, psychological disorder, alopecia, hirsuitism, diseases related to muscle tone, including vascular muscle disorders such as migraine, pre-eclampsia, hypertension and cardiac arrhythmias; bladder muscle tone disorders, such as urinary incontinence, inability to void, urinary tract infection; gastrointestinal smooth muscle tone disorder, such as Gl motility disorder, gastrointestinal reflux diorder (GERD), reflux oesophagitis, paralytic ileus, irritable bowel syndrome (IBS
  • the identification of members of the PKD/REJ family of cation channels is of extreme importance in increasing the understanding of the underlying pathways that lead to the disease states and associated disease states mentioned above, and in developing more effective gene and/or drug therapies to treat these disorders, and also in increasing the understanding of the pathways involved in mammalian reproductive biology.
  • the invention is based on the discovery that the INPIONCH03 and INPIONCH04 proteins are cation channels, and moreover are members of the PKD/REJ family of cation channels.
  • polypeptide which:
  • (i) comprises the amino acid sequence as recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO-10, SEQ ID NO-12, SEQ ID NO:14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:
  • (ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or has an antigenic determinant in common with the polypeptides of (i); or
  • (i) comprises the amino acid sequence as recited in SEQ ID NO:56; (ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or has an antigenic determinant in common with the polypeptides of (i); or
  • a polypeptide which: (i) consists of the amino acid sequence as recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO-12, SEQ ID NO:14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:
  • (ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or having an antigenic determinant in common with the polypeptides of (i); or (iii) is a functional equivalent of (i) or (ii).
  • polypeptide having the sequence recited in SEQ ID NO:2 is referred to hereafter as "INPIONCH03 exon 1 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:4 is referred to hereafter as "INPIONCH03 exon 2 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:6 is referred to hereafter as "INPIONCH03 exon 3 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO: 8 is referred to hereafter as "INPIONCH03 exon 4 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO: 10 is referred to hereafter as "INPIONCH03 exon 5 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO: 12 is referred to hereafter as "INPIONCH03 exon 6 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO: 14 is referred to hereafter as "INPIONCH03 exon 7 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO: 16 is referred to hereafter as "INPIONCH03 exon 8 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO: 18 is referred to hereafter as "INPIONCH03 exon 9 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:20 is referred to hereafter as "INPIONCH03 exon 10 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO:22 is referred to hereafter as "INPIONCH03 exon 11 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:24 is referred to hereafter as "INPIONCH03 exon 12 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:26 is referred to hereafter as "INPIONCH03 exon 13 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:28 is referred to hereafter as "INPIONCH03 exon 14 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:30 is referred to hereafter as "INPIONCH03 exon 15 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO:32 is referred to hereafter as "INPIONCH03 exon 16 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:34 is referred to hereafter as "INPIONCH03 exon 17 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:36 is referred to hereafter as "INPIONCH03 exon 18 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:38 is referred to hereafter as "INPIONCH03 exon 19 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:40 is referred to hereafter as "INPIONCH03 exon 20 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO:42 is referred to hereafter as "INPIONCH03 exon 21 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:44 is referred to hereafter as "INPIONCH03 exon 22 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:46 is referred to hereafter as "INPIONCH03 exon 23 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:48 is referred to hereafter as "INPIONCH03 exon 24 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO: 50 is referred to hereafter as "INPIONCH03 exon 25 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO:52 is referred to hereafter as "INPIONCH03 exon 26 polypeptide".
  • polypeptide having the sequence recited in SEQ ID NO:54 is referred to hereafter as "INPIONCH03 exon 27 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO:56 is referred to hereafter as the "INPIONCH03 polypeptide”.
  • the term "INPIONCH03 polypeptides" as used herein includes polypeptides comprising the INPIONCH03 exon 1 polypeptide, the INPIONCH03 exon 2 polypeptide, the
  • INPIONCH03 exon 7 polypeptide the INPIONCH03 exon 8 polypeptide, the INPIONCH03 exon 9 polypeptide, the INPIONCH03 exon 10 polypeptide, the
  • INPIONCH03 exon 17 polypeptide the INPIONCH03 exon 18 polypeptide, the INPIONCH03 exon 19 polypeptide, the INPIONCH03 exon 20 polypeptide, the
  • INPIONCH03 exon 25 polypeptide the INPIONCH03 exon 26 polypeptide, the INPIONCH03 exon 27 polypeptide and the 1NPIONCH03 polypeptide.
  • (i) comprises the amino acid sequence as recited in SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72,
  • the polypeptide according to this embodiment of the first aspect of the invention comprises the amino acid sequence as recited in SEQ ID NO: 108;
  • (ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or has an antigenic determinant in common with the polypeptides of (i); or
  • (i) consists of the amino acid sequence as recited in SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72, SEQ ID NO:74, SEQ ID NO:76, SEQ ID NO:78, SEQ ID NO:80, SEQ ID NO:82, SEQ ID NO:84, SEQ ID NO:86, SEQ ID NO:88, SEQ ID NO:90, SEQ ID NO:92, SEQ ID NO:94, SEQ ID NO:96, SEQ ID NO:98, SEQ ID NO: 100, SEQ ID NO: 102,
  • SEQ ID NO: 104 SEQ ID NO: 106 and or SEQ ID NO: 108;
  • (ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or having an antigenic determinant in common with the polypeptides of (i); or
  • polypeptide having the sequence recited in SEQ ID NO:58 is referred to hereafter as "INPIONCH04 exon 1 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:60 is referred to hereafter as "INPIONCH04 exon 2 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:62 is referred to hereafter as "INPIONCH04 exon 3 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO:64 is referred to hereafter as "INPIONCH04 exon 4 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO: 66 is referred to hereafter as "INPIONCH04 exon 5 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:68 is referred to hereafter as "INPIONCH04 exon 6 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:70 is referred to hereafter as "INPIONCH04 exon 7 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:72 is referred to hereafter as "INPIONCH04 exon 8 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:74 is referred to hereafter as "INPIONCH04 exon 9 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO:76 is referred to hereafter as "INPIONCH04 exon 10 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:78 is referred to hereafter as "INPIONCH04 exon 11 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:80 is referred to hereafter as "INPIONCH04 exon 12 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO: 82 is referred to hereafter as "INPIONCH04 exon 13 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:84 is referred to hereafter as "INPIONCH04 exon 14 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO:86 is referred to hereafter as "INPIONCH04 exon 15 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:88 is referred to hereafter as "INPIONCH04 exon 16 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:90 is referred to hereafter as "INPIONCH04 exon 17 polypeptide”.
  • the polypeptide having the sequence recited in SEQ ID NO:92 is referred to hereafter as "INPIONCH04 exon 18 polypeptide”.
  • the polypeptide having the sequence recited in SEQ ID NO:94 is referred to hereafter as "INPIONCH04 exon 19 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO:96 is referred to hereafter as "INPIONCH04 exon 20 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO:98 is referred to hereafter as "INPIONCH04 exon 21 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO: 100 is referred to hereafter as "INPIONCH04 exon 22 polypeptide”.
  • the polypeptide having the sequence recited in SEQ ID NO: 102 is referred to hereafter as "INPIONCH04 exon 23 polypeptide".
  • the polypeptide having the sequence recited in SEQ ID NO: 104 is referred to hereafter as "INPIONCH04 exon 24 polypeptide”.
  • polypeptide having the sequence recited in SEQ ID NO: 106 is referred to hereafter as "INPIONCH04 exon 25 polypeptide".
  • polypeptide having the sequence recited in SEQ ID NO: 108 is referred to hereafter as the "INPIONCH04 polypeptide”.
  • INPIONCH04 polypeptides as used herein includes polypeptides comprising the INPIONCH04 exon 1 polypeptide, the INPIONCH04 exon 2 polypeptide, the
  • INPIONCH04 exon 5 polypeptide the INPIONCH04 exon 6 polypeptide, the INPIONCH04 exon 7 polypeptide, the INPIONCH04 exon 8 polypeptide, the
  • INPIONCH04 exon 15 polypeptide the INPIONCH04 exon 16 polypeptide, the INPIONCH04 exon 17 polypeptide, the INPIONCH04 exon 18 polypeptide, the
  • INPIONCH04 exon 23 polypeptide the INPIONCH04 exon 24 polypeptide, the INPIONCH04 exon 25 polypeptide and the INPIONCH04 polypeptide.
  • known genes that are members of the PKD/REJ family of cation channels have N-terminal regions of varying function and sizes.
  • the sequences disclosed in SEQ ID NO:56 and SEQ ID NO: 108 do not contain large N-terminal domains, and would therefore appear similar to PKD2 and PKD2L2.
  • the closest homologues to SEQ ID NO:56 and SEQ ID NO: 108 are the sea urchin REJ3 protein and the PKDREJ 1 human and mouse proteins.
  • PKDREJ 1 proteins both contain substantially larger N-terminal domains than those of the PKD2 and PKD2L2 proteins. Therefore, it is considered by the Applicant that there may be one, and possibly two or more, further exons 5' to SEQ ID NO:l or SEQ ID NO: 57 in the genome that will provide amino acids that are N-terminal to the beginning of the sequences given in SEQ ID NO:56 and SEQ ID NO: 108, respectively.
  • a polypeptide of the present invention preferably comprises the amino acid sequence given in SEQ ID NO:56 or SEQ ID NO: 108 but may also include additional N-terminal amino acid residues.
  • one embodiment of the first aspect of the invention provides homo- or hetero-multimeric complexes comprising the INPIONCH03 and/or INPIONCH04 polypeptides.
  • the invention provides a purified nucleic acid molecule which encodes a polypeptide of the first aspect of the invention.
  • the purified nucleic acid molecule comprises the nucleic acid sequence as recited in SEQ ID NO:l (encoding the INPIONCH03 exon 1 polypeptide), SEQ ID NO:3 (encoding the INPIONCH03 exon 2 polypeptide), SEQ ID NO:5 (encoding the INPIONCH03 exon 3 polypeptide), SEQ ID NO:7 (encoding the INPIONCH03 exon 4 polypeptide), SEQ ID NO:9 (encoding the INPIONCH03 exon 5 polypeptide), SEQ ID NO: 11 (encoding the INPIONCH03 exon 6 polypeptide), SEQ ID NO: 13 (encoding the INPIONCH03 exon 7 polypeptide), SEQ ID NO: 15 (encoding the INPIONCH03 exon 8 polypeptide), SEQ ID NO: 17 (encoding the INPIONCH03 exon 9 polypeptide), SEQ ID NO:19 (encoding the INPIONCH03 exon 10 polypeptide), SEQ ID NO:21 (encoding the
  • the invention further provides that the purified nucleic acid molecule consists of the nucleic acid sequence as recited in SEQ ID NO:l (encoding the INPIONCH03 exon 1 polypeptide), SEQ ID NO:3 (encoding the INPIONCH03 exon 2 polypeptide), SEQ ID NO:5 (encoding the INPIONCH03 exon 3 polypeptide), SEQ ID NO:7 (encoding the INPIONCH03 exon 4 polypeptide), SEQ ID NO:9 (encoding the INPIONCH03 exon 5 polypeptide), SEQ ID NO: 11 (encoding the INPIONCH03 exon 6 polypeptide), SEQ ID NO: 13 (encoding the INPIONCH03 exon 7 polypeptide), SEQ ID NO: 15 (encoding the INPIONCH03 exon 8 polypeptide), SEQ ID NO: 17 (encoding the INPIONCH03 exon 9 polypeptide), SEQ ID NO: 19 (encoding the INPIONCH03 exon 10 polypeptide), SEQ ID NO:21
  • the invention provides a purified nucleic acid molecule which hybridizes under high stringency conditions with a nucleic acid molecule of the second aspect of the invention.
  • the invention provides a vector, such as an expression vector, that contains a nucleic acid molecule of the second or third aspect of the invention.
  • the invention provides a host cell transformed with a vector of the fourth aspect of the invention.
  • the invention provides a ligand which binds specifically to the PKD/REJ cation channel family polypeptide of the first aspect of the invention.
  • the invention provides a compound that is effective to alter the expression of a natural gene which encodes a polypeptide of the first aspect of the invention or to regulate the activity of a polypeptide of the first aspect of the invention.
  • a compound of the seventh aspect of the invention may either increase (agonise) or decrease (antagonise) the level of expression of the gene or the activity of the polypeptide.
  • the identification of the function of the INPIONCH03 and INPIONCH04 polypeptides allows for the design of screening methods capable of identifying compounds that are effective in the treatment and/or diagnosis of disease.
  • Ligands and compounds according to the sixth and seventh aspects of the invention may be identified using such methods. These methods are included as aspects of the present invention.
  • the invention provides a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention, for use in therapy or diagnosis of diseases in which ion channels are implicated.
  • diseases include cardiovascular disease, hypertension, heart arrhythmia, angina, myocardial infarction, tachycardia cerebrovascular disease, thromboembolism, neurological and psychiatric disorders, including stroke, epilepsy, migraine, pain, depression, neuralgia, Alzheimer's disease, brain injury, schizophrenia,
  • the diseases are those in which members of the PKD/REJ family of cation channels are implicated.
  • diseases include renal disease, polycystic kidney disease, glomerulnephritis, fertility disorders, infertility, reproductive disorders, polycystic ovary disease, polycystic diseases, endocrine diseases, testosterone deficiency, testosterone-related disorder, testicular cancer, prostate cancer, gastrointestinal disorders, developmental disorders, brain tumour, sexual dysfunction, psychological disorder, alopecia, hirsuitism, diseases related to muscle tone, including vascular muscle disorders such as migraine, pre-eclampsia, hypertension and cardiac arrhythmias; bladder muscle tone disorders, such as urinary incontinence, inability to void, urinary tract infection; gastrointestinal smooth muscle tone disorder, such as Gl motility disorder, gastrointestinal reflux diorder (GERD), reflux oesophagitis, paralytic ileus, irritable bowel syndrome (IBS), diarrhoea including travellers diarrhoea and viral diarrhoea, constipation and malabsorption; skeletal muscle tone disorder, such
  • a polypeptide of the first aspect of the invention or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention, for use as a contraceptive agent.
  • These molecules may also be used in the manufacture of a medicament suitable for contraceptive use.
  • moieties of the first, second, third, fourth, fifth, sixth or seventh aspect of the invention may also be used in the manufacture of a medicament for the treatment of such diseases.
  • the invention provides a method of diagnosing a disease in a patient, comprising assessing the level of expression of a natural gene encoding a polypeptide of the first aspect of the invention or the activity of a polypeptide of the first aspect of the invention in tissue from said patient and comparing said level of expression or activity to a control level, wherein a level that is different to said control level is indicative of disease.
  • a method will preferably be carried out in vitro.
  • Similar methods may be used for monitoring the therapeutic treatment of disease in a patient, wherein altering the level of expression or activity of a polypeptide or nucleic acid molecule over the period of time towards a control level is indicative of regression of disease.
  • a preferred method for detecting polypeptides of the first aspect of the invention comprises the steps of: (a) contacting a ligand, such as an antibody, of the sixth aspect of the invention with a biological sample under conditions suitable for the formation of a ligand-polypeptide complex; and (b) detecting said complex.
  • a number of different such methods according to the ninth aspect of the invention exist, as the skilled reader will be aware, such as methods of nucleic acid hybridization with short probes, point mutation analysis, polymerase chain reaction (PCR) amplification and methods using antibodies to detect aberrant protein levels. Similar methods may be used on a short or long term basis to allow therapeutic treatment of a disease to be monitored in a patient.
  • the invention also provides kits that are useful in these methods for diagnosing disease.
  • the disease diagnosed by a method of the ninth aspect of the invention is a disease in which members of the PKD/REJ family of cation channels are implicated, as described above.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention, in conjunction with a pharmaceutically- acceptable carrier.
  • the present invention provides a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention, for use in the manufacture of a medicament for the diagnosis or treatment of a disease.
  • a polypeptide of the first aspect of the invention or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention, for use in the manufacture of a contraceptive agent.
  • the disease is a disease in which members of the PKD/REJ family of cation channels are implicated, as described above.
  • the invention provides a method of treating a disease in a patient comprising administering to the patient a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention.
  • a method of preventing pregnancy in a patient comprising administering to the patient a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention.
  • the disease is a disease in which members of the PKD/REJ family of cation channels are implicated, as described above.
  • the polypeptide, nucleic acid molecule, vector, host cell, ligand or compound administered to the patient should be an agonist.
  • the polypeptide, nucleic acid molecule, vector, host cell, ligand or compound administered to the patient should be an antagonist. Examples of such antagonists include antisense nucleic acid molecules, ribozymes and ligands, such as antibodies.
  • the invention provides transgenic or knockout non-human animals that have been transformed to express higher, lower or absent levels of a polypeptide of the first aspect of the invention.
  • Such transgenic animals are very useful models for the study of disease and may also be used in screening regimes for the identification of compounds that are effective in the treatment or diagnosis of such a disease.
  • the disease is a disease in which members of the PKD/REJ family of cation channels are implicated, as described above.
  • polypeptide includes any peptide or protein comprising two or more amino acids joined to each other by peptide bonds or modified peptide bonds, i.e. peptide isosteres. This term refers both to short chains (peptides and oligopeptides) and to longer chains (proteins).
  • the polypeptide of the present invention may be in the form of a mature protein or may be a pre-, pro- or prepro- protein that can be activated by cleavage of the pre-, pro- or prepro- portion to produce an active mature polypeptide.
  • the pre-, pro- or prepro- sequence may be a leader or secretory sequence or may be a sequence that is employed for purification of the mature polypeptide sequence.
  • the polypeptide of the first aspect of the invention may form part of a fusion protein.
  • the mature polypeptide may be fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol).
  • Polypeptides may contain amino acids other than the 20 gene-encoded amino acids, modified either by natural processes, such as by post-translational processing or by chemical modification techniques which are well known in the art.
  • modifications which may commonly be present in polypeptides of the present invention are glycosylation, lipid attachment, sulphation, gamma-carboxylation, for instance of glutamic acid residues, hydroxylation and ADP-ribosylation.
  • Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini.
  • blockage of the amino or carboxyl terminus in a polypeptide, or both, by a covalent modification is common in naturally-occurring and synthetic polypeptides and such modifications may be present in polypeptides of the present invention.
  • polypeptides that occur in a polypeptide often will be a function of how the polypeptide is made.
  • the nature and extent of the modifications in large part will be determined by the post-translational modification capacity of the particular host cell and the modification signals that are present in the amino acid sequence of the polypeptide in question. For instance, glycosylation patterns vary between different types of host cell.
  • the polypeptides of the present invention can be prepared in any suitable manner. Such polypeptides include isolated naturally-occurring polypeptides (for example purified from cell culture), recombinantly-produced polypeptides (including fusion proteins), synthetically-produced polypeptides or polypeptides that are produced by a combination of these methods.
  • the functionally-equivalent polypeptides of the first aspect of the invention may be polypeptides that are homologous to the INPIONCH03 and INPIONCH04 polypeptides.
  • Two polypeptides are said to be "homologous", as the term is used herein, if the sequence of one of the polypeptides has a high enough degree of identity or similarity to the sequence of the other polypeptide. "Identity” indicates that at any particular position in the aligned sequences, the amino acid residue is identical between the sequences. "Similarity” indicates that, at any particular position in the aligned sequences, the amino acid residue is of a similar type between the sequences.
  • Homologous polypeptides therefore include natural biological variants (for example, allelic variants or geographical variations within the species from which the polypeptides are derived) and mutants (such as mutants containing amino acid substitutions, insertions or deletions) of the INPIONCH03 or INPIONCH04 polypeptides.
  • Such mutants may include polypeptides in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code.
  • Such substitutions are among Ala, Val, Leu and He; among Ser and Thr; among the acidic residues Asp and Glu; among Asn and Gin; among the basic residues Lys and Arg; or among the aromatic residues Phe and Tyr.
  • Particularly preferred are variants in which several, i.e. between 5 and 10, 1 and 5, 1 and 3, 1 and 2 or just 1 amino acids are substituted, deleted or added in any combination.
  • silent substitutions, additions and deletions which do not alter the properties and activities of the protein. Also especially preferred in this regard are conservative substitutions.
  • Such mutants also include polypeptides in which one or more of the amino acid residues includes a substituent group.
  • polypeptides of the first aspect of the invention have a degree of sequence identity with the INPIONCH03 or INPIONCH04 polypeptides, or with active fragments thereof, of greater than 80%. More prefe ⁇ ed polypeptides have degrees of identity of greater than 85%, 90%, 95%, 98% or 99%, respectively.
  • the polypeptides whose sequences are recited in accession numbers XP_091397.1, BAB67772.1 and XP_056635.1 are specifically excluded from the scope of this aspect of the invention.
  • the polypeptides of the first aspect of the invention also include fragments of the INPIONCH03 and INPIONCH04 polypeptides and fragments of the functional equivalents of the INPIONCH03 and INPIONCH04 polypeptides, provided that those fragments are members of the PKD/REJ family of cation channels or have an antigenic determinant in common with the INPIONCH03 or INPIONCH04 polypeptides.
  • fragment refers to a polypeptide having an amino acid sequence that is the same as part, but not all, of the amino acid sequence of the INPIONCH03 or INPIONCH04 polypeptides or one of their functional equivalents.
  • the fragments should comprise at least n consecutive amino acids from the sequence and, depending on the particular sequence, n preferably is 7 or more (for example, 8, 10, 12, 14, 16, 18, 20 or more). Small fragments may form an antigenic determinant.
  • polypeptides of the present invention or their immunogenic fragments can be used to generate ligands, such as polyclonal or monoclonal antibodies, that are immunospecific for the polypeptides.
  • ligands such as polyclonal or monoclonal antibodies
  • Such antibodies may be employed to isolate or to identify clones expressing the polypeptides of the invention or to purify the polypeptides by affinity chromatography.
  • the antibodies may also be employed as diagnostic or therapeutic aids, amongst other applications, as will be apparent to the skilled reader.
  • immunospecific means that the antibodies have substantially greater affinity for the polypeptides of the invention than their affinity for other related polypeptides in the prior art.
  • antibody refers to intact molecules as well as to fragments thereof, such as Fab, F(ab')2 and Fv, which are capable of binding to the antigenic determinant in question. Such antibodies thus bind to the polypeptides of the first aspect of the invention.
  • substantially greater affinity we mean that there is a measurable increase in the affinity for a polypeptide of the invention as compared with the affinity for known cell- surface receptors.
  • the affinity is at least 1.5-fold, 2-fold, 5-fold 10-fold, 100-fold, 10 3 -fold, 10 4 - fold, 10 5 -fold or 10 6 -fold greater for a polypeptide of the invention than for known cell- surface receptor polypeptides .
  • a selected mammal such as a mouse, rabbit, goat or horse
  • a polypeptide of the first aspect of the invention may be immunised with a polypeptide of the first aspect of the invention.
  • the polypeptide used to immunise the animal can be derived by recombinant DNA technology or can be synthesized chemically.
  • the polypeptide can be conjugated to a carrier protein.
  • Commonly used carriers to which the polypeptides may be chemically coupled include bovine serum albumin, thyroglobulin and keyhole limpet haemocyanin.
  • the coupled polypeptide is then used to immunise the animal. Serum from the immunised animal is collected and treated according to known procedures, for example by immunoaffinity chromatography.
  • Monoclonal antibodies to the polypeptides of the first aspect of the invention cam also be readily produced by one skilled in the art.
  • the general methodology for making monoclonal antibodies using hybridoma technology is well known (see, for example, Kohler, G. and Milstein, C, Nature 256: 495-497 (1975); Kozbor et al, Immunology Today 4: 72 (1983); Cole et al, 77-96 in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc. (1985).
  • Panels of monoclonal antibodies produced against the polypeptides of the first aspect of the invention can be screened for various properties, i.e., for isotype, epitope, affinity, etc.
  • Monoclonal antibodies are particularly useful in purification of the individual polypeptides against which they are directed.
  • genes encoding the monoclonal antibodies of interest may be isolated from hybridomas, for instance by PCR techniques known in the art, and cloned and expressed in appropriate vectors.
  • Chimeric antibodies, in which non-human variable regions are joined or fused to human constant regions see, for example, Liu et al, Proc. Natl. Acad. Sci. USA, 84, 3439 (1987)), may also be of use.
  • the antibody may be modified to make it less immunogenic in an individual, for example by humanisation (see Jones et al, Nature, 321, 522 (1986); Verhoeyen et al, Science, 239, 1534 (1988); Kabat et al, J. Immunol., 147, 1709 (1991); Queen et al, Proc. Natl Acad. Sci. USA, 86, 10029 (1989); Gorman et al, Proc. Natl Acad. Sci. USA, 88, 34181 (1991); and Hodgson et al, Bio/Technology, 9, 421 (1991)).
  • humanisation see Jones et al, Nature, 321, 522 (1986); Verhoeyen et al, Science, 239, 1534 (1988); Kabat et al, J. Immunol., 147, 1709 (1991); Queen et al, Proc. Natl Acad. Sci. USA, 86, 10029 (1989); Gorman et
  • humanised antibody refers to antibody molecules in which the CDR amino acids and selected other amino acids in the variable domains of the heavy and/or light chains of a non-human donor antibody have been substituted in place of the equivalent amino acids in a human antibody.
  • the humanised antibody thus closely resembles a human antibody but has the binding ability of the donor antibody.
  • the antibody may be a "bispecific" antibody, that is an antibody having two different antigen binding domains, each domain being directed against a different epitope.
  • Phage display technology may be utilised to select genes which encode antibodies with binding activities towards the polypeptides of the invention either from repertoires of PCR amplified V-genes of lymphocytes from humans screened for possessing the relevant antibodies, or from naive libraries (McCafferty, J. et al, (1990), Nature 348, 552-554; Marks, J. et al, (1992) Biotechnology 10, 779-783).
  • the affinity of these antibodies can also be improved by chain shuffling (Clackson, T. et al, (1991) Nature 352, 624-628).
  • Antibodies generated by the above techniques have additional utility in that they may be employed as reagents in immunoassays, radioimmunoassays (RIA) or enzyme-linked immunosorbent assays (ELISA).
  • the antibodies can be labelled with an analytically-detectable reagent such as a radioisotope, a fluorescent molecule or an enzyme.
  • Preferred nucleic acid molecules of the second and third aspects of the invention are those which encode a polypeptide sequence as recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52, SEQ ID NO:54, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO
  • nucleic acid molecules may be used in the methods and applications described herein.
  • the nucleic acid molecules of the invention preferably comprise at least n consecutive nucleotides from the sequences disclosed herein where, depending on the particular sequence, n is 10 or more (for example, 12, 14, 15, 18, 20, 25, 30, 35, 40 or more).
  • nucleic acid molecules of the invention also include sequences that are complementary to nucleic acid molecules described above (for example, for antisense or probing purposes).
  • Nucleic acid molecules of the present invention may be in the form of RNA, such as mRNA, or in the form of DNA, including, for instance cDN A, synthetic DNA or genomic DNA. Such nucleic acid molecules may be obtained by cloning, by chemical synthetic techniques or by a combination thereof.
  • the nucleic acid molecules can be prepared, for example, by chemical synthesis using techniques such as solid phase phosphoramidite chemical synthesis, from genomic or cDNA libraries or by separation from an organism. RNA molecules may generally be generated by the in vitro or in vivo transcription of DNA sequences.
  • the nucleic acid molecules may be double-stranded or single-stranded.
  • Single-stranded DNA may be the coding strand, also known as the sense strand, or it may be the non- coding strand, also referred to as the anti-sense strand.
  • nucleic acid molecule also includes analogues of DNA and RNA, such as those containing modified backbones, and peptide nucleic acids (PNA).
  • PNA peptide nucleic acids
  • PNAs may be pegylated to extend their lifespan in a cell, where they preferentially bind complementary single stranded DNA and RNA and stop transcript elongation (Nielsen, P.E. et al. (1993) Anticancer Drug Des. 8:53-63).
  • a nucleic acid molecule which encodes a polypeptide of this invention may be identical to the coding sequence of one or more of the nucleic acid molecules disclosed herein.
  • SEQ ID NO:2 SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO.12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52, SEQ ID NO:54, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:
  • nucleic acid molecules may include, but are not limited to, the coding sequence for the mature polypeptide by itself; the coding sequence for the mature polypeptide and additional coding sequences, such as those encoding a leader or secretory sequence, such as a pro-, pre- or prepro- polypeptide sequence; the coding sequence of the mature polypeptide, with or without the aforementioned additional coding sequences, together with further additional, non-coding sequences, including non-coding 5' and 3' sequences, such as the transcribed, non-translated sequences that play a role in transcription (including termination signals), ribosome binding and mRNA stability.
  • the nucleic acid molecules may also include additional sequences which encode additional amino acids, such as those which provide additional functionalities.
  • nucleic acid molecules of the second and third aspects of the invention may also encode the fragments or the functional equivalents of the polypeptides and fragments of the first aspect of the invention.
  • a nucleic acid molecule may be a naturally-occurring variant such as a naturally-occurring allelic variant, or the molecule may be a variant that is not known to occur naturally.
  • non-naturally occurring variants of the nucleic acid molecule may be made by mutagenesis techniques, including those applied to nucleic acid molecules, cells or organisms.
  • variants in this regard are variants that differ from the aforementioned nucleic acid molecules by nucleotide substitutions, deletions or insertions.
  • the substitutions, deletions or insertions may involve one or more nucleotides.
  • the variants may be altered in coding or non-coding regions or both. Alterations in the coding regions may produce conservative or non-conservative amino acid substitutions, deletions or insertions.
  • the nucleic acid molecules of the invention can also be engineered, using methods generally known in the art, for a variety of reasons, including modifying the cloning, processing, and/or expression of the gene product (the polypeptide).
  • DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides are included as techniques which may be used to engineer the nucleotide sequences.
  • Site-directed mutagenesis may be used to insert new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, introduce mutations and so forth.
  • Nucleic acid molecules which encode a polypeptide of the first aspect of the invention may be ligated to a heterologous sequence so that the combined nucleic acid molecule encodes a fusion protein.
  • Such combined nucleic acid molecules are included within the second or third aspects of the invention.
  • a fusion protein that can be recognised by a commercially-available antibody.
  • a fusion protein may also be engineered to contain a cleavage site located between the sequence of the polypeptide of the invention and the sequence of a heterologous protein so that the polypeptide may be cleaved and purified away from the heterologous protein.
  • the nucleic acid molecules of the invention also include antisense molecules that are partially complementary to nucleic acid molecules encoding polypeptides of the present invention and that therefore hybridize to the encoding nucleic acid molecules (hybridization).
  • antisense molecules such as oligonucleotides, can be designed to recognise, specifically bind to and prevent transcription of a target nucleic acid encoding a polypeptide of the invention, as will be known by those of ordinary skill in the art (see, for example, Cohen, J.S., Trends in Pharm. Sci., 10, 435 (1989), Okano, J. Neurochem. 56, 560 (1991); O'Connor, J. Neurochem 56, 560 (1991); Lee et al, Nucleic Acids Res 6, 3073 (1979); Cooney et al, Science 241, 456 (1988); Dervan et al, Science 251, 1360 (1991).
  • hybridization refers to the association of two nucleic acid molecules with one another by hydrogen bonding. Typically, one molecule will be fixed to a solid support and the other will be free in solution. Then, the two molecules may be placed in contact with one another under conditions that favour hydrogen bonding. Factors that affect this bonding include: the type and volume of solvent; reaction temperature; time of hybridization; agitation; agents to block the non-specific attachment of the liquid phase molecule to the solid support (Denhardt's reagent or BLOTTO); the concentration of the molecules; use of compounds to increase the rate of association of molecules (dextran sulphate or polyethylene glycol); and the stringency of the washing conditions following hybridization (see Sambrook et al. [supra]).
  • the inhibition of hybridization of a completely complementary molecule to a target molecule may be examined using a hybridization assay, as known in the art (see, for example, Sambrook et al [supra]).
  • a substantially homologous molecule will then compete for and inhibit the binding of a completely homologous molecule to the target molecule under various conditions of stringency, as taught in Wahl, G.M. and S.L. Berger (1987; Methods Enzymol. 152:399-407) and Kimmel, A.R. (1987; Methods Enzymol. 152:507- 511).
  • Stringency refers to conditions in a hybridization reaction that favour the association of very similar molecules over association of molecules that differ.
  • High stringency hybridisation conditions are defined as overnight incubation at 42°C in a solution comprising 50% formamide, 5XSSC (150mM NaCl, 15mM trisodium citrate), 50mM sodium phosphate (pH7.6), 5x Denhardts solution, 10% dextran sulphate, and 20 microgram/ml denatured, sheared salmon sperm DNA, followed by washing the filters in 0.1X SSC at approximately 65°C.
  • Low stringency conditions involve the hybridisation reaction being carried out at 35°C (see Sambrook et al. [supra]).
  • the conditions used for hybridization are those of high stringency.
  • Preferred embodiments of this aspect of the invention -ire nucleic acid molecules that are at least 70% identical over their entire length to a nucleic acid molecule encoding the INPIONCH03 and INPIONCH04 polypeptides and nucleic acid molecules that are substantially complementary to such nucleic acid molecules.
  • a nucleic acid molecule according to this aspect of the invention comprises a region that is at least 80% identical over its entire length to such coding sequences, or is a nucleic acid molecule that is complementary thereto.
  • nucleic acid molecules at least 90%, preferably at least 95%, more preferably at least 98%, 99% or more identical over their entire length to the same are particularly preferred.
  • nucleic acid molecules that encode polypeptides which retain substantially the same biological function or activity as the INPIONCH03 and INPIONCH04 polypeptides are specifically excluded from the scope of this aspect of the invention.
  • the invention also provides a process for detecting a nucleic acid molecule of the invention, comprising the steps of: (a) contacting a nucleic probe according to the invention with a biological sample under hybridizing conditions to form duplexes; and (b) detecting any such duplexes that are formed.
  • a nucleic acid molecule as described above may be used as a hybridization probe for RNA, cDNA or genomic DNA, in order to isolate full-length cDNAs and genomic clones encoding the INPIONCH03 or INPIONCH04 polypeptides and to isolate cDNA and genomic clones of homologous or orthologous genes that have a high sequence similarity to the gene encoding this polypeptide.
  • the sequencing process may be automated using machines such as the Hamilton Micro Lab 2200 (Hamilton, Reno, NV), the Peltier Thermal Cycler (PTC200; MJ Research, Watertown, MA) and the ABI Catalyst and 373 and 377 DNA Sequencers (Perkin Elmer).
  • machines such as the Hamilton Micro Lab 2200 (Hamilton, Reno, NV), the Peltier Thermal Cycler (PTC200; MJ Research, Watertown, MA) and the ABI Catalyst and 373 and 377 DNA Sequencers (Perkin Elmer).
  • One method for isolating a nucleic acid molecule encoding a polypeptide with an equivalent function to that of the INPIONCH03 and INPIONCH04 polypeptides is to probe a genomic or cDNA library with a natural or artificially-designed probe using standard procedures that are recognised in the art (see, for example, "Current Protocols in Molecular Biology", Ausubel et al. (eds). Greene Publishing Association and John Wiley Interscience, New York, 1989,1992).
  • Probes comprising at least 15, preferably at least 30, and more preferably at least 50, contiguous bases that correspond to, or are complementary to, nucleic acid sequences from the appropriate encoding gene (SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l 1, SEQ ID NO-13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:27, SEQ ID NO:29, SEQ ID NO:31, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:37, SEQ ID NO:39, SEQ ID NO:41, SEQ ID NO:43, SEQ ID NO:45, SEQ ID NO:47, SEQ ID NO:49, SEQ ID NO-51, SEQ ID NO:53, SEQ ID NO:55, SEQ ID NO:57, SEQ ID NO:59, SEQ ID
  • Such probes may be labelled with an analytically-detectable reagent to facilitate their identification.
  • Useful reagents include, but are not limited to, radioisotopes, fluorescent dyes and enzymes that are capable of catalysing the formation of a detectable product.
  • the ordinarily skilled artisan will be capable of isolating complementary copies of genomic DNA, cDNA or RNA polynucleotides encoding proteins of interest from human, mammalian or other animal sources and screening such sources for related sequences, for example, for additional members of the family, type and/or subtype.
  • isolated cDNA sequences will be incomplete, in that the region encoding the polypeptide will be cut short, normally at the 5' end.
  • Several methods are available to obtain full length cDNAs, or to extend short cDNAs. Such sequences may be extended utilising a partial nucleotide sequence and employing various methods known in the art to detect upstream sequences such as promoters and regulatory elements. For example, one method which may be employed is based on the method of Rapid Amplification of cDNA Ends (RACE; see, for example, Frohman et al, PNAS USA 85, 8998-9002, 1988).
  • RACE Rapid Amplification of cDNA Ends
  • Another method which may be used is capture PCR which involves PCR amplification of DNA fragments adjacent a known sequence in human and yeast artificial chromosome DNA (Lagerstrom, M. et al. (1991) PCR Methods Applic, 1, 111-119). Another method which may be used to retrieve unknown sequences is that of Parker, J.D. et al. (1991); Nucleic Acids Res. 19:3055-3060). Additionally, one may use PCR, nested primers, and PromoterFinderTM libraries to walk genomic DNA (Clontech, Palo Alto, CA). This process avoids the need to screen libraries and is useful in finding intron/exon j unctions .
  • libraries that have been size- selected to include larger cDNAs.
  • random-primed libraries are preferable, in that they will contain more sequences that contain the 5' regions of genes. Use of a randomly primed library may be especially preferable for situations in which an oligo d(T) library does not yield a full-length cDNA.
  • Genomic libraries may be useful for extension of sequence into 5' non-transcribed regulatory regions.
  • the nucleic acid molecules of the present invention may be used for chromosome localisation.
  • a nucleic acid molecule is specifically targeted to, and can hybridize with, a particular location on an individual human chromosome.
  • the mapping of relevant sequences to chromosomes according to the present invention is an important step in the confirmatory correlation of those sequences with the gene-associated disease. Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. Such data are found in, for example, V. McKusick, Mendelian Inheritance in Man (available on-line through Johns Hopkins University Welch Medical Library).
  • the relationships between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance of physically adjacent genes). This provides valuable information to investigators searching for disease genes using positional cloning or other gene discovery techniques. Once the disease or syndrome has been crudely localised by genetic linkage to a particular genomic region, any sequences mapping to that area may represent associated or regulatory genes for further investigation.
  • the nucleic acid molecule may also be used to detect differences in the chromosomal location due to translocation, inversion, etc. among normal, carrier, or affected individuals.
  • the nucleic acid molecules of the present invention are also valuable for tissue localisation.
  • Such techniques allow the determination of expression patterns of the polypeptide in tissues by detection of the mRNAs that encode them.
  • These techniques include in situ hybridization techniques and nucleotide amplification techniques, such as PCR. Results from these studies provide an indication of the normal functions of the polypeptide in the organism.
  • comparative studies of the normal expression pattern of mRNAs with that of mRNAs encoded by a mutant gene provide valuable insights into the role of mutant polypeptides in disease. Such inappropriate expression may be of a temporal, spatial or quantitative nature.
  • RNA interference (Elbashir, SM et al, Nature 2001, 411, 494-498) is one method of sequence specific post- transcriptional gene silencing that may be employed. Short dsRNA oligonucleotides are synthesised in vitro and introduced into a cell. The sequence specific binding of these dsRNA oligonucleotides triggers the degradation of target mRNA, reducing or ablating target protein expression.
  • Efficacy of the gene silencing approaches assessed above may be assessed through the measurement of polypeptide expression (for example, by Western blotting), and at the RNA level using TaqMan-based methodologies.
  • the vectors of the present invention comprise nucleic acid molecules of the invention and may be cloning or expression vectors.
  • the host cells of the invention which may be transformed, transfected or transduced with the vectors of the invention may be prokaryotic or eukaryotic.
  • the polypeptides of the invention may be prepared in recombinant form by expression of their encoding nucleic acid molecules in vectors contained within a host cell. Such expression methods are well known to those of skill in the art and many are described in detail by Sambrook et al (supra) and Fernandez & Hoeffler (1998, eds. "Gene expression systems. Using nature for the art of expression”. Academic Press, San Diego, London, Boston, New York, Sydney, Tokyo, Toronto).
  • any system or vector that is suitable to maintain, propagate or express nucleic acid molecules to produce a polypeptide in the required host may be used.
  • the appropriate nucleotide sequence may be inserted into an expression system by any of a variety of well- known and routine techniques, such as, for example, those described in Sambrook et al, (supra).
  • the encoding gene can be placed under the control of a control element such as a promoter, ribosome binding site (for bacterial expression) and, optionally, an operator, so that the DNA sequence encoding the desired polypeptide is transcribed into RNA in the transformed host cell.
  • suitable expression systems include, for example, chromosomal, episomal and virus-derived systems, including, for example, vectors derived from: bacterial plasmids, bacteriophage, transposons, yeast episomes, insertion elements, yeast chromosomal elements, viruses such as baculoviruses, papova viruses such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, or combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, including cosmids and phagemids.
  • Human artificial chromosomes may also be employed to deliver larger fragments of DNA than can be contained and expressed in a plasmid.
  • Particularly suitable expression systems include microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (for example, baculovirus); plant cell systems transformed with virus expression vectors (for example, cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or with bacterial expression vectors (for example, Ti or pBR322 plasmids); or animal cell systems.
  • Cell-free translation systems can also be employed to produce the polypeptides of the invention.
  • nucleic acid molecules encoding a polypeptide of the present invention into host cells can be effected by methods described in many standard laboratory manuals, such as Davis et al, Basic Methods in Molecular Biology (1986) and Sambrook et al, (supra). Particularly suitable methods include calcium phosphate transfection, DEAE-dextran mediated transfection, transvection, microinjection, cationic lipid-mediated transfection, electroporation, transduction, scrape loading, ballistic introduction or infection (see Sambrook et al, 1989 [supra]; Ausubel et al, 1991 [supra]; Spector, Goldman & Leinwald, 1998). In eukaryotic cells, expression systems may either be transient (for example, episomal) or permanent (chromosomal integration) according to the needs of the system.
  • the encoding nucleic acid molecule may or may not include a sequence encoding a control sequence, such as a signal peptide or leader sequence, as desired, for example, for secretion of the translated polypeptide into the lumen of the endoplasmic reticulum, into the periplasmic space or into the extracellular environment.
  • a control sequence such as a signal peptide or leader sequence
  • These signals may be endogenous to the polypeptide or they may be heterologous signals.
  • Leader sequences can be removed by the bacterial host in post-translational processing.
  • regulatory sequences that allow for regulation of the expression of the polypeptide relative to the growth of the host cell.
  • regulatory sequences are those which cause the expression of a gene to be increased or decreased in response to a chemical or physical stimulus, including the presence of a regulatory compound or to various temperature or metabolic conditions.
  • Regulatory sequences are those non-translated regions of the vector, such as enhancers, promoters and 5' and 3' untranslated regions. These interact with host cellular proteins to carry out transcription and translation. Such regulatory sequences may vary in their strength and specificity. Depending on the vector system and host utilised, any number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used.
  • inducible promoters such as the hybrid lacZ promoter of the Bluescript phagemid (Stratagene, LaJolla, CA) or pSportlTM plasmid (Gibco BRL) and the like may be used.
  • the baculovirus polyhedrin promoter may be used in insect cells. Promoters or enhancers derived from the genomes of plant cells (for example, heat shock, RUBISCO and storage protein genes) or from plant viruses (for example, viral promoters or leader sequences) may be cloned into the vector. In mammalian cell systems, promoters from mammalian genes or from mammalian viruses are preferable. If it is necessary to generate a cell line that contains multiple copies of the sequence, vectors based on SV40 or EBV may be used with an appropriate selectable marker.
  • An expression vector is constructed so that the particular nucleic acid coding sequence is located in the vector with the appropriate regulatory sequences, the positioning and orientation of the coding sequence with respect to the regulatory sequences being such that the coding sequence is transcribed under the "control" of the regulatory sequences, i.e., RNA polymerase which binds to the DNA molecule at the control sequences transcribes the coding sequence.
  • control i.e., RNA polymerase which binds to the DNA molecule at the control sequences transcribes the coding sequence.
  • control sequences and other regulatory sequences may be ligated to the nucleic acid coding sequence prior to insertion into a vector.
  • the coding sequence can be cloned directly into an expression vector that already contains the control sequences and an appropriate restriction site.
  • cell lines which stably express the polypeptide of interest may be transformed using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells may be allowed to grow for 1-2 days in an enriched media before they are switched to selective media.
  • the purpose of the selectable marker is to confer resistance to selection, and its presence allows growth and recovery of cells that successfully express the introduced sequences.
  • Resistant clones of stably transformed cells may be proliferated using tissue culture techniques appropriate to the cell type.
  • Mammalian cell lines available as hosts for expression are known in the art and include many immortalised cell lines available from the American Type Culture Collection (ATCC) including, but not limited to, Chinese hamster ovary (CHO), HeLa, baby hamster kidney (BHK), monkey kidney (COS), C127, 3T3, BHK, HEK 293, Bowes melanoma and human hepatocellular carcinoma (for example Hep G2) cells and a number of other cell lines.
  • ATCC American Type Culture Collection
  • baculovirus/insect cell expression systems are commercially available in kit form from, inter alia, Invitrogen, San Diego CA (the "MaxBac” kit). These techniques are generally known to those skilled in the art and are described fully in Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987). Particularly suitable host cells for use in this system include insect cells such as Drosophila S2 and Spodoptera Sf9 cells. There are many plant cell culture and whole plant genetic expression systems known in the art. Examples of suitable plant cellular genetic expression systems include those described in US 5,693,506; US 5,659,122; and US 5,608,143. Additional examples of genetic expression in plant cell culture has been described by Zenk, Phytochemistry 30, 3861-3863 (1991).
  • all plants from which protoplasts can be isolated and cultured to give whole regenerated plants can be utilised, so that whole plants are recovered which contain the transferred gene.
  • Practically all plants can be regenerated from cultured cells or tissues, including but not limited to all major species of sugar cane, sugar beet, cotton, fruit and other trees, legumes and vegetables.
  • Examples of particularly prefe ⁇ ed bacterial host cells include streptococci, staphylococci, E. coli, Streptomyces and Bacillus subtilis cells.
  • Examples of particularly suitable host cells for fungal expression include yeast cells (for example, S. cerevisiae) and Aspergillus cells.
  • any number of selection systems are known in the art that may be used to recover transformed cell lines. Examples include the herpes simplex virus thymidine kinase (Wigler, M. et al. (1977) Cell 11:223-32) and adenine phosphoribosyltransferase (Lowy, I. et al. (1980) Cell 22:817-23) genes that can be employed in tk " or aprt* cells, respectively.
  • antimetabolite, antibiotic or herbicide resistance can be used as the basis for selection; for example, dihydrofolate reductase (DHFR) that confers resistance to methotrexate (Wigler, M. et al. (1980) Proc. Natl. Acad. Sci. 77:3567-70); npt, which confers resistance to the aminoglycosides neomycin and G-418 (Colbere-Garapin, F. et al (1981) J. Mol. Biol. 150:1-14) and als or pat, which confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively. Additional selectable genes have been described, examples of which will be clear to those of skill in the art.
  • marker gene expression suggests that the gene of interest is also present, its presence and expression may need to be confirmed.
  • a marker gene can be placed in tandem with a sequence encoding a polypeptide of the invention under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well.
  • host cells that contain a nucleic acid sequence encoding a polypeptide of the invention and which express said polypeptide may be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA- DNA or DNA-RNA hybridizations and protein bioassays, for example, fluorescence activated cell sorting (FACS) or immunoassay techniques (such as the enzyme-linked immunosorbent assay [ELISA] and radioimmunoassay [RIA]), that include membrane, solution, or chip based technologies for the detection and/or quantification of nucleic acid or protein (see Hampton, R. et al. (1990) Serological Methods, a Laboratory Manual, APS Press, St Paul, MN) and Maddox, D.E. et al. (1983) J. Exp. Med, 158, 1211-1216).
  • FACS fluorescence activated cell sorting
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • Means for producing labelled hybridization or PCR probes for detecting sequences related to nucleic acid molecules encoding polypeptides of the present invention include oligolabelling, nick translation, end-labelling or PCR amplification using a labelled polynucleotide.
  • Suitable reporter molecules or labels include radionuclides, enzymes and fluorescent, chemiluminescent or chromogenic agents as well as substrates, cofactors, inhibitors, magnetic particles, and the like.
  • Nucleic acid molecules according to the present invention may also be used to create transgenic animals, particularly rodent animals. Such transgenic animals form a further aspect of the present invention. This may be done locally by modification of somatic cells, or by germ line therapy to incorporate heritable modifications. Such transgenic animals may be particularly useful in the generation of animal models for drug molecules effective as modulators of the polypeptides of the present invention.
  • the polypeptide can be recovered and purified from recombinant cell cultures by well- known methods including ammonium sulphate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. High performance liquid chromatography is particularly useful for purification. Well known techniques for refolding proteins may be employed to regenerate an active conformation when the polypeptide is denatured during isolation and or purification.
  • Specialised vector constructions may also be used to facilitate purification of proteins, as desired, by joining sequences encoding the polypeptides of the invention to a nucleotide sequence encoding a polypeptide domain that will facilitate purification of soluble proteins.
  • purification-facilitating domains include metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilised metals, protein A domains that allow purification on immobilised immunoglobulin, and the domain utilised in the FLAGS extension/affinity purification system (Immunex Corp., Seattle, WA).
  • cleavable linker sequences such as those specific for Factor XA or enterokinase (Invitrogen, San Diego, CA) between the purification domain and the polypeptide of the invention may be used to facilitate purification.
  • One such expression vector provides for expression of a fusion protein containing the polypeptide of the invention fused to several histidine residues preceding a thioredoxin or an enterokinase cleavage site. The histidine residues facilitate purification by IMAC (immobilised metal ion affinity chromatography as described in Porath, J. et al. (1992), Prot. Exp. Purif.
  • the polypeptide is to be expressed for use in screening assays, generally it is preferred that it be produced at the surface of the host cell in which it is expressed. In this event, the host cells may be harvested prior to use in the screening assay, for example using techniques such as fluorescence activated cell sorting (FACS) or immunoaffinity techniques. If the polypeptide is secreted into the medium, the medium can be recovered in order to recover and purify the expressed polypeptide. If polypeptide is produced intracellularly, the cells must first be lysed before the polypeptide is recovered.
  • the polypeptide of the invention can be used to screen libraries of compounds in any of a variety of drug screening techniques.
  • Such compounds may activate (agonise) or inhibit (antagonise) the level of expression of the gene or the activity of the polypeptide of the invention and form a further aspect of the present invention.
  • Preferred compounds are effective to alter the expression of a natural gene which encodes a polypeptide of the first aspect of the invention or to regulate the activity of a polypeptide of the first aspect of the invention.
  • Agonist or antagonist compounds may be isolated from, for example, cells, cell-free preparations, chemical libraries or natural product mixtures. These agonists or antagonists may be natural or modified substrates, ligands, enzymes, receptors or structural or functional mimetics. For a suitable review of such screening techniques, see Coligan et al.
  • Compounds that are most likely to be good antagonists are molecules that bind to the polypeptide of the invention without inducing the biological effects of the polypeptide upon binding to it.
  • Potential antagonists include small organic molecules, peptides, polypeptides and antibodies that bind to the polypeptide of the invention and thereby inhibit or extinguish its activity. In this fashion, binding of the polypeptide to normal cellular binding molecules may be inhibited, such that the normal biological activity of the polypeptide is prevented.
  • the polypeptide of the invention that is employed in such a screening technique may be free in solution, affixed to a solid support, borne on a cell surface or located intracellularly.
  • screening procedures may involve using appropriate cells or cell membranes that express the polypeptide that are contacted with a test compound to observe binding, or stimulation or inhibition of a functional response.
  • the functional response of the cells contacted with the test compound is then compared with control cells that were not contacted with the test compound.
  • Such an assay may assess whether the test compound results in a signal generated by activation of the polypeptide, using an appropriate detection system.
  • Inhibitors of activation are generally assayed in the presence of a known agonist and the effect on activation by the agonist in the presence of the test compound is observed.
  • a preferred method for identifying an agonist or antagonist compound of a polypeptide of the present invention comprises: (a) contacting a cell expressing on the surface thereof the polypeptide according to the first aspect of the invention, the polypeptide being associated with a second component capable of providing a detectable signal in response to the binding of a compound to the polypeptide, with a compound to be screened under conditions to permit binding to the polypeptide; and
  • a further prefe ⁇ ed method for identifying an agonist or antagonist of a polypeptide of the invention comprises:
  • the general methods that are described above may further comprise conducting the identification of agonist or antagonist in the presence of labelled or unlabelled ligand for the polypeptide.
  • the method for identifying an agonist or antagonist of a polypeptide of the present invention comprises: determining the inhibition of binding of a ligand to cells which have a polypeptide of the invention on the surface thereof, or to cell membranes containing such a polypeptide, in the presence of a candidate compound under conditions to permit binding to the polypeptide, and determining the amount of ligand bound to the polypeptide.
  • a compound capable of causing reduction of binding of a ligand is considered to be an agonist or antagonist.
  • the ligand is labelled.
  • a method of screening for a polypeptide antagonist or agonist compound comprises the steps of:
  • step (a) incubating a labelled ligand with a whole cell expressing a polypeptide according to the invention on the cell surface, or a cell membrane containing a polypeptide of the invention, (b) measuring the amount of labelled ligand bound to the whole cell or the cell membrane; (c) adding a candidate compound to a mixture of labelled ligand and the whole cell or the cell membrane of step (a) and allowing the mixture to attain equilibrium;
  • step (d) measuring the amount of labelled ligand bound to the whole cell or the cell membrane after step (c); and (e) comparing the difference in the labelled ligand bound in step (b) and (d), such that the compound which causes the reduction in binding in step (d) is considered to be an agonist or antagonist.
  • simple binding assays may be used, in which the adherence of a test compound to a surface bearing the polypeptide is detected by means of a label directly or indirectly associated with the test compound or in an assay involving competition with a labelled competitor.
  • competitive drug screening assays may be used, in which neutralising antibodies that are capable of binding the polypeptide specifically compete with a test compound for binding. In this manner, the antibodies can be used to detect the presence of any test compound that possesses specific binding affinity for the polypeptide.
  • Assays may also be designed to detect the effect of added test compounds on the production of mRNA encoding the polypeptide in cells.
  • an ELISA may be constructed that measures secreted or cell-associated levels of polypeptide using monoclonal or polyclonal antibodies by standard methods known in the art, and this can be used to search for compounds that may inhibit or enhance the production of the polypeptide from suitably manipulated cells or tissues. The formation of binding complexes between the polypeptide and the compound being tested may then be measured.
  • Assay methods that are also included within the terms of the present invention are those that involve the use of the genes and polypeptides of the invention in overexpression or ablation assays. Such assays involve the manipulation of levels of these genes/polypeptides in cells and assessment of the impact of this manipulation event on the physiology of the manipulated cells. For example, such experiments reveal details of signalling and metabolic pathways in which the particular genes/polypeptides are implicated, generate information regarding the identities of polypeptides with which the studied polypeptides interact and provide clues as to methods by which related genes and proteins are regulated.
  • Another technique for drug screening which may be used provides for high throughput screening of compounds having suitable binding affinity to the polypeptide of interest (see International patent application WO84/03564).
  • This method large numbers of different small test compounds are synthesised on a solid substrate, which may then be reacted with the polypeptide of the invention and washed.
  • One way of immobilising the polypeptide is to use non-neutralising antibodies. Bound polypeptide may then be detected using methods that are well known in the art. Purified polypeptide can also be coated directly onto plates for use in the aforementioned drug screening techniques.
  • the polypeptide of the invention may be used to identify membrane-bound or soluble receptors, through standard receptor binding techniques that are known in the art, such as ligand binding and crosslinking assays in which the polypeptide is labelled with a radioactive isotope, is chemically modified, or is fused to a peptide sequence that facilitates its detection or purification, and incubated with a source of the putative receptor (for example, a composition of cells, cell membranes, cell supernatants, tissue extracts, or bodily fluids).
  • a source of the putative receptor for example, a composition of cells, cell membranes, cell supernatants, tissue extracts, or bodily fluids.
  • the efficacy of binding may be measured using biophysical techniques such as surface plasmon resonance (supplied by Biacore AB, Uppsala, Sweden) and spectroscopy.
  • Binding assays may be used for the purification and cloning of the receptor, but may also identify agonists and antagonists of the polypeptide, that compete with the binding of the polypeptide to its receptor. Standard methods for conducting screening assays are well understood in the art.
  • the invention also includes a screening kit useful in the methods for identifying agonists, antagonists, ligands, receptors, substrates, enzymes, that are described above.
  • the invention includes the agonists, antagonists, ligands, receptors, substrates and enzymes, and other compounds which modulate the activity or antigenicity of the polypeptide of the invention discovered by the methods that are described above.
  • the invention also provides pharmaceutical compositions comprising a polypeptide, nucleic acid, ligand or compound of the invention in combination with a suitable pharmaceutical carrier. These compositions may be suitable as therapeutic or diagnostic reagents, as vaccines, or as other immunogenic compositions, as outlined in detail below.
  • composition containing a polypeptide, nucleic acid, ligand or compound [X] is "substantially free of impurities [herein, Y] when at least
  • X comprises at least about 90% by weight of the total of X+Y in the composition, more preferably at least about 95%, 98% or even 99% by weight.
  • compositions should preferably comprise a therapeutically effective amount of the polypeptide, nucleic acid molecule, ligand, or compound of the invention.
  • therapeutically effective amount refers to an amount of a therapeutic agent needed to treat, ameliorate, or prevent a targeted disease or condition, or to exhibit a detectable therapeutic or preventative effect.
  • the therapeutically effective dose can be estimated initially either in cell culture assays, for example, of neoplastic cells, or in animal models, usually mice, rabbits, dogs, or pigs. The animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • an effective amount for a human subject will depend upon the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. This amount can be determined by routine experimentation and is within the judgement of the clinician. Generally, an effective dose will be from 0.01 mg/kg to 50 mg/kg, preferably 0.05 mg/kg to 10 mg/kg.
  • Compositions may be administered individually to a patient or may be administered in combination with other agents, drugs or hormones.
  • a pharmaceutical composition may also contain a pharmaceutically acceptable ca ⁇ ier, for administration of a therapeutic agent.
  • Such carriers include antibodies and other polypeptides, genes and other therapeutic agents such as liposomes, provided that the carrier does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity.
  • Suitable ca ⁇ iers may be large, slowly metabolised macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers and inactive virus particles.
  • Pharmaceutically acceptable salts can be used therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulphates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulphates, and the like
  • organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • Pharmaceutically acceptable carriers in therapeutic compositions may additionally contain liquids such as water, saline, glycerol and ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such compositions.
  • Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for ingestion by
  • compositions of the invention can be administered directly to the subject.
  • the subjects to be treated can be animals; in particular, human subjects can be treated.
  • the pharmaceutical compositions utilised in this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra- arterial, intramedullary, intrathecal, intraventricular, transdermal or transcutaneous applications (for example, see WO98/20734), subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, intravaginal or rectal means.
  • Gene guns or hyposprays may also be used to administer the pharmaceutical compositions of the invention.
  • the therapeutic compositions may be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared.
  • Direct delivery of the compositions will generally be accomplished by injection, subcutaneously, intraperitoneally, intravenously or intramuscularly, or delivered to the interstitial space of a tissue.
  • the compositions can also be administered into a lesion. Dosage treatment may be a single dose schedule or a multiple dose schedule.
  • One approach comprises administering to a subject an inhibitor compound (antagonist) as described above, along with a pharmaceutically acceptable carrier in an amount effective to inhibit the function of the polypeptide, such as by blocking the binding of ligands, substrates, enzymes, receptors, or by inhibiting a second signal, and thereby alleviating the abnormal condition.
  • an inhibitor compound as described above
  • a pharmaceutically acceptable carrier in an amount effective to inhibit the function of the polypeptide, such as by blocking the binding of ligands, substrates, enzymes, receptors, or by inhibiting a second signal, and thereby alleviating the abnormal condition.
  • antagonists are antibodies.
  • such antibodies are chimeric and/or humanised to minimise their immunogenicity, as described previously.
  • polypeptide that retain binding affinity for the ligand, substrate, enzyme, receptor, in question, may be administered.
  • polypeptide may be administered in the form of fragments that retain the relevant portions.
  • expression of the gene encoding the polypeptide can be inhibited using expression blocking techniques, such as the use of antisense nucleic acid molecules (as described above), either internally generated or separately administered.
  • Modifications of gene expression can be obtained by designing complementary sequences or antisense molecules (DNA, RNA, or PNA) to the control, 5' or regulatory regions (signal sequence, promoters, enhancers and introns) of the gene encoding the polypeptide.
  • inhibition can be achieved using "triple helix" base-pairing methodology. Triple helix pairing is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or regulatory molecules.
  • the complementary sequence or antisense molecule may also be designed to block translation of mRNA by preventing the transcript from binding to ribosomes.
  • Such oligonucleotides may be administered or may be generated in situ from expression in vivo.
  • Ribozymes are catalytically active RNAs that can be natural or synthetic (see for example Usman, N, et al, Curr. Opin. Struct. Biol (1996) 6(4), 527-33). Synthetic ribozymes can be designed to specifically cleave mRNAs at selected positions thereby preventing translation of the mRNAs into functional polypeptide. Ribozymes may be synthesised with a natural ribose phosphate backbone and natural bases, as normally found in RNA molecules. Alternatively the ribozymes may be synthesised with non-natural backbones, for example, 2'-O-methyl RNA, to provide protection from ribonuclease degradation and may contain modified bases.
  • RNA molecules may be modified to increase intracellular stability and half-life. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5' and/or 3' ends of the molecule or the use of phosphorothioate or 2' O-methyl rather than phosphodiesterase linkages within the backbone of the molecule. This concept is inherent in the production of PNAs and can be extended in all of these molecules by the inclusion of non-traditional bases such as inosine, queosine and butosine, as well as acetyl-, methyl-, thio- and similarly modified forms of adenine, cytidine, guanine, thymine and uridine which are not as easily recognised by endogenous endonucleases.
  • One approach comprises administering to a subject a therapeutically effective amount of a compound that activates the polypeptide, i.e., an agonist as described above, to alleviate the abnormal condition.
  • a therapeutic amount of the polypeptide in combination with a suitable pharmaceutical carrier may be administered to restore the relevant physiological balance of polypeptide.
  • Gene therapy may be employed to effect the endogenous production of the polypeptide by the relevant cells in the subject. Gene therapy is used to treat permanently the inappropriate production of the polypeptide by replacing a defective gene with a co ⁇ ected therapeutic gene.
  • Gene therapy of the present invention can occur in vivo or ex vivo.
  • Ex vivo gene therapy requires the isolation and purification of patient cells, the introduction of a therapeutic gene and introduction of the genetically altered cells back into the patient.
  • in vivo gene therapy does not require isolation and purification of a patient's cells.
  • the therapeutic gene is typically "packaged" for administration to a patient.
  • Gene delivery vehicles may be non-viral, such as liposomes, or replication-deficient viruses, such as adenovirus as described by Berkner, K.L., in Cu ⁇ . Top. Microbiol. Immunol., 158, 39-66 (1992) or adeno-associated virus (AAV) vectors as described by Muzyczka, N., in Cu ⁇ . Top. Microbiol. Immunol., 158, 97-129 (1992) and U.S. Patent No. 5,252,479.
  • a nucleic acid molecule encoding a polypeptide of the invention may be engineered for expression in a replication-defective retroviral vector.
  • This expression construct may then be isolated and introduced into a packaging cell transduced with a retroviral plasmid vector containing RNA encoding the polypeptide, such that the packaging cell now produces infectious viral particles containing the gene of interest.
  • These producer cells may be administered to a subject for engineering cells in vivo and expression of the polypeptide in vivo (see Chapter 20, Gene Therapy and other Molecular Genetic-based Therapeutic Approaches, (and references cited therein) in Human Molecular Genetics (1996), T Strachan and A P Read, BIOS Scientific Publishers Ltd).
  • Another approach is the administration of "naked DNA" in which the therapeutic gene is directly injected into the bloodstream or muscle tissue.
  • the invention provides that they can be used in vaccines to raise antibodies against the disease causing agent.
  • Vaccines according to the invention may either be prophylactic (ie. to prevent infection) or therapeutic (ie. to treat disease after infection).
  • Such vaccines comprise immunising antigen(s), immunogen(s), polypeptide(s), protein(s) or nucleic acid, usually in combination with pharmaceutically-acceptable carriers as described above, which include any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition. Additionally, these carriers may function as immunostimulating agents ("adjuvants").
  • the antigen or immunogen may be conjugated to a bacterial toxoid, such as a toxoid from diphtheria, tetanus, cholera, H. pylori, and other pathogens.
  • vaccines comprising polypeptides are preferably administered parenterally (for instance, subcutaneous, intramuscular, intravenous, or intradermal injection).
  • parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti- oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the recipient, and aqueous and non-aqueous sterile suspensions which may include suspending agents or thickening agents.
  • the vaccine formulations of the invention may be presented in unit-dose or multi-dose containers.
  • sealed ampoules and vials and may be stored in a freeze-dried condition requiring only the addition of the sterile liquid carrier immediately prior to use.
  • the dosage will depend on the specific activity of the vaccine and can be readily determined by routine experimentation.
  • jet injection may also be useful in the formulation of vaccine compositions.
  • This invention also relates to the use of nucleic acid molecules according to the present invention as diagnostic reagents. Detection of a mutated form of the gene characterised by the nucleic acid molecules of the invention which is associated with a dysfunction will provide a diagnostic tool that can add to, or define, a diagnosis of a disease, or susceptibility to a disease, which results from under-expression, over-expression or altered spatial or temporal expression of the gene. Individuals carrying mutations in the gene may be detected at the DNA level by a variety of techniques.
  • Nucleic acid molecules for diagnosis may be obtained from a subject's cells, such as from blood, urine, saliva, tissue biopsy or autopsy material.
  • the genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR, ligase chain reaction (LCR), strand displacement amplification (SDA), or other amplification techniques (see Saiki et al, Nature, 324, 163-166 (1986); Bej, et al, Crit. Rev. Biochem. Molec. Biol., 26, 301-334 (1991); Birkenmeyer et al, J. Virol. Meth., 35, 117-126 (1991); Van Brunt, J., Bio/Technology, 8, 291-294 (1990)) prior to analysis.
  • LCR ligase chain reaction
  • SDA strand displacement amplification
  • this aspect of the invention provides a method of diagnosing a disease in a patient, comprising assessing the level of expression of a natural gene encoding a polypeptide according to the invention and comparing said level of expression to a control level, wherein a level that is different to said control level is indicative of disease.
  • the method may comprise the steps of: a)contacting a sample of tissue from the patient with a nucleic acid probe under stringent conditions that allow the formation of a hybrid complex between a nucleic acid molecule of the invention and the probe; b)contacting a control sample with said probe under the same conditions used in step a); c)and detecting the presence of hybrid complexes in said samples; wherein detection of levels of the hybrid complex in the patient sample that differ from levels of the hybrid complex in the control sample is indicative of disease.
  • a further aspect of the invention comprises a diagnostic method comprising the steps of: a)obtaining a tissue sample from a patient being tested for disease; b)isolating a nucleic acid molecule according to the invention from said tissue sample; and c)diagnosing the patient for disease by detecting the presence of a mutation in the nucleic acid molecule which is associated with disease.
  • an amplification step for example using PCR, may be included. Deletions and insertions can be detected by a change in the size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to labelled RNA of the invention or alternatively, labelled antisense DNA sequences of the invention. Perfectly-matched sequences can be distinguished from mismatched duplexes by RNase digestion or by assessing differences in melting temperatures.
  • the presence or absence of the mutation in the patient may be detected by contacting DNA with a nucleic acid probe that hybridises to the DNA under stringent conditions to form a hybrid double-stranded molecule, the hybrid double-stranded molecule having an unhybridised portion of the nucleic acid probe strand at any portion co ⁇ esponding to a mutation associated with disease; and detecting the presence or absence of an unhybridised portion of the probe strand as an indication of the presence or absence of a disease-associated mutation in the co ⁇ esponding portion of the DNA strand.
  • Such diagnostics are particularly useful for prenatal and even neonatal testing.
  • Point mutations and other sequence differences between the reference gene and "mutant" genes can be identified by other well-known techniques, such as direct DNA sequencing or single-strand conformational polymorphism, (see Orita et al, Genomics, 5, 874-879 (1989)).
  • a sequencing primer may be used with double-stranded PCR product or a single-stranded template molecule generated by a modified PCR.
  • the sequence determination is performed by conventional procedures with radiolabelled nucleotides or by automatic sequencing procedures with fluorescent-tags.
  • Cloned DNA segments may also be used as probes to detect specific DNA segments. The sensitivity of this method is greatly enhanced when combined with PCR.
  • point mutations and other sequence variations can be detected as described above, for example, through the use of allele-specific oligonucleotides for PCR amplification of sequences that differ by single nucleotides.
  • DNA sequence differences may also be detected by alterations in the electrophoretic mobility of DNA fragments in gels, with or without denaturing agents, or by direct DNA sequencing (for example, Myers et al, Science (1985) 230:1242). Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and SI protection or the chemical cleavage method (see Cotton et al, Proc. Natl. Acad. Sci. USA (1985) 85: 4397-4401).
  • mutations such as microdeletions, aneuploidies, translocations, inversions, can also be detected by in situ analysis (see, for example, Keller et al, DNA Probes, 2nd Ed., Stockton Press, New York, N.Y., USA (1993)), that is, DNA or RNA sequences in cells can be analysed for mutations without need for their isolation and/or immobilisation onto a membrane.
  • Fluorescence in situ hybridization is presently the most commonly applied method and numerous reviews of FISH have appeared (see, for example, Trachuck et al, Science, 250, 559-562 (1990), and Trask et al, Trends, Genet., 7, 149-154 (1991)).
  • an array of oligonucleotide probes comprising a nucleic acid molecule according to the invention can be constructed to conduct efficient screening of genetic variants, mutations and polymorphisms.
  • Array technology methods are well known and have general applicability and can be used to address a variety of questions in molecular genetics including gene expression, genetic linkage, and genetic variability (see for example: M.Chee et al, Science (1996), Vol 274, pp 610-613).
  • the a ⁇ ay is prepared and used according to the methods described in PCT application WO95/11995 (Chee et al); Lockhart, D. J. et al. (1996) Nat. Biotech. 14: 1675-1680); and Schena, M. et al. (1996) Proc. Natl. Acad. Sci. 93: 10614-10619).
  • Oligonucleotide pairs may range from two to over one million.
  • the oligomers are synthesized at designated areas on a substrate using a light-directed chemical process.
  • the substrate may be paper, nylon or other type of membrane, filter, chip, glass slide or any other suitable solid support.
  • an oligonucleotide may be synthesized on the surface of the substrate by using a chemical coupling procedure and an ink jet application apparatus, as described in PCT application W095/251116 (Baldeschweiler et at).
  • a "gridded" a ⁇ ay analogous to a dot (or slot) blot may be used to arrange and link cDNA fragments or oligonucleotides to the surface of a substrate using a vacuum system, thermal, UV, mechanical or chemical bonding procedures.
  • An a ⁇ ay such as those described above, may be produced by hand or by using available devices (slot blot or dot blot apparatus), materials (any suitable solid support), and machines (including robotic instruments), and may contain 8, 24, 96, 384, 1536 or 6144 oligonucleotides, or any other number between two and over one million which lends itself to the efficient use of commercially-available instrumentation.
  • diseases may be diagnosed by methods comprising determining, from a sample derived from a subject, an abnormally decreased or increased level of polypeptide or mRNA. Decreased or increased expression can be measured at the RNA level using any of the methods well known in the art for the quantitation of polynucleotides, such as, for example, nucleic acid amplification, for instance PCR, RT-PCR, RNase protection, Northern blotting and other hybridization methods.
  • nucleic acid amplification for instance PCR, RT-PCR, RNase protection, Northern blotting and other hybridization methods.
  • Assay techmques that can be used to determine levels of a polypeptide of the present invention in a sample derived from a host are well-known to those of skill in the art and are discussed in some detail above (including radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays).
  • This aspect of the invention provides a diagnostic method which comprises the steps of: (a) contacting a ligand as described above with a biological sample under conditions suitable for the formation of a ligand- polypeptide complex; and (b) detecting said complex.
  • Protocols such as ELISA, RIA, and FACS for measuring polypeptide levels may additionally provide a basis for diagnosing altered or abnormal levels of polypeptide expression.
  • Normal or standard values for polypeptide expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, preferably humans, with antibody to the polypeptide under conditions suitable for complex formation The amount of standard complex formation may be quantified by various methods, such as by photometric means.
  • Antibodies which specifically bind to a polypeptide of the invention may be used for the diagnosis of conditions or diseases characterised by expression of the polypeptide, or in assays to monitor patients being treated with the polypeptides, nucleic acid molecules, ligands and other compounds of the invention.
  • Antibodies useful for diagnostic purposes may be prepared in the same manner as those described above for therapeutics. Diagnostic assays for the polypeptide include methods that utilise the antibody and a label to detect the polypeptide in human body fluids or extracts of cells or tissues.
  • the antibodies may be used with or without modification, and may be labelled by joining them, either covalently or non-covalently, with a reporter molecule.
  • a wide variety of reporter molecules known in the art may be used, several of which are described above.
  • a diagnostic kit of the present invention may comprise: (a) a nucleic acid molecule of the present invention
  • a diagnostic kit may comprise a first container containing a nucleic acid probe that hybridises under stringent conditions with a nucleic acid molecule according to the invention; a second container containing primers useful for amplifying the nucleic acid molecule; and instructions for using the probe and primers for facilitating the diagnosis of disease.
  • the kit may further comprise a third container holding an agent for digesting unhybridised RNA.
  • a diagnostic kit may comprise an a ⁇ ay of nucleic acid molecules, at least one of which may be a nucleic acid molecule according to the invention.
  • kits may comprise one or more antibodies that bind to a polypeptide according to the invention; and a reagent useful for the detection of a binding reaction between the antibody and the polypeptide.
  • a disease or susceptibility to disease in which ion channels are implicated particularly cardiovascular disease, hypertension, heart arrhythmia, angina, myocardial infarction, tachycardia cerebrovascular disease, thromboembolism, neurological and psychiatric disorders, including stroke, epilepsy, migraine, pain, depression, neuralgia, Alzheimer's disease, brain injury, schizophrenia, Himtington's disease, motor neurone disease, Parkinson's disease, neuropathy, neuromuscular disorder, micturition disorder, impotence, diabetes, dermatitis, pulmonary disease, asthma, cystic fibrosis, mucous membrane disorders including sinusitis, COPD, rhinitis and cough, leukemia, ocular disease, glaucoma
  • the diseases are those in which members of the PKD/REJ family of cation channels are implicated.
  • diseases include renal disease, polycystic kidney disease, glomerulnephritis, fertility disorders, infertility, reproductive disorders, polycystic ovary disease, polycystic diseases, endocrine diseases, testosterone deficiency, testosterone-related disorder, testicular cancer, prostate cancer, gastrointestinal disorders, developmental disorders, brain tumour, sexual dysfunction, psychological disorder, alopecia, hirsuitism, diseases related to muscle tone, including vascular muscle disorders such as migraine, pre-eclampsia, hypertension and cardiac a ⁇ hythmias; bladder muscle tone disorders, such as urinary incontinence, inability to void, urinary tract infection; gastrointestinal smooth muscle tone disorder, such as Gl motility disorder, gastrointestinal reflux diorder (GERD), reflux oesophagitis, paralytic ileus, irritable bowel syndrome (IBS), dia ⁇ hoea
  • Figure 1 PKD/REJ cation channel family domain/topology diagram highlighting the conserved features.
  • Figure 2 Top ten results from BLAST against NCBI non-redundant database using SEQ ID NO:56 (INPIONCH03 polypeptide sequence).
  • Figure 3 Alignment generated by BLAST between SEQ ID NO:56 (INPIONCH03 polypeptide sequence) and the top biochemically annotated hit, the receptor for egg jelly 3 protein (REJ3).
  • Figure 4 conserved domain search using the INPIONCH03 query sequence.
  • Figure 5 INPIONCH03 topology prediction using the TMHMM transmembrane domain prediction programme
  • Figure 6 Top ten results from BLAST against NCBI non-redundant database using SEQ ID NO: 108 (INPIONCH04 polypeptide sequence).
  • Figure 7 Alignment generated by BLAST between SEQ ID NO: 108 (INPIONCH04 polypeptide sequence) and the top biochemically annotated hit, the receptor for egg jelly 3 protein (REJ3).
  • Figure 10 Normalised expression of INPIONCH03 in 22 samples derived from 18 normal human tissues.
  • Figure 11 Normalised expression of INPIONCH04 in 22 samples derived from 18 normal human tissues.
  • the INPIONCH03 polypeptide sequence shown in SEQ ID NO:56, was used as a BLAST query against the NCBI non-redundant Sequence database. Several of the top ten matches are members of the PKD/REJ family of cation channels. The top ten hits all align to the query sequence with highly significant E-values (8e "39 to 0.0) ( Figure 2). Figure 3 shows the alignment of the INPIONCH03 polypeptide query sequence to the sequence of the top biochemically annotated hit, the Strongylocentrotus purpuratus receptor for egg jelly 3 protein (REJ3). These results indicate that the INPIONCH03 polypeptide is a member of the PKD/REJ family of cation channels.
  • the INPIONCH03 polypeptide sequence (SEQ ID NO: 56) was analysed against the amino acid sequences of conserved domain in the Pfam and SMART public domain databases.
  • Figure 4 shows the results of this conserved domain search.
  • the INPIONCH03 query sequence returns matches to the PLAT (or LH2) domain, an ion transport-associated domain and the latrophilin/CL-1 like GPS domain.
  • the results of the conserved domain search therefore confirm that the INPIONCH03 polypeptide is a member of the PKD/REJ family of cation channels, as shown in Example 1 above.
  • Example 3 INPIONCH03 Topology Information from TMHMM Prediction Program
  • the INPIONCH03 polypeptide sequence (SEQ ID NO: 56) was analysed using the TMHMM (transmembrane hidden Markov model) programme for prediction of transmembrane domains.
  • the TMHMM programme predicts over 75% of transmembrane topologies co ⁇ ectly and is therefore a reliable tool for investigating protein topology (Krogh et al, Journal of Molecular Biology, 2001, 305(3):567-580).
  • the results of the TMHMM analysis of INPIONCH03 are shown in Figure 5.
  • Figure 5 shows that the TMHMM confidently predicts that the INPIONCH03 polypeptide contains eleven transmembrane domains, in accordance with other members of the PKD/REJ family of cation channels.
  • this topology prediction therefore provides further evidence that the INPIONCH03 polypeptide is a member of the PKD/REJ family of cation channels.
  • the INPIONCH04 polypeptide sequence shown in SEQ ID NO: 108, was used as a BLAST query against the NCBI non-redundant Sequence database. Several of the top ten matches are members of the PKD/REJ family of cation channels. The top ten hits all align to the query sequence with highly significant E-values (2e ' to e " ) ( Figure 6). Figure 7 shows the alignment of the INPIONCH04 polypeptide query sequence to the sequence of the top biochemically annotated hit, the Strongylocentrotus purpuratus receptor for egg jelly 3 protein (REJ3). These results indicate that the INPIONCH04 polypeptide is a member of the PKD/REJ family of cation channels.
  • REJ3 Strongylocentrotus purpuratus receptor for egg jelly 3 protein
  • the INPIONCH04 polypeptide sequence (SEQ ID NO: 108) was analysed against the amino acid sequences of conserved domain in the Pfam and SMART public domain databases.
  • Figure 8 shows the results of this conserved domain search.
  • the INPIONCH04 query sequence returns matches to the PLAT (or LH2) domain, an ion transport-associated domain and the latrophilin/CL-1 like GPS domain.
  • the results of the conserved domain search therefore confirm that the INPIONCH04 polypeptide is a member of the PKD/REJ family of cation channels, as shown in Example 4 above.
  • the INPIONCH04 polypeptide sequence (SEQ ID NO: 108) was analysed using the TMHMM (transmembrane hidden Markov model) programme for prediction of transmembrane domains.
  • the TMHMM programme predicts over 75% of transmembrane topologies co ⁇ ectly and is therefore a reliable tool for investigating protein topology (Krogh et al., Journal of Molecular Biology, 2001, 3O5(3):567-580).
  • the results of the TMHMM analysis of INPIONCH04 are shown in Figure 9.
  • Figure 9 shows that the TMHMM confidently predicts that the INPIONCH04 polypeptide contains eleven transmembrane domains, in accordance with other members of the PKD/REJ family of cation channels. In combination with the BLAST results described in Example 4, and the conserved domain search results described in Example 5, this topology prediction therefore provides further evidence that the INPIONCH04 polypeptide is a member of the PKD/REJ family of cation channels.
  • Taqman RT- PCR quantitation was used.
  • the TaqMan 3'- 5' exonuclease assay signals the formation of PCR amplicons by a process involving the nucleolytic degradation of a double labelled fluorogenic probe that hybridises to the target template at a site between the two primer recognition sequences (cf. U. S. Patent 5,876,930).
  • the ABI Prism 7700 automates the detection and quantitative measurement of these signals, which are stoichiometrically related to the quantities of amplicons produced, during each cycle of amplification.
  • this technology permits simplified and potentially highly accurate quantification of target sequences in the reactions.
  • Figure 10 shows normalised expression of INPIONCH03 in 22 samples derived from 18 normal human tissues.
  • Figure 11 shows normalised expression of INPIONCH04 in 22 samples derived from 18 normal human tissues.
  • Taqman RT-PCR was carried out using 15ng of the indicated cDNA using primers/probes specific for INPIONCH03 and 18s rRNA as described in the detailed description.
  • a standard curve for target and internal control was also carried out, using between 25ng to 0.39ng of cDNA template of a typical tissue sample.
  • Cycle threshold (Ct) determinations i.e. non-integer calculations of the number of cycles required for reporter dye fluorescence resulting from the synthesis of PCR products to become significantly higher than background fluorescence levels were performed by the instrument for each reaction using default parameters.
  • Ct Cycle threshold
  • the levels of target cDNA in each sample were normalised to the level of expression of target in a comparative sample, in the case of INPIONCH03, heart and in the case of
  • INPIONCH04 testis.
  • the levels of 18s cDNA in each sample were also normalised to the level of expression of 18s in heart and testis, respectively.
  • INPIONCH03 and INPIONCH04 were then normalised to the expression levels of 18s.
  • Figures 10 and 11 represent the fold expression of normalised target sequence relative to the level of expression in heart or testis cDNA, which is set arbitrarily to 1. Each sample was quantitated in 2 individual experiments. Figures 10 and 11 show the mean ⁇ SEM for the multiple experiments.
  • the primer/probe set recognises a sequence spanning a proposed exon-exon boundary within the INPIONCH03 coding sequence.
  • the PCR reaction was also carried out on cDNA made in the absence of reverse transcriptase enzyme. No signal was seen in these reactions (data not shown), indicating that results shown in Figure 10 are not affected by genomic contamination of the RNA sample and that the region of INPIONCH03 detected is present in cDNA.
  • the primer/probe set recognises a sequence within a proposed exon within the INPIONCH04 coding sequence.
  • the PCR reaction was also carried out on cDNA made in the absence of reverse transcriptase enzyme. A signal was seen in these reactions (data not shown), this was quantified and subtracted from the quantity calculated from the signal seen with cDNA synthesized in the presence of reverse transcriptase.
  • the results shown in Figure 11 are co ⁇ ected for genomic contamination of the RNA sample and indicate that the region of INPIONCH04 detected is present in cDNA.
  • RNA prepared from non-diseased organs was purchased from either Ambion Europe (Huntingdon, UK) or Clontech (BD, Franklin Lakes, NJ).
  • Oligonucleotide primers and probes were designed using Primer Express software (Applied Biosystems, Foster City CA) with a GC-content of 40-60%, no G-nucleotide at the 5'-end of the probe, and no more than 4 contiguous Gs.
  • INPIONCH03 Probe CGCTTGCCCTGCACCACCATGTA
  • INPIONCH04 Probe TGCAAAGAACTCCAATAGATTTTACCTCCACCA
  • TAMRA fluorescent quencher dye
  • Mem 582nm
  • Primers were obtained from Sigma Genosys, UK and probes were obtained from Eurogentec, Belgium.
  • Primer/probe concentrations were titrated in the range of 50nM to 900nM and optimal concentrations for efficient PCR reactions were determined.
  • Optimal primer and probe concentrations vary in between 1 OOnM and 900nM depending on the target gene that is amplified.
  • cDNA was prepared using components from Applied Biosystems, Foster City CA. 50 ⁇ l reactions were prepared in 0.5ml RNase free tubes. Reactions contain 500ng total RNA; lx reverse transcriptase buffer; 5.5mM MgC12; ImM dNTP's; 2.5 ⁇ l random hexamers; 20U RNase inhibitor; and 62.5U reverse transcriptase.
  • reaction were prepared in 0.5 ml thin-walled, optical grade PCR 96 well plates (Applied Biosystems, Foster City CA). Reactions contained: lx final concentration of TaqMan Universal Master Mix (a proprietary mixture of AmpliTaq Gold DNA polymerase, AmpEraseX UNG, dNTPs with UTP, passive reference dye and optimised buffer components, Applied Biosystems, Foster City CA); lOOnM Taqman probe; 900nM forward primer; 900nM reverse primer and 15ng of cDNA template.
  • TaqMan Universal Master Mix a proprietary mixture of AmpliTaq Gold DNA polymerase, AmpEraseX UNG, dNTPs with UTP, passive reference dye and optimised buffer components, Applied Biosystems, Foster City CA
  • lOOnM Taqman probe 900nM forward primer
  • 900nM reverse primer 15ng of cDNA template.
  • Standard procedures for the operation of the ABI Prism 7700 or similar detection system were used. This includes, for example with the ABI Prism 7700, use of all default program settings with the exception of reaction volume which is changed from 50 to 25 ul.
  • Thermal cycling conditions consist of two min at 50°C, 10 min at 95°C, followed by 40 cycles of 15 sec at 95°C and 1 min at 60 C.
  • Cycle threshold (Ct) determinations i.e. non-integer calculations of the number of cycles required for reporter dye fluorescence resulting from the synthesis of PCR products to become significantly higher than background fluorescence levels were automatically performed by the instrument for each reaction using default parameters.
  • Assays for target sequences and ribosomal 18s (reference) sequences in the same cDNA samples were performed in separate reaction tubes. Within each experiment, a standard curve was carried out on a typical tissue sample, from 25ng to 0.39ng of cDNA template. From this standard curve, the amount of actual starting target or 18s cDNA in each test sample was determined.
  • the levels of target cDNA in each sample were normalised to the level of expression of target in a comparative sample.
  • the levels of internal control cDNA in each sample were normalised to the level of expression of internal control in a comparative sample.
  • the data was then represented as fold expression of normalised target sequence relative to the level of expression in the comparative sample, which is set arbitrarily to 1.
  • the data provided in this example show that the INPIONCH03 and INPIONCH04 polypeptides are expressed in a variety of human tissues. High levels of INPIONCH03 are expressed in brain, cervix, placenta, bladder, testis, small intestine and heart. The INPIONCH04 protein shows widespread expression, with highest levels in testis and high levels also found in brain, skeletal muscle, ovary, lung and adrenal tissues. Experiments will now be undertaken to verify the full-length transcripts of the INPIONCH03 and INPIONCH04 polypeptides.

Abstract

This invention relates to novel proteins, termed INPIONCH03 and INPIONCH04, herein identified as members of the PKD/REJ family of cation channels and to the use of these proteins and nucleic acid sequences from the encoding genes in the diagnosis, prevention and treatment of disease.

Description

Cation Channel Proteins
This invention relates to novel proteins, termed INPIONCH03 and INPIONCH04, herein identified as members of the PKD/REJ family of cation channels and to the use of these proteins and nucleic acid sequences from the encoding genes in the diagnosis, prevention and treatment of disease.
All publications, patents and patent applications cited herein are incorporated in full by reference.
Background
The process of drug discovery is presently undergoing a fundamental revolution as the era of functional genomics comes of age. The term "functional genomics" applies to an approach utilising bioinformatics tools to ascribe function to protein sequences of interest. Such tools are becoming increasingly necessary as the speed of generation of sequence data is rapidly outpacing the ability of research laboratories to assign functions to these protein sequences. As bioinformatics tools increase in potency and in accuracy, these tools are rapidly replacing the conventional techniques of biochemical characterisation. Indeed, the advanced bioinformatics tools used in identifying the present invention are now capable of outputting results in which a high degree of confidence can be placed.
Various institutions and commercial organisations are examining sequence data as they become available and significant discoveries are being made on an on-going basis. However, there remains a continuing need to identify and characterise further genes and the polypeptides that they encode, as targets for research and for drug discovery.
Introduction to the PKD/REJ Family of Cation Channels
The PKD/REJ (polycystic kidney disease-receptor for egg jelly) family is small family of modular proteins whose members possess a characteristic combination of structural and functional features. A generic topology for members of the PKD/REJ family of cation channels is shown in Figure 1. The characteristic structural and functional features of the PKD/REJ family include at the N-terminus a REJ (sea urchin receptor for egg jelly)-like domain and a GPS domain (GPCR proteolytic site), followed by eleven transmembrane domains, a cytoplasmic PLAT (or LH2) domain (polycystin-1 lipoxygenase, alpha-toxin or lipoxygenase homology domain; a lipid binding site) and an ion-transfer domain (similar to those found in cationic ion channels). The different members of the family have varied sequences at the extreme N-terminus and C-terminus. The known members of the PKD/REJ family are summarised below in Table 1.
Table 1: Members of the PKD/REJ Family of Cation Channels
Figure imgf000003_0001
Figure imgf000004_0001
Figure imgf000005_0001
Ion channels may be useful for the treatment, prevention and/or diagnosis of medical conditions and diseases which include, but are not limited to, cardiovascular disease, hypertension, heart arrhythmia, angina, myocardial infarction, tachycardia, cerebrovascular disease, thromboembolism, neurological and psychiatric disorders, including stroke, epilepsy, migraine, pain, depression, neuralgia, Alzheimer's disease, brain injury, schizophrenia, Huntington's disease, motor neurone disease, Parkinson's disease, neuropathy, neuromuscular disorder, micturition disorder, impotence, diabetes, dermatitis, pulmonary disease, asthma, cystic fϊbrosis, mucous membrane disorders including sinusitis, COPD, rhinitis and cough, leukemia, ocular disease, glaucoma, retinopathy and immune disorders.
Diseases linked to the PKD/REJ family of Cation channels
Polycystic kidney disease
Mutations to the PKD1 and PKD2 genes account for approximately 95% of autosomal dominant polycystic kidney disease (ADPKD) cases (Nomura et al, J. Biol. Chem., 1998, Vol. 273, No. 40, pp 25967-25973). ADPKD is a very common monogenic disease, with an estimated frequency of occurrence of 1 in 1000 births, and is characterised by progressive cystic development in the kidney and eventual renal failure.
Reproductive health Members of the PKD/REJ family of cation channels, as well as playing a central role in polycystic kidney disease, are also implicated in reproductive health due to the presence of the REJ-like domain. The acrosome reaction, which occurs as the sperm penetrates the egg during fertilisation and is triggered by the suREJ protein in sea urchins, is characterised by various signal transduction events including Ca2+, Na+, H+ and K+ transportation, cyclic nucleotide changes and protein kinase C activation. The suREJ protein is thought to be involved in the initial Ca2+ influx that triggers the acrosome reaction. The sequence similarity and expression pattern of the PKDREJ protein suggests it is a mammalian equivalent of the suREJ protein and therefore may have a central role in human fertilisation via an involvement in the acrosome reaction. Since the acrosome reaction is required for animal fertilisation it offers the potential for the development of novel methods of non-hormonal contraception. The precise steps involved in the initiation of the acrosome reaction in animals have not yet been determined (Hughes et al, Hum. Mol. Genet., 1999, 8(3):543-9). Modulation of the acrosome reaction in humans would be of great importance in reproductive health, since it would allow new forms of infertility remedies or contraceptive methods to be devised.
PKD2L2 is also believed to play a role in fertilisation because of its specific expression in the testis. PKDILI localises to the Ley dig cells of the testis, a known location of testosterone production, suggesting a possible role in fertility and/or sexual maturation. PKDILI is also expressed in the heart, and although no role for this localised expression of PKDILI has yet been suggested, it may be implicated in cardiac diseases.
Thus, members of the PKD/REJ family of cation channels may be useful as contraceptive agents or for the treatment, prevention and or diagnosis of medical conditions and diseases which include, but are not limited to, renal disease, polycystic kidney disease, glomerulonephritis, fertility disorders, infertility, reproductive disorders, polycystic ovary disease, polycystic diseases, endocrine diseases, testosterone deficiency, testosterone-related disorder, testicular cancer, prostate cancer, gastrointestinal disorders, developmental disorders, brain tumour, sexual dysfunction, psychological disorder, alopecia, hirsuitism, diseases related to muscle tone, including vascular muscle disorders such as migraine, pre-eclampsia, hypertension and cardiac arrhythmias; bladder muscle tone disorders, such as urinary incontinence, inability to void, urinary tract infection; gastrointestinal smooth muscle tone disorder, such as Gl motility disorder, gastrointestinal reflux diorder (GERD), reflux oesophagitis, paralytic ileus, irritable bowel syndrome (IBS), diarrhoea including travellers diarrhoea and viral diarrhoea, constipation and malabsorption; skeletal muscle tone disorder, such as spacticity, and muscle fatigue disorders such as myotonias, myasthenia gravis, floppy baby syndrome, ME, frailty of old age; lung disorders such as COPD, asthma, pulmonary hypertension; adrenal disorders, such as adrenal hypertension, Conn's syndrome, Cushing disease and Addison's disease.
The identification of members of the PKD/REJ family of cation channels is of extreme importance in increasing the understanding of the underlying pathways that lead to the disease states and associated disease states mentioned above, and in developing more effective gene and/or drug therapies to treat these disorders, and also in increasing the understanding of the pathways involved in mammalian reproductive biology.
THE INVENTION
The invention is based on the discovery that the INPIONCH03 and INPIONCH04 proteins are cation channels, and moreover are members of the PKD/REJ family of cation channels.
In one embodiment of the first aspect of the invention, there is provided a polypeptide which:
(i) comprises the amino acid sequence as recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO-10, SEQ ID NO-12, SEQ ID NO:14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID
NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52, SEQ ID NO:54 and/or SEQ ID NO:56; (ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or has an antigenic determinant in common with the polypeptides of (i); or
(iii) is a functional equivalent of (i) or (ii).
Preferably, the polypeptide according to this first aspect of the invention:
(i) comprises the amino acid sequence as recited in SEQ ID NO:56; (ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or has an antigenic determinant in common with the polypeptides of (i); or
(iii) is a functional equivalent of (i) or (ii).
According to a second embodiment of this first aspect of the invention, there is provided a polypeptide which: (i) consists of the amino acid sequence as recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO-12, SEQ ID NO:14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID
NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52, SEQ ID NO:54 and/or SEQ ID NO:56;
(ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or having an antigenic determinant in common with the polypeptides of (i); or (iii) is a functional equivalent of (i) or (ii).
The polypeptide having the sequence recited in SEQ ID NO:2 is referred to hereafter as "INPIONCH03 exon 1 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:4 is referred to hereafter as "INPIONCH03 exon 2 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:6 is referred to hereafter as "INPIONCH03 exon 3 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 8 is referred to hereafter as "INPIONCH03 exon 4 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 10 is referred to hereafter as "INPIONCH03 exon 5 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 12 is referred to hereafter as "INPIONCH03 exon 6 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 14 is referred to hereafter as "INPIONCH03 exon 7 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 16 is referred to hereafter as "INPIONCH03 exon 8 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 18 is referred to hereafter as "INPIONCH03 exon 9 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:20 is referred to hereafter as "INPIONCH03 exon 10 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:22 is referred to hereafter as "INPIONCH03 exon 11 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:24 is referred to hereafter as "INPIONCH03 exon 12 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:26 is referred to hereafter as "INPIONCH03 exon 13 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:28 is referred to hereafter as "INPIONCH03 exon 14 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:30 is referred to hereafter as "INPIONCH03 exon 15 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:32 is referred to hereafter as "INPIONCH03 exon 16 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:34 is referred to hereafter as "INPIONCH03 exon 17 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:36 is referred to hereafter as "INPIONCH03 exon 18 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:38 is referred to hereafter as "INPIONCH03 exon 19 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:40 is referred to hereafter as "INPIONCH03 exon 20 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:42 is referred to hereafter as "INPIONCH03 exon 21 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:44 is referred to hereafter as "INPIONCH03 exon 22 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:46 is referred to hereafter as "INPIONCH03 exon 23 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:48 is referred to hereafter as "INPIONCH03 exon 24 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 50 is referred to hereafter as "INPIONCH03 exon 25 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:52 is referred to hereafter as "INPIONCH03 exon 26 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:54 is referred to hereafter as "INPIONCH03 exon 27 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:56 is referred to hereafter as the "INPIONCH03 polypeptide". The term "INPIONCH03 polypeptides" as used herein includes polypeptides comprising the INPIONCH03 exon 1 polypeptide, the INPIONCH03 exon 2 polypeptide, the
INPIONCH03 exon 3 polypeptide, the INPIONCH03 exon 4 polypeptide, the
INPIONCH03 exon 5 polypeptide, the INPIONCH03 exon 6 polypeptide, the
INPIONCH03 exon 7 polypeptide, the INPIONCH03 exon 8 polypeptide, the INPIONCH03 exon 9 polypeptide, the INPIONCH03 exon 10 polypeptide, the
INPIONCH03 exon 11 polypeptide, the INPIONCH03 exon 12 polypeptide, the
INPIONCH03 exon 13 polypeptide, the INPIONCH03 exon 14 polypeptide, the
INPIONCH03 exon 15 polypeptide, the INPIONCH03 exon 16 polypeptide, the
INPIONCH03 exon 17 polypeptide, the INPIONCH03 exon 18 polypeptide, the INPIONCH03 exon 19 polypeptide, the INPIONCH03 exon 20 polypeptide, the
INPIONCH03 exon 21 polypeptide, the INPIONCH03 exon 22 polypeptide, the
INPIONCH03 exon 23 polypeptide, the INPIONCH03 exon 24 polypeptide, the
INPIONCH03 exon 25 polypeptide, the INPIONCH03 exon 26 polypeptide, the INPIONCH03 exon 27 polypeptide and the 1NPIONCH03 polypeptide.
In a third embodiment of the first aspect of the invention, there is provided a polypeptide which:
(i) comprises the amino acid sequence as recited in SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72,
SEQ ID NO:74, SEQ ID NO:76, SEQ ID NO:78, SEQ ID NO:80, SEQ ID NO:82, SEQ ID NO:84, SEQ ID NO:86, SEQ ID NO:88, SEQ ID NO:90, SEQ ID NO:92, SEQ ID NO:94, SEQ ID NO:96, SEQ ID NO:98s SEQ ID NO: 100, SEQ ID NO: 102, SEQ ID NO:104, SEQ ID NO:106 and/or SEQ ID NO:108; (ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or has an antigenic determinant in common with the polypeptides of (i); or
(iii) is a functional equivalent of (i) or (ii).
Preferably, the polypeptide according to this embodiment of the first aspect of the invention: (i) comprises the amino acid sequence as recited in SEQ ID NO: 108;
(ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or has an antigenic determinant in common with the polypeptides of (i); or
(iii) is a functional equivalent of (i) or (ii).
According to a fourth embodiment of this first aspect of the invention, there is provided a polypeptide which:
(i) consists of the amino acid sequence as recited in SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72, SEQ ID NO:74, SEQ ID NO:76, SEQ ID NO:78, SEQ ID NO:80, SEQ ID NO:82, SEQ ID NO:84, SEQ ID NO:86, SEQ ID NO:88, SEQ ID NO:90, SEQ ID NO:92, SEQ ID NO:94, SEQ ID NO:96, SEQ ID NO:98, SEQ ID NO: 100, SEQ ID NO: 102,
SEQ ID NO: 104, SEQ ID NO: 106 and or SEQ ID NO: 108;
(ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or having an antigenic determinant in common with the polypeptides of (i); or
(iii) is a functional equivalent of (i) or (ii). The polypeptide having the sequence recited in SEQ ID NO:58 is referred to hereafter as "INPIONCH04 exon 1 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:60 is referred to hereafter as "INPIONCH04 exon 2 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:62 is referred to hereafter as "INPIONCH04 exon 3 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:64 is referred to hereafter as "INPIONCH04 exon 4 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 66 is referred to hereafter as "INPIONCH04 exon 5 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:68 is referred to hereafter as "INPIONCH04 exon 6 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:70 is referred to hereafter as "INPIONCH04 exon 7 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:72 is referred to hereafter as "INPIONCH04 exon 8 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:74 is referred to hereafter as "INPIONCH04 exon 9 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:76 is referred to hereafter as "INPIONCH04 exon 10 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:78 is referred to hereafter as "INPIONCH04 exon 11 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:80 is referred to hereafter as "INPIONCH04 exon 12 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 82 is referred to hereafter as "INPIONCH04 exon 13 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:84 is referred to hereafter as "INPIONCH04 exon 14 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:86 is referred to hereafter as "INPIONCH04 exon 15 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:88 is referred to hereafter as "INPIONCH04 exon 16 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:90 is referred to hereafter as "INPIONCH04 exon 17 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:92 is referred to hereafter as "INPIONCH04 exon 18 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:94 is referred to hereafter as "INPIONCH04 exon 19 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:96 is referred to hereafter as "INPIONCH04 exon 20 polypeptide". The polypeptide having the sequence recited in SEQ ID NO:98 is referred to hereafter as "INPIONCH04 exon 21 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 100 is referred to hereafter as "INPIONCH04 exon 22 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 102 is referred to hereafter as "INPIONCH04 exon 23 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 104 is referred to hereafter as "INPIONCH04 exon 24 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 106 is referred to hereafter as "INPIONCH04 exon 25 polypeptide". The polypeptide having the sequence recited in SEQ ID NO: 108 is referred to hereafter as the "INPIONCH04 polypeptide".
The term "INPIONCH04 polypeptides" as used herein includes polypeptides comprising the INPIONCH04 exon 1 polypeptide, the INPIONCH04 exon 2 polypeptide, the
INPIONCH04 exon 3 polypeptide, the INPIONCH04 exon 4 polypeptide, the
INPIONCH04 exon 5 polypeptide, the INPIONCH04 exon 6 polypeptide, the INPIONCH04 exon 7 polypeptide, the INPIONCH04 exon 8 polypeptide, the
INPIONCH04 exon 9 polypeptide, the INPIONCH04 exon 10 polypeptide, the
INPIONCH04 exon 11 polypeptide, the INPIONCH04 exon 12 polypeptide, the
INPIONCH04 exon 13 polypeptide, the INPIONCH04 exon 14 polypeptide, the
INPIONCH04 exon 15 polypeptide, the INPIONCH04 exon 16 polypeptide, the INPIONCH04 exon 17 polypeptide, the INPIONCH04 exon 18 polypeptide, the
INPIONCH04 exon 19 polypeptide, the INPIONCH04 exon 20 polypeptide, the
INPIONCH04 exon 21 polypeptide, the INPIONCH04 exon 22 polypeptide, the
INPIONCH04 exon 23 polypeptide, the INPIONCH04 exon 24 polypeptide, the INPIONCH04 exon 25 polypeptide and the INPIONCH04 polypeptide. As previously described, known genes that are members of the PKD/REJ family of cation channels have N-terminal regions of varying function and sizes. The sequences disclosed in SEQ ID NO:56 and SEQ ID NO: 108 do not contain large N-terminal domains, and would therefore appear similar to PKD2 and PKD2L2. However, as is illustrated below in the Examples, the closest homologues to SEQ ID NO:56 and SEQ ID NO: 108 are the sea urchin REJ3 protein and the PKDREJ 1 human and mouse proteins. The REJ3 and PKDREJ 1 proteins both contain substantially larger N-terminal domains than those of the PKD2 and PKD2L2 proteins. Therefore, it is considered by the Applicant that there may be one, and possibly two or more, further exons 5' to SEQ ID NO:l or SEQ ID NO: 57 in the genome that will provide amino acids that are N-terminal to the beginning of the sequences given in SEQ ID NO:56 and SEQ ID NO: 108, respectively. Thus, a polypeptide of the present invention preferably comprises the amino acid sequence given in SEQ ID NO:56 or SEQ ID NO: 108 but may also include additional N-terminal amino acid residues. As described above, known genes that are members of the PKD/REJ family of cation channels are implicated in the formation of multimeric polypeptide complexes. Accordingly, it is considered by the Applicant that the INPIONCH03 and INPIONCH04 polypeptides of the invention may be involved in the formation of homo- and hetero- multimeric polypeptide complexes. Furthermore, it is envisaged by the Applicant that these multimeric polypeptide complexes are likely to possess novel or modulated function, and therefore may be useful as contraceptive agents and in the treatment of the disease states and associated disease states mentioned above. Therefore, one embodiment of the first aspect of the invention provides homo- or hetero-multimeric complexes comprising the INPIONCH03 and/or INPIONCH04 polypeptides.
In a second aspect, the invention provides a purified nucleic acid molecule which encodes a polypeptide of the first aspect of the invention.
Preferably, the purified nucleic acid molecule comprises the nucleic acid sequence as recited in SEQ ID NO:l (encoding the INPIONCH03 exon 1 polypeptide), SEQ ID NO:3 (encoding the INPIONCH03 exon 2 polypeptide), SEQ ID NO:5 (encoding the INPIONCH03 exon 3 polypeptide), SEQ ID NO:7 (encoding the INPIONCH03 exon 4 polypeptide), SEQ ID NO:9 (encoding the INPIONCH03 exon 5 polypeptide), SEQ ID NO: 11 (encoding the INPIONCH03 exon 6 polypeptide), SEQ ID NO: 13 (encoding the INPIONCH03 exon 7 polypeptide), SEQ ID NO: 15 (encoding the INPIONCH03 exon 8 polypeptide), SEQ ID NO: 17 (encoding the INPIONCH03 exon 9 polypeptide), SEQ ID NO:19 (encoding the INPIONCH03 exon 10 polypeptide), SEQ ID NO:21 (encoding the INPIONCH03 exon 11 polypeptide), SEQ ID NO:23 (encoding the INPIONCH03 exon 12 polypeptide), SEQ ID NO:25 (encoding the INPIONCH03 exon 13 polypeptide), SEQ ID NO:27 (encoding the INPIONCH03 exon 14 polypeptide), SEQ ID NO:29 (encoding the INPIONCH03 exon 15 polypeptide), SEQ ID NO:31 (encoding the INPIONCH03 exon 16 polypeptide), SEQ ID NO:33 (encoding the INPIONCH03 exon 17 polypeptide), SEQ ID NO:35 (encoding the INPIONCH03 exon 18 polypeptide), SEQ ID NO:37 (encoding the INPIONCH03 exon 19 polypeptide), SEQ ID NO:39 (encoding the INPIONCH03 exon 20 polypeptide), SEQ ID NO:41 (encoding the INPIONCH03 exon 21 polypeptide), SEQ ID NO:43 (encoding the INPIONCH03 exon 22 polypeptide), SEQ ID NO:45 (encoding the INPIONCH03 exon 23 polypeptide), SEQ ID NO:47 (encoding the INPIONCH03 exon 24 polypeptide), SEQ ID NO:49 (encoding the INPIONCH03 exon 25 polypeptide), SEQ ID NO:51 (encoding the INPIONCH03 exon 26 polypeptide), SEQ ID NO:53 (encoding the INPIONCH03 exon 27 polypeptide), SEQ ID NO:55 (encoding the INPIONCH03 polypeptide), SEQ ID NO:57 (encoding the INPIONCH04 exon 1 polypeptide), SEQ ID NO:59 (encoding the INPIONCH04 exon 2 polypeptide), SEQ ID NO:61 (encoding the INPIONCH04 exon 3 polypeptide), SEQ ID NO:63 (encoding the INPIONCH04 exon 4 polypeptide), SEQ ID NO:65 (encoding the INPIONCH04 exon 5 polypeptide), SEQ ID NO:67 (encoding the INPIONCH04 exon 6 polypeptide), SEQ ID NO:69 (encoding the INPIONCH04 exon 7 polypeptide), SEQ ID NO:71 (encoding the INPIONCH04 exon 8 polypeptide), SEQ ID NO:73 (encoding the INPIONCH04 exon 9 polypeptide), SEQ ID NO:75 (encoding the INPIONCH04 exon 10 polypeptide), SEQ ID NO:77 (encoding the INPIONCH04 exon 11 polypeptide), SEQ ID NO:79 (encoding the INPIONCH04 exon 12 polypeptide), SEQ ID NO:81 (encoding the INPIONCH04 exon 13 polypeptide), SEQ ID NO:83 (encoding the INPIONCH04 exon 14 polypeptide), SEQ ID NO:85 (encoding the INPIONCH04 exon 15 polypeptide), SEQ ID NO:87 (encoding the INPIONCH04 exon 16 polypeptide), SEQ ID NO:89 (encoding the LNPIONCH04 exon 17 polypeptide), SEQ ID NO:91 (encoding the INPIONCH04 exon 18 polypeptide), SEQ ID NO:93 (encoding the INPIONCH04 exon 19 polypeptide), SEQ ID NO:95 (encoding the INPIONCH04 exon 20 polypeptide), SEQ ID NO:97 (encoding the INPIONCH04 exon 21 polypeptide), SEQ ID NO:99 (encoding the INPIONCH04 exon 22 polypeptide), SEQ ID NO: 101 (encoding the INPIONCH04 exon 23 polypeptide), SEQ ID NO: 103 (encoding the INPIONCH04 exon 24 polypeptide), SEQ ID NO: 105 (encoding the INPIONCH04 exon 25 polypeptide) and/or SEQ ID NO: 107 (encoding the INPIONCH04 polypeptide) or is a redundant equivalent or fragment of any one of these sequences.
The invention further provides that the purified nucleic acid molecule consists of the nucleic acid sequence as recited in SEQ ID NO:l (encoding the INPIONCH03 exon 1 polypeptide), SEQ ID NO:3 (encoding the INPIONCH03 exon 2 polypeptide), SEQ ID NO:5 (encoding the INPIONCH03 exon 3 polypeptide), SEQ ID NO:7 (encoding the INPIONCH03 exon 4 polypeptide), SEQ ID NO:9 (encoding the INPIONCH03 exon 5 polypeptide), SEQ ID NO: 11 (encoding the INPIONCH03 exon 6 polypeptide), SEQ ID NO: 13 (encoding the INPIONCH03 exon 7 polypeptide), SEQ ID NO: 15 (encoding the INPIONCH03 exon 8 polypeptide), SEQ ID NO: 17 (encoding the INPIONCH03 exon 9 polypeptide), SEQ ID NO: 19 (encoding the INPIONCH03 exon 10 polypeptide), SEQ ID NO:21 (encoding the INPIONCH03 exon 11 polypeptide), SEQ ID NO:23 (encoding the INPIONCH03 exon 12 polypeptide), SEQ ID NO:25 (encoding the INPIONCH03 exon 13 polypeptide), SEQ ID NO:27 (encoding the INPIONCH03 exon 14 polypeptide), SEQ ID NO:29 (encoding the INPIONCH03 exon 15 polypeptide), SEQ ID NO:31 (encoding the INPIONCH03 exon 16 polypeptide), SEQ ID NO:33 (encoding the INPIONCH03 exon 17 polypeptide), SEQ ID NO:35 (encoding the INPIONCH03 exon 18 polypeptide), SEQ ID NO:37 (encoding the INPIONCH03 exon 19 polypeptide), SEQ ID NO:39 (encoding the INPIONCH03 exon 20 polypeptide), SEQ ID NO:41 (encoding the INPIONCH03 exon 21 polypeptide), SEQ ID NO:43 (encoding the INPIONCH03 exon 22 polypeptide), SEQ ID NO:45 (encoding the INPIONCH03 exon 23 polypeptide), SEQ ID NO:47 (encoding the INPIONCH03 exon 24 polypeptide), SEQ ID NO:49 (encoding the INPIONCH03 exon 25 polypeptide), SEQ ID NO:51 (encoding the INPIONCH03 exon 26 polypeptide), SEQ ID NO:53 (encoding the INPIONCH03 exon 27 polypeptide), SEQ ID NO:55 (encoding the INPIONCH03 polypeptide), SEQ ID NO:57 (encoding the INPIONCH04 exon 1 polypeptide), SEQ ID NO:59 (encoding the INPIONCH04 exon 2 polypeptide), SEQ ID NO:61 (encoding the INPIONCH04 exon 3 polypeptide), SEQ ID NO:63 (encoding the INPIONCH04 exon 4 polypeptide), SEQ ID NO:65 (encoding the INPIONCH04 exon 5 polypeptide), SEQ ID NO:67 (encoding the INPIONCH04 exon 6 polypeptide), SEQ ID NO:69 (encoding the INPIONCH04 exon 7 polypeptide), SEQ ID NO:71 (encoding the INPIONCH04 exon 8 polypeptide), SEQ ID NO:73 (encoding the INPIONCH04 exon 9 polypeptide), SEQ ID NO:75 (encoding the INPIONCH04 exon 10 polypeptide), SEQ ID NO:77 (encoding the INPIONCH04 exon 11 polypeptide), SEQ ID NO:79 (encoding the INPIONCH04 exon 12 polypeptide), SEQ ID NO: 81 (encoding the INPIONCH04 exon 13 polypeptide), SEQ ID NO:83 (encoding the INPIONCH04 exon 14 polypeptide), SEQ ID NO:85 (encoding the INPIONCH04 exon 15 polypeptide), SEQ ID NO:87 (encoding the INPIONCH04 exon 16 polypeptide), SEQ ID NO:89 (encoding the INPIONCH04 exon 17 polypeptide), SEQ ID NO:91 (encoding the INPIONCH04 exon 18 polypeptide), SEQ ID NO:93 (encoding the INPIONCH04 exon 19 polypeptide), SEQ ID NO:95 (encoding the INPIONCH04 exon 20 polypeptide), SEQ ID NO:97 (encoding the INPIONCH04 exon 21 polypeptide), SEQ ID NO:99 (encoding the INPIONCH04 exon 22 polypeptide), SEQ ID NO: 101 (encoding the INPIONCH04 exon 23 polypeptide), SEQ ID NO: 103 (encoding the INPIONCH04 exon 24 polypeptide), SEQ ID NO: 105 (encoding the INPIONCH04 exon 25 polypeptide) and/or SEQ ID NO: 107 (encoding the INPIONCH04 polypeptide), or is a redundant equivalent or fragment of either of these sequences.
In a third aspect, the invention provides a purified nucleic acid molecule which hybridizes under high stringency conditions with a nucleic acid molecule of the second aspect of the invention.
In a fourth aspect, the invention provides a vector, such as an expression vector, that contains a nucleic acid molecule of the second or third aspect of the invention. In a fifth aspect, the invention provides a host cell transformed with a vector of the fourth aspect of the invention.
In a sixth aspect, the invention provides a ligand which binds specifically to the PKD/REJ cation channel family polypeptide of the first aspect of the invention.
In a seventh aspect, the invention provides a compound that is effective to alter the expression of a natural gene which encodes a polypeptide of the first aspect of the invention or to regulate the activity of a polypeptide of the first aspect of the invention.
A compound of the seventh aspect of the invention may either increase (agonise) or decrease (antagonise) the level of expression of the gene or the activity of the polypeptide.
Importantly, the identification of the function of the INPIONCH03 and INPIONCH04 polypeptides allows for the design of screening methods capable of identifying compounds that are effective in the treatment and/or diagnosis of disease. Ligands and compounds according to the sixth and seventh aspects of the invention may be identified using such methods. These methods are included as aspects of the present invention.
In an eighth aspect, the invention provides a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention, for use in therapy or diagnosis of diseases in which ion channels are implicated. Such diseases include cardiovascular disease, hypertension, heart arrhythmia, angina, myocardial infarction, tachycardia cerebrovascular disease, thromboembolism, neurological and psychiatric disorders, including stroke, epilepsy, migraine, pain, depression, neuralgia, Alzheimer's disease, brain injury, schizophrenia,
Huntington's disease, motor neurone disease, Parkinson's disease, neuropathy, neuromuscular disorder, micturition disorder, impotence, diabetes, dermatitis, pulmonary disease, asthma, cystic fibrosis, mucous membrane disorders including sinusitis, COPD, rhinitis and cough, leukemia, ocular disease, glaucoma, retinopathy and immune disorders. Preferably, the diseases are those in which members of the PKD/REJ family of cation channels are implicated. Examples of such diseases include renal disease, polycystic kidney disease, glomerulnephritis, fertility disorders, infertility, reproductive disorders, polycystic ovary disease, polycystic diseases, endocrine diseases, testosterone deficiency, testosterone-related disorder, testicular cancer, prostate cancer, gastrointestinal disorders, developmental disorders, brain tumour, sexual dysfunction, psychological disorder, alopecia, hirsuitism, diseases related to muscle tone, including vascular muscle disorders such as migraine, pre-eclampsia, hypertension and cardiac arrhythmias; bladder muscle tone disorders, such as urinary incontinence, inability to void, urinary tract infection; gastrointestinal smooth muscle tone disorder, such as Gl motility disorder, gastrointestinal reflux diorder (GERD), reflux oesophagitis, paralytic ileus, irritable bowel syndrome (IBS), diarrhoea including travellers diarrhoea and viral diarrhoea, constipation and malabsorption; skeletal muscle tone disorder, such as spacticity, and muscle fatigue disorders such as myotonias, myasthenia gravis, floppy baby syndrome, ME, frailty of old age; lung disorders such as COPD, asthma, pulmonary hypertension; adrenal disorders, such as adrenal hypertension, Conn's syndrome, Cushing disease and Addison's disease. These molecules may also be used in the manufacture of a medicament for the treatment of such diseases. These moieties of the first, second, third, fourth, fifth, sixth or seventh aspect of the invention may also be used in the manufacture of a medicament for the treatment of such diseases.
In a further embodiment according to the eighth aspect of the invention, there is provided a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention, for use as a contraceptive agent. These molecules may also be used in the manufacture of a medicament suitable for contraceptive use. These moieties of the first, second, third, fourth, fifth, sixth or seventh aspect of the invention may also be used in the manufacture of a medicament for the treatment of such diseases.
In a ninth aspect, the invention provides a method of diagnosing a disease in a patient, comprising assessing the level of expression of a natural gene encoding a polypeptide of the first aspect of the invention or the activity of a polypeptide of the first aspect of the invention in tissue from said patient and comparing said level of expression or activity to a control level, wherein a level that is different to said control level is indicative of disease. Such a method will preferably be carried out in vitro. Similar methods may be used for monitoring the therapeutic treatment of disease in a patient, wherein altering the level of expression or activity of a polypeptide or nucleic acid molecule over the period of time towards a control level is indicative of regression of disease. A preferred method for detecting polypeptides of the first aspect of the invention comprises the steps of: (a) contacting a ligand, such as an antibody, of the sixth aspect of the invention with a biological sample under conditions suitable for the formation of a ligand-polypeptide complex; and (b) detecting said complex.
A number of different such methods according to the ninth aspect of the invention exist, as the skilled reader will be aware, such as methods of nucleic acid hybridization with short probes, point mutation analysis, polymerase chain reaction (PCR) amplification and methods using antibodies to detect aberrant protein levels. Similar methods may be used on a short or long term basis to allow therapeutic treatment of a disease to be monitored in a patient. The invention also provides kits that are useful in these methods for diagnosing disease.
Preferably, the disease diagnosed by a method of the ninth aspect of the invention is a disease in which members of the PKD/REJ family of cation channels are implicated, as described above.
In a tenth aspect, the invention provides for the use of a polypeptide of the first aspect of the invention as a cation channel. For example, cells may be modified to express the cation channels of the invention at their cell membrane at varying levels, thus allowing for the selective modulation of cell membrane cation permeability. Manipulation of the cation permeability of the cell membrane in this way, for example in Xenopus oocytes, permits the investigation of the properties of the lipid bilayer and of other proteins (including other ion channels) within the lipid bilayer, as well as cation-regulated cellular signalling pathways and is likely to be of significant utility in this respect.
In an eleventh aspect, the invention provides a pharmaceutical composition comprising a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention, in conjunction with a pharmaceutically- acceptable carrier. In a twelfth aspect, the present invention provides a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention, for use in the manufacture of a medicament for the diagnosis or treatment of a disease.
In a second embodiment of the twelfth aspect of the invention, there is provided a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention, for use in the manufacture of a contraceptive agent.
Preferably, the disease is a disease in which members of the PKD/REJ family of cation channels are implicated, as described above. In a thirteenth aspect, the invention provides a method of treating a disease in a patient comprising administering to the patient a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention. In a second embodiment of the thirteenth aspect of the invention, there is provided a method of preventing pregnancy in a patient comprising administering to the patient a polypeptide of the first aspect of the invention, or a nucleic acid molecule of the second or third aspect of the invention, or a vector of the fourth aspect of the invention, or a host cell of the fifth aspect of the invention, or a ligand of the sixth aspect of the invention, or a compound of the seventh aspect of the invention. Preferably, the disease is a disease in which members of the PKD/REJ family of cation channels are implicated, as described above.
For diseases in which the expression of a natural gene encoding a polypeptide of the first aspect of the invention, or in which the activity of a polypeptide of the first aspect of the invention, is lower in a diseased patient when compared to the level of expression or activity in a healthy patient, the polypeptide, nucleic acid molecule, vector, host cell, ligand or compound administered to the patient should be an agonist. Conversely, for diseases in which the expression of the natural gene or activity of the polypeptide is higher in a diseased patient when compared to the level of expression or activity in a healthy patient, the polypeptide, nucleic acid molecule, vector, host cell, ligand or compound administered to the patient should be an antagonist. Examples of such antagonists include antisense nucleic acid molecules, ribozymes and ligands, such as antibodies.
In a fourteenth aspect, the invention provides transgenic or knockout non-human animals that have been transformed to express higher, lower or absent levels of a polypeptide of the first aspect of the invention. Such transgenic animals are very useful models for the study of disease and may also be used in screening regimes for the identification of compounds that are effective in the treatment or diagnosis of such a disease.
Preferably, the disease is a disease in which members of the PKD/REJ family of cation channels are implicated, as described above.
A summary of standard techniques and procedures which may be employed in order to utilise the invention is given below. It will be understood that this invention is not limited to the particular methodology, protocols, cell lines, vectors and reagents described. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and it is not intended that this terminology should limit the scope of the present invention. The extent of the invention is limited only by the terms of the appended claims. Standard abbreviations for nucleotides and amino acids are used in this specification.
The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology, microbiology, recombinant DNA technology and immunology, which are within the skill of those working in the art.
Such techniques are explained fully in the literature. Examples of particularly suitable texts for consultation include the following: Sambrook Molecular Cloning; A Laboratory
Manual, Second Edition (1989); DNA Cloning, Volumes I and II (D.N Glover ed. 1985);
Oligonucleotide Synthesis (M.J. Gait ed. 1984); Nucleic Acid Hybridization (B.D. Hames
& S.J. Higgins eds. 1984); Transcription and Translation (B.D. Hames & S.J. Higgins eds.
1984); Animal Cell Culture (R.I. Freshney ed. 1986); Immobilized Cells and Enzymes (IRL Press, 1986); B. Perbal, A Practical Guide to Molecular Cloning (1984); the Methods in Enzymology series (Academic Press, Inc.), especially volumes 154 & 155; Gene Transfer Vectors for Mammalian Cells (J.H. Miller and M.P. Calos eds. 1987, Cold Spring Harbor Laboratory); Immunochemical Methods in Cell and Molecular Biology (Mayer and Walker, eds. 1987, Academic Press, London); Scopes, (1987) Protein Purification: Principles and Practice, Second Edition (Springer Verlag, N.Y.); and Handbook of Experimental Immunology, Volumes I-IV (D.M. Weir and C. C. Blackwell eds. 1986).
As used herein, the term "polypeptide" includes any peptide or protein comprising two or more amino acids joined to each other by peptide bonds or modified peptide bonds, i.e. peptide isosteres. This term refers both to short chains (peptides and oligopeptides) and to longer chains (proteins). The polypeptide of the present invention may be in the form of a mature protein or may be a pre-, pro- or prepro- protein that can be activated by cleavage of the pre-, pro- or prepro- portion to produce an active mature polypeptide. In such polypeptides, the pre-, pro- or prepro- sequence may be a leader or secretory sequence or may be a sequence that is employed for purification of the mature polypeptide sequence. The polypeptide of the first aspect of the invention may form part of a fusion protein. For example, it is often advantageous to include one or more additional amino acid sequences which may contain secretory or leader sequences, pro-sequences, sequences which aid in purification, or sequences that confer higher protein stability, for example during recombinant production. Alternatively or additionally, the mature polypeptide may be fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol).
Polypeptides may contain amino acids other than the 20 gene-encoded amino acids, modified either by natural processes, such as by post-translational processing or by chemical modification techniques which are well known in the art. Among the known modifications which may commonly be present in polypeptides of the present invention are glycosylation, lipid attachment, sulphation, gamma-carboxylation, for instance of glutamic acid residues, hydroxylation and ADP-ribosylation. Other potential modifications include acetylation, acylation, amidation, covalent attachment of flavin, covalent attachment of a haeme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid derivative, covalent attachment of phosphatidylinositol, cross-linking, cyclization, disulphide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, GPI anchor formation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, transfer- RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. In fact, blockage of the amino or carboxyl terminus in a polypeptide, or both, by a covalent modification is common in naturally-occurring and synthetic polypeptides and such modifications may be present in polypeptides of the present invention.
The modifications that occur in a polypeptide often will be a function of how the polypeptide is made. For polypeptides that are made recombinantly, the nature and extent of the modifications in large part will be determined by the post-translational modification capacity of the particular host cell and the modification signals that are present in the amino acid sequence of the polypeptide in question. For instance, glycosylation patterns vary between different types of host cell. The polypeptides of the present invention can be prepared in any suitable manner. Such polypeptides include isolated naturally-occurring polypeptides (for example purified from cell culture), recombinantly-produced polypeptides (including fusion proteins), synthetically-produced polypeptides or polypeptides that are produced by a combination of these methods. The functionally-equivalent polypeptides of the first aspect of the invention may be polypeptides that are homologous to the INPIONCH03 and INPIONCH04 polypeptides. Two polypeptides are said to be "homologous", as the term is used herein, if the sequence of one of the polypeptides has a high enough degree of identity or similarity to the sequence of the other polypeptide. "Identity" indicates that at any particular position in the aligned sequences, the amino acid residue is identical between the sequences. "Similarity" indicates that, at any particular position in the aligned sequences, the amino acid residue is of a similar type between the sequences. Degrees of identity and similarity can be readily calculated (Computational Molecular Biology, Lesk, A.M., ed., Oxford University Press, New York, 1988; Biocomputing. Informatics and Genome Projects, Smith, D.W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1, Griffin, A.M., and Griffin, H.G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991).
Homologous polypeptides therefore include natural biological variants (for example, allelic variants or geographical variations within the species from which the polypeptides are derived) and mutants (such as mutants containing amino acid substitutions, insertions or deletions) of the INPIONCH03 or INPIONCH04 polypeptides. Such mutants may include polypeptides in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code. Typical such substitutions are among Ala, Val, Leu and He; among Ser and Thr; among the acidic residues Asp and Glu; among Asn and Gin; among the basic residues Lys and Arg; or among the aromatic residues Phe and Tyr. Particularly preferred are variants in which several, i.e. between 5 and 10, 1 and 5, 1 and 3, 1 and 2 or just 1 amino acids are substituted, deleted or added in any combination. Especially preferred are silent substitutions, additions and deletions, which do not alter the properties and activities of the protein. Also especially preferred in this regard are conservative substitutions. Such mutants also include polypeptides in which one or more of the amino acid residues includes a substituent group.
Typically, greater than 30% identity between two polypeptides is considered to be an indication of functional equivalence. Preferably, functionally equivalent polypeptides of the first aspect of the invention have a degree of sequence identity with the INPIONCH03 or INPIONCH04 polypeptides, or with active fragments thereof, of greater than 80%. More prefeπed polypeptides have degrees of identity of greater than 85%, 90%, 95%, 98% or 99%, respectively. The polypeptides whose sequences are recited in accession numbers XP_091397.1, BAB67772.1 and XP_056635.1 are specifically excluded from the scope of this aspect of the invention.
The functionally-equivalent polypeptides of the first aspect of the invention may also be polypeptides which have been identified using one or more techniques of structural alignment. For example, the Inpharmatica Genome Threader technology that forms one aspect of the search tools used to generate the Biopendium™ search database may be used (see co-pending PCT application PCT/GBO 1/01105, published as WO 01/69507) to identify polypeptides of presently-unknown function which, while having low sequence identity as compared to the INPIONCH03 and INPIONCH04 polypeptides, are predicted to be members of the PKD/REJ cation channel family, by virtue of sharing significant structural homology with the INPIONCH03 or INPIONCH04 polypeptide sequences. By "significant structural homology" is meant that the Inpharmatica Genome Threader predicts two proteins to share structural homology with a certainty of 10% and above. The polypeptides of the first aspect of the invention also include fragments of the INPIONCH03 and INPIONCH04 polypeptides and fragments of the functional equivalents of the INPIONCH03 and INPIONCH04 polypeptides, provided that those fragments are members of the PKD/REJ family of cation channels or have an antigenic determinant in common with the INPIONCH03 or INPIONCH04 polypeptides. As used herein, the term "fragment" refers to a polypeptide having an amino acid sequence that is the same as part, but not all, of the amino acid sequence of the INPIONCH03 or INPIONCH04 polypeptides or one of their functional equivalents. The fragments should comprise at least n consecutive amino acids from the sequence and, depending on the particular sequence, n preferably is 7 or more (for example, 8, 10, 12, 14, 16, 18, 20 or more). Small fragments may form an antigenic determinant.
Such fragments may be "free-standing", i.e. not part of or fused to other amino acids or polypeptides, or they may be comprised within a larger polypeptide of which they form a part or region. When comprised within a larger polypeptide, the fragment of the invention most preferably forms a single continuous region. For instance, certain preferred embodiments relate to a fragment having a pre- and/or pro- polypeptide region fused to the amino terminus of the fragment and/or an additional region fused to the carboxyl terminus of the fragment. However, several fragments may be comprised within a single larger polypeptide.
The polypeptides of the present invention or their immunogenic fragments (comprising at least one antigenic determinant) can be used to generate ligands, such as polyclonal or monoclonal antibodies, that are immunospecific for the polypeptides. Such antibodies may be employed to isolate or to identify clones expressing the polypeptides of the invention or to purify the polypeptides by affinity chromatography. The antibodies may also be employed as diagnostic or therapeutic aids, amongst other applications, as will be apparent to the skilled reader.
The term "immunospecific" means that the antibodies have substantially greater affinity for the polypeptides of the invention than their affinity for other related polypeptides in the prior art. As used herein, the term "antibody" refers to intact molecules as well as to fragments thereof, such as Fab, F(ab')2 and Fv, which are capable of binding to the antigenic determinant in question. Such antibodies thus bind to the polypeptides of the first aspect of the invention. By "substantially greater affinity" we mean that there is a measurable increase in the affinity for a polypeptide of the invention as compared with the affinity for known cell- surface receptors.
Preferably, the affinity is at least 1.5-fold, 2-fold, 5-fold 10-fold, 100-fold, 103-fold, 104- fold, 105-fold or 106-fold greater for a polypeptide of the invention than for known cell- surface receptor polypeptides .
If polyclonal antibodies are desired, a selected mammal, such as a mouse, rabbit, goat or horse, may be immunised with a polypeptide of the first aspect of the invention. The polypeptide used to immunise the animal can be derived by recombinant DNA technology or can be synthesized chemically. If desired, the polypeptide can be conjugated to a carrier protein. Commonly used carriers to which the polypeptides may be chemically coupled include bovine serum albumin, thyroglobulin and keyhole limpet haemocyanin. The coupled polypeptide is then used to immunise the animal. Serum from the immunised animal is collected and treated according to known procedures, for example by immunoaffinity chromatography. Monoclonal antibodies to the polypeptides of the first aspect of the invention cam also be readily produced by one skilled in the art. The general methodology for making monoclonal antibodies using hybridoma technology is well known (see, for example, Kohler, G. and Milstein, C, Nature 256: 495-497 (1975); Kozbor et al, Immunology Today 4: 72 (1983); Cole et al, 77-96 in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc. (1985).
Panels of monoclonal antibodies produced against the polypeptides of the first aspect of the invention can be screened for various properties, i.e., for isotype, epitope, affinity, etc. Monoclonal antibodies are particularly useful in purification of the individual polypeptides against which they are directed. Alternatively, genes encoding the monoclonal antibodies of interest may be isolated from hybridomas, for instance by PCR techniques known in the art, and cloned and expressed in appropriate vectors. Chimeric antibodies, in which non-human variable regions are joined or fused to human constant regions (see, for example, Liu et al, Proc. Natl. Acad. Sci. USA, 84, 3439 (1987)), may also be of use.
The antibody may be modified to make it less immunogenic in an individual, for example by humanisation (see Jones et al, Nature, 321, 522 (1986); Verhoeyen et al, Science, 239, 1534 (1988); Kabat et al, J. Immunol., 147, 1709 (1991); Queen et al, Proc. Natl Acad. Sci. USA, 86, 10029 (1989); Gorman et al, Proc. Natl Acad. Sci. USA, 88, 34181 (1991); and Hodgson et al, Bio/Technology, 9, 421 (1991)). The term "humanised antibody", as used herein, refers to antibody molecules in which the CDR amino acids and selected other amino acids in the variable domains of the heavy and/or light chains of a non-human donor antibody have been substituted in place of the equivalent amino acids in a human antibody. The humanised antibody thus closely resembles a human antibody but has the binding ability of the donor antibody.
In a further alternative, the antibody may be a "bispecific" antibody, that is an antibody having two different antigen binding domains, each domain being directed against a different epitope.
Phage display technology may be utilised to select genes which encode antibodies with binding activities towards the polypeptides of the invention either from repertoires of PCR amplified V-genes of lymphocytes from humans screened for possessing the relevant antibodies, or from naive libraries (McCafferty, J. et al, (1990), Nature 348, 552-554; Marks, J. et al, (1992) Biotechnology 10, 779-783). The affinity of these antibodies can also be improved by chain shuffling (Clackson, T. et al, (1991) Nature 352, 624-628).
Antibodies generated by the above techniques, whether polyclonal or monoclonal, have additional utility in that they may be employed as reagents in immunoassays, radioimmunoassays (RIA) or enzyme-linked immunosorbent assays (ELISA). In these applications, the antibodies can be labelled with an analytically-detectable reagent such as a radioisotope, a fluorescent molecule or an enzyme.
Preferred nucleic acid molecules of the second and third aspects of the invention are those which encode a polypeptide sequence as recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52, SEQ ID NO:54, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72, SEQ ID NO:74, SEQ ID NO:76, SEQ ID NO:78, SEQ ID NO:80, SEQ ID NO:82, SEQ ID NO:84, SEQ ID NO:86, SEQ ID NO:88, SEQ ID NO:90, SEQ ID NO:92, SEQ ID NO:94, SEQ ID NO:96, SEQ ID NO:98, SEQ ID NO: 100, SEQ ID NO: 102, SEQ ID NO: 104, SEQ ID NO: 106 and/or SEQ ID NO: 108 and functionally equivalent polypeptides. These nucleic acid molecules may be used in the methods and applications described herein. The nucleic acid molecules of the invention preferably comprise at least n consecutive nucleotides from the sequences disclosed herein where, depending on the particular sequence, n is 10 or more (for example, 12, 14, 15, 18, 20, 25, 30, 35, 40 or more).
The nucleic acid molecules of the invention also include sequences that are complementary to nucleic acid molecules described above (for example, for antisense or probing purposes). Nucleic acid molecules of the present invention may be in the form of RNA, such as mRNA, or in the form of DNA, including, for instance cDN A, synthetic DNA or genomic DNA. Such nucleic acid molecules may be obtained by cloning, by chemical synthetic techniques or by a combination thereof. The nucleic acid molecules can be prepared, for example, by chemical synthesis using techniques such as solid phase phosphoramidite chemical synthesis, from genomic or cDNA libraries or by separation from an organism. RNA molecules may generally be generated by the in vitro or in vivo transcription of DNA sequences.
The nucleic acid molecules may be double-stranded or single-stranded. Single-stranded DNA may be the coding strand, also known as the sense strand, or it may be the non- coding strand, also referred to as the anti-sense strand.
The term "nucleic acid molecule" also includes analogues of DNA and RNA, such as those containing modified backbones, and peptide nucleic acids (PNA). The term "PNA", as used herein, refers to an antisense molecule or an anti-gene agent which comprises an oligonucleotide of at least five nucleotides in length linked to a peptide backbone of amino acid residues, which preferably ends in lysine. The terminal lysine confers solubility to the composition. PNAs may be pegylated to extend their lifespan in a cell, where they preferentially bind complementary single stranded DNA and RNA and stop transcript elongation (Nielsen, P.E. et al. (1993) Anticancer Drug Des. 8:53-63).
A nucleic acid molecule which encodes a polypeptide of this invention may be identical to the coding sequence of one or more of the nucleic acid molecules disclosed herein.
These molecules also may have a different sequence which, as a result of the degeneracy of the genetic code, encodes a polypeptide of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO.12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52, SEQ ID NO:54, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72, SEQ ID NO:74, SEQ ID NO:76, SEQ ID NO:78, SEQ ID NO:80, SEQ ID NO:82, SEQ ID NO:84, SEQ ID NO:86, SEQ ID NO:88, SEQ ID NO:90, SEQ ID NO:92, SEQ ID NO:94, SEQ ID NO:96, SEQ ID NO:98, SEQ ID NO: 100, SEQ ID NO: 102, SEQ ID NO: 104, SEQ ID NO: 106 or SEQ ID NO: 108. Such nucleic acid molecules may include, but are not limited to, the coding sequence for the mature polypeptide by itself; the coding sequence for the mature polypeptide and additional coding sequences, such as those encoding a leader or secretory sequence, such as a pro-, pre- or prepro- polypeptide sequence; the coding sequence of the mature polypeptide, with or without the aforementioned additional coding sequences, together with further additional, non-coding sequences, including non-coding 5' and 3' sequences, such as the transcribed, non-translated sequences that play a role in transcription (including termination signals), ribosome binding and mRNA stability. The nucleic acid molecules may also include additional sequences which encode additional amino acids, such as those which provide additional functionalities. The nucleic acid molecules of the second and third aspects of the invention may also encode the fragments or the functional equivalents of the polypeptides and fragments of the first aspect of the invention. Such a nucleic acid molecule may be a naturally-occurring variant such as a naturally-occurring allelic variant, or the molecule may be a variant that is not known to occur naturally. Such non-naturally occurring variants of the nucleic acid molecule may be made by mutagenesis techniques, including those applied to nucleic acid molecules, cells or organisms.
Among variants in this regard are variants that differ from the aforementioned nucleic acid molecules by nucleotide substitutions, deletions or insertions. The substitutions, deletions or insertions may involve one or more nucleotides. The variants may be altered in coding or non-coding regions or both. Alterations in the coding regions may produce conservative or non-conservative amino acid substitutions, deletions or insertions. The nucleic acid molecules of the invention can also be engineered, using methods generally known in the art, for a variety of reasons, including modifying the cloning, processing, and/or expression of the gene product (the polypeptide). DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides are included as techniques which may be used to engineer the nucleotide sequences. Site-directed mutagenesis may be used to insert new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, introduce mutations and so forth.
Nucleic acid molecules which encode a polypeptide of the first aspect of the invention may be ligated to a heterologous sequence so that the combined nucleic acid molecule encodes a fusion protein. Such combined nucleic acid molecules are included within the second or third aspects of the invention. For example, to screen peptide libraries for inhibitors of the activity of the polypeptide, it may be useful to express, using such a combined nucleic acid molecule, a fusion protein that can be recognised by a commercially-available antibody. A fusion protein may also be engineered to contain a cleavage site located between the sequence of the polypeptide of the invention and the sequence of a heterologous protein so that the polypeptide may be cleaved and purified away from the heterologous protein.
The nucleic acid molecules of the invention also include antisense molecules that are partially complementary to nucleic acid molecules encoding polypeptides of the present invention and that therefore hybridize to the encoding nucleic acid molecules (hybridization). Such antisense molecules, such as oligonucleotides, can be designed to recognise, specifically bind to and prevent transcription of a target nucleic acid encoding a polypeptide of the invention, as will be known by those of ordinary skill in the art (see, for example, Cohen, J.S., Trends in Pharm. Sci., 10, 435 (1989), Okano, J. Neurochem. 56, 560 (1991); O'Connor, J. Neurochem 56, 560 (1991); Lee et al, Nucleic Acids Res 6, 3073 (1979); Cooney et al, Science 241, 456 (1988); Dervan et al, Science 251, 1360 (1991).
The term "hybridization" as used here refers to the association of two nucleic acid molecules with one another by hydrogen bonding. Typically, one molecule will be fixed to a solid support and the other will be free in solution. Then, the two molecules may be placed in contact with one another under conditions that favour hydrogen bonding. Factors that affect this bonding include: the type and volume of solvent; reaction temperature; time of hybridization; agitation; agents to block the non-specific attachment of the liquid phase molecule to the solid support (Denhardt's reagent or BLOTTO); the concentration of the molecules; use of compounds to increase the rate of association of molecules (dextran sulphate or polyethylene glycol); and the stringency of the washing conditions following hybridization (see Sambrook et al. [supra]).
The inhibition of hybridization of a completely complementary molecule to a target molecule may be examined using a hybridization assay, as known in the art (see, for example, Sambrook et al [supra]). A substantially homologous molecule will then compete for and inhibit the binding of a completely homologous molecule to the target molecule under various conditions of stringency, as taught in Wahl, G.M. and S.L. Berger (1987; Methods Enzymol. 152:399-407) and Kimmel, A.R. (1987; Methods Enzymol. 152:507- 511).
"Stringency" refers to conditions in a hybridization reaction that favour the association of very similar molecules over association of molecules that differ. High stringency hybridisation conditions are defined as overnight incubation at 42°C in a solution comprising 50% formamide, 5XSSC (150mM NaCl, 15mM trisodium citrate), 50mM sodium phosphate (pH7.6), 5x Denhardts solution, 10% dextran sulphate, and 20 microgram/ml denatured, sheared salmon sperm DNA, followed by washing the filters in 0.1X SSC at approximately 65°C. Low stringency conditions involve the hybridisation reaction being carried out at 35°C (see Sambrook et al. [supra]). Preferably, the conditions used for hybridization are those of high stringency. Preferred embodiments of this aspect of the invention -ire nucleic acid molecules that are at least 70% identical over their entire length to a nucleic acid molecule encoding the INPIONCH03 and INPIONCH04 polypeptides and nucleic acid molecules that are substantially complementary to such nucleic acid molecules. Preferably, a nucleic acid molecule according to this aspect of the invention comprises a region that is at least 80% identical over its entire length to such coding sequences, or is a nucleic acid molecule that is complementary thereto. In this regard, nucleic acid molecules at least 90%, preferably at least 95%, more preferably at least 98%, 99% or more identical over their entire length to the same are particularly preferred. Prefeπed embodiments in this respect are nucleic acid molecules that encode polypeptides which retain substantially the same biological function or activity as the INPIONCH03 and INPIONCH04 polypeptides. Nucleic acid molecules that encode the polypeptides whose sequences are recited in accession numbers XP_091397.1, BAB67772.1 and XP_056635.1 are specifically excluded from the scope of this aspect of the invention.
The invention also provides a process for detecting a nucleic acid molecule of the invention, comprising the steps of: (a) contacting a nucleic probe according to the invention with a biological sample under hybridizing conditions to form duplexes; and (b) detecting any such duplexes that are formed.
As discussed additionally below in connection with assays that may be utilised according to the invention, a nucleic acid molecule as described above may be used as a hybridization probe for RNA, cDNA or genomic DNA, in order to isolate full-length cDNAs and genomic clones encoding the INPIONCH03 or INPIONCH04 polypeptides and to isolate cDNA and genomic clones of homologous or orthologous genes that have a high sequence similarity to the gene encoding this polypeptide.
In this regard, the following techniques, among others known in the art, may be utilised and are discussed below for purposes of illustration. Methods for DNA sequencing and analysis are well known and are generally available in the art and may, indeed, be used to practice many of the embodiments of the invention discussed herein. Such methods may employ such enzymes as the Klenow fragment of DNA polymerase I, Sequenase (US Biochemical Corp, Cleveland, OH), Taq polymerase (Perkin Elmer), thermostable T7 polymerase (Amersham, Chicago, IL), or combinations of polymerases and proof-reading exonucleases such as those found in the ELONGASE Amplification System marketed by Gibco/BRL (Gaithersburg, MD). Preferably, the sequencing process may be automated using machines such as the Hamilton Micro Lab 2200 (Hamilton, Reno, NV), the Peltier Thermal Cycler (PTC200; MJ Research, Watertown, MA) and the ABI Catalyst and 373 and 377 DNA Sequencers (Perkin Elmer).
One method for isolating a nucleic acid molecule encoding a polypeptide with an equivalent function to that of the INPIONCH03 and INPIONCH04 polypeptides is to probe a genomic or cDNA library with a natural or artificially-designed probe using standard procedures that are recognised in the art (see, for example, "Current Protocols in Molecular Biology", Ausubel et al. (eds). Greene Publishing Association and John Wiley Interscience, New York, 1989,1992). Probes comprising at least 15, preferably at least 30, and more preferably at least 50, contiguous bases that correspond to, or are complementary to, nucleic acid sequences from the appropriate encoding gene (SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l 1, SEQ ID NO-13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:27, SEQ ID NO:29, SEQ ID NO:31, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:37, SEQ ID NO:39, SEQ ID NO:41, SEQ ID NO:43, SEQ ID NO:45, SEQ ID NO:47, SEQ ID NO:49, SEQ ID NO-51, SEQ ID NO:53, SEQ ID NO:55, SEQ ID NO:57, SEQ ID NO:59, SEQ ID NO:61, SEQ ID NO:63, SEQ ID NO:65, SEQ ID NO:67, SEQ ID NO:69, SEQ ID NO:71, SEQ ID NO:73, SEQ ID NO:75, SEQ ID NO:79, SEQ ID NO:81, SEQ ID NO:83, SEQ ID NO:85, SEQ ID NO:87, , SEQ ID NO:89, SEQ ID NO:91, SEQ ID NO:93, SEQ ID NO:95, SEQ ID NO:97, SEQ ID NO:99, SEQ ID NO-101, SEQ ID NO: 103, SEQ ID NO: 105 and SEQ ID NO: 107), are particularly useful probes. Such probes may be labelled with an analytically-detectable reagent to facilitate their identification. Useful reagents include, but are not limited to, radioisotopes, fluorescent dyes and enzymes that are capable of catalysing the formation of a detectable product. Using these probes, the ordinarily skilled artisan will be capable of isolating complementary copies of genomic DNA, cDNA or RNA polynucleotides encoding proteins of interest from human, mammalian or other animal sources and screening such sources for related sequences, for example, for additional members of the family, type and/or subtype.
In many cases, isolated cDNA sequences will be incomplete, in that the region encoding the polypeptide will be cut short, normally at the 5' end. Several methods are available to obtain full length cDNAs, or to extend short cDNAs. Such sequences may be extended utilising a partial nucleotide sequence and employing various methods known in the art to detect upstream sequences such as promoters and regulatory elements. For example, one method which may be employed is based on the method of Rapid Amplification of cDNA Ends (RACE; see, for example, Frohman et al, PNAS USA 85, 8998-9002, 1988). Recent modifications of this technique, exemplified by the Marathon™ technology (Clontech Laboratories Inc.), for example, have significantly simplified the search for longer cDNAs. A slightly different technique, termed "restriction-site" PCR, uses universal primers to retrieve unknown nucleic acid sequence adjacent a known locus (Sarkar, G. (1993) PCR Methods Applic. 2:318-322). Inverse PCR may also be used to amplify or to extend sequences using divergent primers based on a known region (Triglia, T. et al. (1988) Nucleic Acids Res. 16:8186). Another method which may be used is capture PCR which involves PCR amplification of DNA fragments adjacent a known sequence in human and yeast artificial chromosome DNA (Lagerstrom, M. et al. (1991) PCR Methods Applic, 1, 111-119). Another method which may be used to retrieve unknown sequences is that of Parker, J.D. et al. (1991); Nucleic Acids Res. 19:3055-3060). Additionally, one may use PCR, nested primers, and PromoterFinder™ libraries to walk genomic DNA (Clontech, Palo Alto, CA). This process avoids the need to screen libraries and is useful in finding intron/exon j unctions .
When screening for full-length cDNAs, it is preferable to use libraries that have been size- selected to include larger cDNAs. Also, random-primed libraries are preferable, in that they will contain more sequences that contain the 5' regions of genes. Use of a randomly primed library may be especially preferable for situations in which an oligo d(T) library does not yield a full-length cDNA. Genomic libraries may be useful for extension of sequence into 5' non-transcribed regulatory regions.
In one embodiment of the invention, the nucleic acid molecules of the present invention may be used for chromosome localisation. In this technique, a nucleic acid molecule is specifically targeted to, and can hybridize with, a particular location on an individual human chromosome. The mapping of relevant sequences to chromosomes according to the present invention is an important step in the confirmatory correlation of those sequences with the gene-associated disease. Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. Such data are found in, for example, V. McKusick, Mendelian Inheritance in Man (available on-line through Johns Hopkins University Welch Medical Library). The relationships between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance of physically adjacent genes). This provides valuable information to investigators searching for disease genes using positional cloning or other gene discovery techniques. Once the disease or syndrome has been crudely localised by genetic linkage to a particular genomic region, any sequences mapping to that area may represent associated or regulatory genes for further investigation. The nucleic acid molecule may also be used to detect differences in the chromosomal location due to translocation, inversion, etc. among normal, carrier, or affected individuals.
The nucleic acid molecules of the present invention are also valuable for tissue localisation. Such techniques allow the determination of expression patterns of the polypeptide in tissues by detection of the mRNAs that encode them. These techniques include in situ hybridization techniques and nucleotide amplification techniques, such as PCR. Results from these studies provide an indication of the normal functions of the polypeptide in the organism. In addition, comparative studies of the normal expression pattern of mRNAs with that of mRNAs encoded by a mutant gene provide valuable insights into the role of mutant polypeptides in disease. Such inappropriate expression may be of a temporal, spatial or quantitative nature.
Gene silencing approaches may also be undertaken to down-regulate endogenous expression of a gene encoding a polypeptide of the invention. RNA interference (RNAi) (Elbashir, SM et al, Nature 2001, 411, 494-498) is one method of sequence specific post- transcriptional gene silencing that may be employed. Short dsRNA oligonucleotides are synthesised in vitro and introduced into a cell. The sequence specific binding of these dsRNA oligonucleotides triggers the degradation of target mRNA, reducing or ablating target protein expression.
Efficacy of the gene silencing approaches assessed above may be assessed through the measurement of polypeptide expression (for example, by Western blotting), and at the RNA level using TaqMan-based methodologies.
The vectors of the present invention comprise nucleic acid molecules of the invention and may be cloning or expression vectors. The host cells of the invention, which may be transformed, transfected or transduced with the vectors of the invention may be prokaryotic or eukaryotic. The polypeptides of the invention may be prepared in recombinant form by expression of their encoding nucleic acid molecules in vectors contained within a host cell. Such expression methods are well known to those of skill in the art and many are described in detail by Sambrook et al (supra) and Fernandez & Hoeffler (1998, eds. "Gene expression systems. Using nature for the art of expression". Academic Press, San Diego, London, Boston, New York, Sydney, Tokyo, Toronto).
Generally, any system or vector that is suitable to maintain, propagate or express nucleic acid molecules to produce a polypeptide in the required host may be used. The appropriate nucleotide sequence may be inserted into an expression system by any of a variety of well- known and routine techniques, such as, for example, those described in Sambrook et al, (supra). Generally, the encoding gene can be placed under the control of a control element such as a promoter, ribosome binding site (for bacterial expression) and, optionally, an operator, so that the DNA sequence encoding the desired polypeptide is transcribed into RNA in the transformed host cell.
Examples of suitable expression systems include, for example, chromosomal, episomal and virus-derived systems, including, for example, vectors derived from: bacterial plasmids, bacteriophage, transposons, yeast episomes, insertion elements, yeast chromosomal elements, viruses such as baculoviruses, papova viruses such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, or combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, including cosmids and phagemids. Human artificial chromosomes (HACs) may also be employed to deliver larger fragments of DNA than can be contained and expressed in a plasmid.
Particularly suitable expression systems include microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (for example, baculovirus); plant cell systems transformed with virus expression vectors (for example, cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or with bacterial expression vectors (for example, Ti or pBR322 plasmids); or animal cell systems. Cell-free translation systems can also be employed to produce the polypeptides of the invention.
Introduction of nucleic acid molecules encoding a polypeptide of the present invention into host cells can be effected by methods described in many standard laboratory manuals, such as Davis et al, Basic Methods in Molecular Biology (1986) and Sambrook et al, (supra). Particularly suitable methods include calcium phosphate transfection, DEAE-dextran mediated transfection, transvection, microinjection, cationic lipid-mediated transfection, electroporation, transduction, scrape loading, ballistic introduction or infection (see Sambrook et al, 1989 [supra]; Ausubel et al, 1991 [supra]; Spector, Goldman & Leinwald, 1998). In eukaryotic cells, expression systems may either be transient (for example, episomal) or permanent (chromosomal integration) according to the needs of the system.
The encoding nucleic acid molecule may or may not include a sequence encoding a control sequence, such as a signal peptide or leader sequence, as desired, for example, for secretion of the translated polypeptide into the lumen of the endoplasmic reticulum, into the periplasmic space or into the extracellular environment. These signals may be endogenous to the polypeptide or they may be heterologous signals. Leader sequences can be removed by the bacterial host in post-translational processing.
In addition to control sequences, it may be desirable to add regulatory sequences that allow for regulation of the expression of the polypeptide relative to the growth of the host cell. Examples of regulatory sequences are those which cause the expression of a gene to be increased or decreased in response to a chemical or physical stimulus, including the presence of a regulatory compound or to various temperature or metabolic conditions. Regulatory sequences are those non-translated regions of the vector, such as enhancers, promoters and 5' and 3' untranslated regions. These interact with host cellular proteins to carry out transcription and translation. Such regulatory sequences may vary in their strength and specificity. Depending on the vector system and host utilised, any number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used. For example, when cloning in bacterial systems, inducible promoters such as the hybrid lacZ promoter of the Bluescript phagemid (Stratagene, LaJolla, CA) or pSportl™ plasmid (Gibco BRL) and the like may be used. The baculovirus polyhedrin promoter may be used in insect cells. Promoters or enhancers derived from the genomes of plant cells (for example, heat shock, RUBISCO and storage protein genes) or from plant viruses (for example, viral promoters or leader sequences) may be cloned into the vector. In mammalian cell systems, promoters from mammalian genes or from mammalian viruses are preferable. If it is necessary to generate a cell line that contains multiple copies of the sequence, vectors based on SV40 or EBV may be used with an appropriate selectable marker.
An expression vector is constructed so that the particular nucleic acid coding sequence is located in the vector with the appropriate regulatory sequences, the positioning and orientation of the coding sequence with respect to the regulatory sequences being such that the coding sequence is transcribed under the "control" of the regulatory sequences, i.e., RNA polymerase which binds to the DNA molecule at the control sequences transcribes the coding sequence. In some cases it may be necessary to modify the sequence so that it may be attached to the control sequences with the appropriate orientation; i.e., to maintain the reading frame.
The control sequences and other regulatory sequences may be ligated to the nucleic acid coding sequence prior to insertion into a vector. Alternatively, the coding sequence can be cloned directly into an expression vector that already contains the control sequences and an appropriate restriction site.
For long-term, high-yield production of a recombinant polypeptide, stable expression is preferred. For example, cell lines which stably express the polypeptide of interest may be transformed using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells may be allowed to grow for 1-2 days in an enriched media before they are switched to selective media. The purpose of the selectable marker is to confer resistance to selection, and its presence allows growth and recovery of cells that successfully express the introduced sequences. Resistant clones of stably transformed cells may be proliferated using tissue culture techniques appropriate to the cell type.
Mammalian cell lines available as hosts for expression are known in the art and include many immortalised cell lines available from the American Type Culture Collection (ATCC) including, but not limited to, Chinese hamster ovary (CHO), HeLa, baby hamster kidney (BHK), monkey kidney (COS), C127, 3T3, BHK, HEK 293, Bowes melanoma and human hepatocellular carcinoma (for example Hep G2) cells and a number of other cell lines.
In the baculovirus system, the materials for baculovirus/insect cell expression systems are commercially available in kit form from, inter alia, Invitrogen, San Diego CA (the "MaxBac" kit). These techniques are generally known to those skilled in the art and are described fully in Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987). Particularly suitable host cells for use in this system include insect cells such as Drosophila S2 and Spodoptera Sf9 cells. There are many plant cell culture and whole plant genetic expression systems known in the art. Examples of suitable plant cellular genetic expression systems include those described in US 5,693,506; US 5,659,122; and US 5,608,143. Additional examples of genetic expression in plant cell culture has been described by Zenk, Phytochemistry 30, 3861-3863 (1991).
In particular, all plants from which protoplasts can be isolated and cultured to give whole regenerated plants can be utilised, so that whole plants are recovered which contain the transferred gene. Practically all plants can be regenerated from cultured cells or tissues, including but not limited to all major species of sugar cane, sugar beet, cotton, fruit and other trees, legumes and vegetables.
Examples of particularly prefeπed bacterial host cells include streptococci, staphylococci, E. coli, Streptomyces and Bacillus subtilis cells. Examples of particularly suitable host cells for fungal expression include yeast cells (for example, S. cerevisiae) and Aspergillus cells.
Any number of selection systems are known in the art that may be used to recover transformed cell lines. Examples include the herpes simplex virus thymidine kinase (Wigler, M. et al. (1977) Cell 11:223-32) and adenine phosphoribosyltransferase (Lowy, I. et al. (1980) Cell 22:817-23) genes that can be employed in tk" or aprt* cells, respectively.
Also, antimetabolite, antibiotic or herbicide resistance can be used as the basis for selection; for example, dihydrofolate reductase (DHFR) that confers resistance to methotrexate (Wigler, M. et al. (1980) Proc. Natl. Acad. Sci. 77:3567-70); npt, which confers resistance to the aminoglycosides neomycin and G-418 (Colbere-Garapin, F. et al (1981) J. Mol. Biol. 150:1-14) and als or pat, which confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively. Additional selectable genes have been described, examples of which will be clear to those of skill in the art.
Although the presence or absence of marker gene expression suggests that the gene of interest is also present, its presence and expression may need to be confirmed. For example, if the relevant sequence is inserted within a marker gene sequence, transformed cells containing the appropriate sequences can be identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a sequence encoding a polypeptide of the invention under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well.
Alternatively, host cells that contain a nucleic acid sequence encoding a polypeptide of the invention and which express said polypeptide may be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA- DNA or DNA-RNA hybridizations and protein bioassays, for example, fluorescence activated cell sorting (FACS) or immunoassay techniques (such as the enzyme-linked immunosorbent assay [ELISA] and radioimmunoassay [RIA]), that include membrane, solution, or chip based technologies for the detection and/or quantification of nucleic acid or protein (see Hampton, R. et al. (1990) Serological Methods, a Laboratory Manual, APS Press, St Paul, MN) and Maddox, D.E. et al. (1983) J. Exp. Med, 158, 1211-1216).
A wide variety of labels and conjugation techniques are known by those skilled in the art and may be used in various nucleic acid and amino acid assays. Means for producing labelled hybridization or PCR probes for detecting sequences related to nucleic acid molecules encoding polypeptides of the present invention include oligolabelling, nick translation, end-labelling or PCR amplification using a labelled polynucleotide.
Alternatively, the sequences encoding the polypeptide of the invention may be cloned into a vector for the production of an mRNA probe. Such vectors are known in the art, are commercially available, and may be used to synthesise RNA probes in vitro by addition of an appropriate RNA polymerase such as T7, T3 or SP6 and labelled nucleotides. These procedures may be conducted using a variety of commercially available kits (Pharmacia &
Upjohn, (Kalamazoo, MI); Promega (Madison WI); and U.S. Biochemical Corp., Cleveland, OH)).
Suitable reporter molecules or labels, which may be used for ease of detection, include radionuclides, enzymes and fluorescent, chemiluminescent or chromogenic agents as well as substrates, cofactors, inhibitors, magnetic particles, and the like.
Nucleic acid molecules according to the present invention may also be used to create transgenic animals, particularly rodent animals. Such transgenic animals form a further aspect of the present invention. This may be done locally by modification of somatic cells, or by germ line therapy to incorporate heritable modifications. Such transgenic animals may be particularly useful in the generation of animal models for drug molecules effective as modulators of the polypeptides of the present invention. The polypeptide can be recovered and purified from recombinant cell cultures by well- known methods including ammonium sulphate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. High performance liquid chromatography is particularly useful for purification. Well known techniques for refolding proteins may be employed to regenerate an active conformation when the polypeptide is denatured during isolation and or purification.
Specialised vector constructions may also be used to facilitate purification of proteins, as desired, by joining sequences encoding the polypeptides of the invention to a nucleotide sequence encoding a polypeptide domain that will facilitate purification of soluble proteins. Examples of such purification-facilitating domains include metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilised metals, protein A domains that allow purification on immobilised immunoglobulin, and the domain utilised in the FLAGS extension/affinity purification system (Immunex Corp., Seattle, WA). The inclusion of cleavable linker sequences such as those specific for Factor XA or enterokinase (Invitrogen, San Diego, CA) between the purification domain and the polypeptide of the invention may be used to facilitate purification. One such expression vector provides for expression of a fusion protein containing the polypeptide of the invention fused to several histidine residues preceding a thioredoxin or an enterokinase cleavage site. The histidine residues facilitate purification by IMAC (immobilised metal ion affinity chromatography as described in Porath, J. et al. (1992), Prot. Exp. Purif. 3: 263-281) while the thioredoxin or enterokinase cleavage site provides a means for purifying the polypeptide from the fusion protein. A discussion of vectors which contain fusion proteins is provided in Kroll, D.J. et al. (1993; DNA Cell Biol. 12:441-453).
If the polypeptide is to be expressed for use in screening assays, generally it is preferred that it be produced at the surface of the host cell in which it is expressed. In this event, the host cells may be harvested prior to use in the screening assay, for example using techniques such as fluorescence activated cell sorting (FACS) or immunoaffinity techniques. If the polypeptide is secreted into the medium, the medium can be recovered in order to recover and purify the expressed polypeptide. If polypeptide is produced intracellularly, the cells must first be lysed before the polypeptide is recovered. The polypeptide of the invention can be used to screen libraries of compounds in any of a variety of drug screening techniques. Such compounds may activate (agonise) or inhibit (antagonise) the level of expression of the gene or the activity of the polypeptide of the invention and form a further aspect of the present invention. Preferred compounds are effective to alter the expression of a natural gene which encodes a polypeptide of the first aspect of the invention or to regulate the activity of a polypeptide of the first aspect of the invention. Agonist or antagonist compounds may be isolated from, for example, cells, cell-free preparations, chemical libraries or natural product mixtures. These agonists or antagonists may be natural or modified substrates, ligands, enzymes, receptors or structural or functional mimetics. For a suitable review of such screening techniques, see Coligan et al. , Current Protocols in Immunology l(2):Chapter 5 (1991). Compounds that are most likely to be good antagonists are molecules that bind to the polypeptide of the invention without inducing the biological effects of the polypeptide upon binding to it. Potential antagonists include small organic molecules, peptides, polypeptides and antibodies that bind to the polypeptide of the invention and thereby inhibit or extinguish its activity. In this fashion, binding of the polypeptide to normal cellular binding molecules may be inhibited, such that the normal biological activity of the polypeptide is prevented.
The polypeptide of the invention that is employed in such a screening technique may be free in solution, affixed to a solid support, borne on a cell surface or located intracellularly. In general, such screening procedures may involve using appropriate cells or cell membranes that express the polypeptide that are contacted with a test compound to observe binding, or stimulation or inhibition of a functional response. The functional response of the cells contacted with the test compound is then compared with control cells that were not contacted with the test compound. Such an assay may assess whether the test compound results in a signal generated by activation of the polypeptide, using an appropriate detection system. Inhibitors of activation are generally assayed in the presence of a known agonist and the effect on activation by the agonist in the presence of the test compound is observed.
A preferred method for identifying an agonist or antagonist compound of a polypeptide of the present invention comprises: (a) contacting a cell expressing on the surface thereof the polypeptide according to the first aspect of the invention, the polypeptide being associated with a second component capable of providing a detectable signal in response to the binding of a compound to the polypeptide, with a compound to be screened under conditions to permit binding to the polypeptide; and
(b) determining whether the compound binds to and activates or inhibits the polypeptide by measuring the level of a signal generated from the interaction of the compound with the polypeptide.
A further prefeπed method for identifying an agonist or antagonist of a polypeptide of the invention comprises:
(a) contacting a cell expressing on the surface thereof the polypeptide, the polypeptide being associated with a second component capable of providing a detectable signal in response to the binding of a compound to the polypeptide, with a compound to be screened under conditions to permit binding to the polypeptide; and
(b) determining whether the compound binds to and activates or inhibits the polypeptide by comparing the level of a signal generated from the interaction of the compound with the polypeptide with the level of a signal in the absence of the compound. In further prefeπed embodiments, the general methods that are described above may further comprise conducting the identification of agonist or antagonist in the presence of labelled or unlabelled ligand for the polypeptide.
In another embodiment of the method for identifying an agonist or antagonist of a polypeptide of the present invention comprises: determining the inhibition of binding of a ligand to cells which have a polypeptide of the invention on the surface thereof, or to cell membranes containing such a polypeptide, in the presence of a candidate compound under conditions to permit binding to the polypeptide, and determining the amount of ligand bound to the polypeptide. A compound capable of causing reduction of binding of a ligand is considered to be an agonist or antagonist. Preferably the ligand is labelled.
More particularly, a method of screening for a polypeptide antagonist or agonist compound comprises the steps of:
(a) incubating a labelled ligand with a whole cell expressing a polypeptide according to the invention on the cell surface, or a cell membrane containing a polypeptide of the invention, (b) measuring the amount of labelled ligand bound to the whole cell or the cell membrane; (c) adding a candidate compound to a mixture of labelled ligand and the whole cell or the cell membrane of step (a) and allowing the mixture to attain equilibrium;
(d) measuring the amount of labelled ligand bound to the whole cell or the cell membrane after step (c); and (e) comparing the difference in the labelled ligand bound in step (b) and (d), such that the compound which causes the reduction in binding in step (d) is considered to be an agonist or antagonist.
In certain of the embodiments described above, simple binding assays may be used, in which the adherence of a test compound to a surface bearing the polypeptide is detected by means of a label directly or indirectly associated with the test compound or in an assay involving competition with a labelled competitor. In another embodiment, competitive drug screening assays may be used, in which neutralising antibodies that are capable of binding the polypeptide specifically compete with a test compound for binding. In this manner, the antibodies can be used to detect the presence of any test compound that possesses specific binding affinity for the polypeptide.
Assays may also be designed to detect the effect of added test compounds on the production of mRNA encoding the polypeptide in cells. For example, an ELISA may be constructed that measures secreted or cell-associated levels of polypeptide using monoclonal or polyclonal antibodies by standard methods known in the art, and this can be used to search for compounds that may inhibit or enhance the production of the polypeptide from suitably manipulated cells or tissues. The formation of binding complexes between the polypeptide and the compound being tested may then be measured.
Assay methods that are also included within the terms of the present invention are those that involve the use of the genes and polypeptides of the invention in overexpression or ablation assays. Such assays involve the manipulation of levels of these genes/polypeptides in cells and assessment of the impact of this manipulation event on the physiology of the manipulated cells. For example, such experiments reveal details of signalling and metabolic pathways in which the particular genes/polypeptides are implicated, generate information regarding the identities of polypeptides with which the studied polypeptides interact and provide clues as to methods by which related genes and proteins are regulated.
Another technique for drug screening which may be used provides for high throughput screening of compounds having suitable binding affinity to the polypeptide of interest (see International patent application WO84/03564). In this method, large numbers of different small test compounds are synthesised on a solid substrate, which may then be reacted with the polypeptide of the invention and washed. One way of immobilising the polypeptide is to use non-neutralising antibodies. Bound polypeptide may then be detected using methods that are well known in the art. Purified polypeptide can also be coated directly onto plates for use in the aforementioned drug screening techniques.
The polypeptide of the invention may be used to identify membrane-bound or soluble receptors, through standard receptor binding techniques that are known in the art, such as ligand binding and crosslinking assays in which the polypeptide is labelled with a radioactive isotope, is chemically modified, or is fused to a peptide sequence that facilitates its detection or purification, and incubated with a source of the putative receptor (for example, a composition of cells, cell membranes, cell supernatants, tissue extracts, or bodily fluids). The efficacy of binding may be measured using biophysical techniques such as surface plasmon resonance (supplied by Biacore AB, Uppsala, Sweden) and spectroscopy. Binding assays may be used for the purification and cloning of the receptor, but may also identify agonists and antagonists of the polypeptide, that compete with the binding of the polypeptide to its receptor. Standard methods for conducting screening assays are well understood in the art. The invention also includes a screening kit useful in the methods for identifying agonists, antagonists, ligands, receptors, substrates, enzymes, that are described above.
The invention includes the agonists, antagonists, ligands, receptors, substrates and enzymes, and other compounds which modulate the activity or antigenicity of the polypeptide of the invention discovered by the methods that are described above. The invention also provides pharmaceutical compositions comprising a polypeptide, nucleic acid, ligand or compound of the invention in combination with a suitable pharmaceutical carrier. These compositions may be suitable as therapeutic or diagnostic reagents, as vaccines, or as other immunogenic compositions, as outlined in detail below.
According to the terminology used herein, a composition containing a polypeptide, nucleic acid, ligand or compound [X] is "substantially free of impurities [herein, Y] when at least
85% by weight of the total X+Y in the composition is X. Preferably, X comprises at least about 90% by weight of the total of X+Y in the composition, more preferably at least about 95%, 98% or even 99% by weight.
The pharmaceutical compositions should preferably comprise a therapeutically effective amount of the polypeptide, nucleic acid molecule, ligand, or compound of the invention. The term "therapeutically effective amount" as used herein refers to an amount of a therapeutic agent needed to treat, ameliorate, or prevent a targeted disease or condition, or to exhibit a detectable therapeutic or preventative effect. For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays, for example, of neoplastic cells, or in animal models, usually mice, rabbits, dogs, or pigs. The animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
The precise effective amount for a human subject will depend upon the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. This amount can be determined by routine experimentation and is within the judgement of the clinician. Generally, an effective dose will be from 0.01 mg/kg to 50 mg/kg, preferably 0.05 mg/kg to 10 mg/kg. Compositions may be administered individually to a patient or may be administered in combination with other agents, drugs or hormones. A pharmaceutical composition may also contain a pharmaceutically acceptable caπier, for administration of a therapeutic agent. Such carriers include antibodies and other polypeptides, genes and other therapeutic agents such as liposomes, provided that the carrier does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity. Suitable caπiers may be large, slowly metabolised macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers and inactive virus particles.
Pharmaceutically acceptable salts can be used therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulphates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. A thorough discussion of pharmaceutically acceptable carriers is available in Remington's Pharmaceutical Sciences (Mack Pub. Co., NJ. 1991). Pharmaceutically acceptable carriers in therapeutic compositions may additionally contain liquids such as water, saline, glycerol and ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such compositions. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for ingestion by the patient.
Once formulated, the compositions of the invention can be administered directly to the subject. The subjects to be treated can be animals; in particular, human subjects can be treated. The pharmaceutical compositions utilised in this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra- arterial, intramedullary, intrathecal, intraventricular, transdermal or transcutaneous applications (for example, see WO98/20734), subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, intravaginal or rectal means. Gene guns or hyposprays may also be used to administer the pharmaceutical compositions of the invention. Typically, the therapeutic compositions may be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared.
Direct delivery of the compositions will generally be accomplished by injection, subcutaneously, intraperitoneally, intravenously or intramuscularly, or delivered to the interstitial space of a tissue. The compositions can also be administered into a lesion. Dosage treatment may be a single dose schedule or a multiple dose schedule.
If the activity of the polypeptide of the invention is in excess in a particular disease state, several approaches are available. One approach comprises administering to a subject an inhibitor compound (antagonist) as described above, along with a pharmaceutically acceptable carrier in an amount effective to inhibit the function of the polypeptide, such as by blocking the binding of ligands, substrates, enzymes, receptors, or by inhibiting a second signal, and thereby alleviating the abnormal condition. Preferably, such antagonists are antibodies. Most preferably, such antibodies are chimeric and/or humanised to minimise their immunogenicity, as described previously.
In another approach, soluble forms of the polypeptide that retain binding affinity for the ligand, substrate, enzyme, receptor, in question, may be administered. Typically, the polypeptide may be administered in the form of fragments that retain the relevant portions.
In an alternative approach, expression of the gene encoding the polypeptide can be inhibited using expression blocking techniques, such as the use of antisense nucleic acid molecules (as described above), either internally generated or separately administered. Modifications of gene expression can be obtained by designing complementary sequences or antisense molecules (DNA, RNA, or PNA) to the control, 5' or regulatory regions (signal sequence, promoters, enhancers and introns) of the gene encoding the polypeptide. Similarly, inhibition can be achieved using "triple helix" base-pairing methodology. Triple helix pairing is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or regulatory molecules. Recent therapeutic advances using triplex DNA have been described in the literature (Gee, J.E. et al. (1994) In: Huber, B.E. and B.I. Can, Molecular and Immunologic Approaches, Futura Publishing Co., Mt. Kisco, NY). The complementary sequence or antisense molecule may also be designed to block translation of mRNA by preventing the transcript from binding to ribosomes. Such oligonucleotides may be administered or may be generated in situ from expression in vivo.
In addition, expression of the polypeptide of the invention may be prevented by using ribozymes specific to its encoding mRNA sequence. Ribozymes are catalytically active RNAs that can be natural or synthetic (see for example Usman, N, et al, Curr. Opin. Struct. Biol (1996) 6(4), 527-33). Synthetic ribozymes can be designed to specifically cleave mRNAs at selected positions thereby preventing translation of the mRNAs into functional polypeptide. Ribozymes may be synthesised with a natural ribose phosphate backbone and natural bases, as normally found in RNA molecules. Alternatively the ribozymes may be synthesised with non-natural backbones, for example, 2'-O-methyl RNA, to provide protection from ribonuclease degradation and may contain modified bases.
RNA molecules may be modified to increase intracellular stability and half-life. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5' and/or 3' ends of the molecule or the use of phosphorothioate or 2' O-methyl rather than phosphodiesterase linkages within the backbone of the molecule. This concept is inherent in the production of PNAs and can be extended in all of these molecules by the inclusion of non-traditional bases such as inosine, queosine and butosine, as well as acetyl-, methyl-, thio- and similarly modified forms of adenine, cytidine, guanine, thymine and uridine which are not as easily recognised by endogenous endonucleases.
For treating abnormal conditions related to an under-expression of the polypeptide of the invention and its activity, several approaches are also available. One approach comprises administering to a subject a therapeutically effective amount of a compound that activates the polypeptide, i.e., an agonist as described above, to alleviate the abnormal condition. Alternatively, a therapeutic amount of the polypeptide in combination with a suitable pharmaceutical carrier may be administered to restore the relevant physiological balance of polypeptide. Gene therapy may be employed to effect the endogenous production of the polypeptide by the relevant cells in the subject. Gene therapy is used to treat permanently the inappropriate production of the polypeptide by replacing a defective gene with a coπected therapeutic gene.
Gene therapy of the present invention can occur in vivo or ex vivo. Ex vivo gene therapy requires the isolation and purification of patient cells, the introduction of a therapeutic gene and introduction of the genetically altered cells back into the patient. In contrast, in vivo gene therapy does not require isolation and purification of a patient's cells.
The therapeutic gene is typically "packaged" for administration to a patient. Gene delivery vehicles may be non-viral, such as liposomes, or replication-deficient viruses, such as adenovirus as described by Berkner, K.L., in Cuπ. Top. Microbiol. Immunol., 158, 39-66 (1992) or adeno-associated virus (AAV) vectors as described by Muzyczka, N., in Cuπ. Top. Microbiol. Immunol., 158, 97-129 (1992) and U.S. Patent No. 5,252,479. For example, a nucleic acid molecule encoding a polypeptide of the invention may be engineered for expression in a replication-defective retroviral vector. This expression construct may then be isolated and introduced into a packaging cell transduced with a retroviral plasmid vector containing RNA encoding the polypeptide, such that the packaging cell now produces infectious viral particles containing the gene of interest. These producer cells may be administered to a subject for engineering cells in vivo and expression of the polypeptide in vivo (see Chapter 20, Gene Therapy and other Molecular Genetic-based Therapeutic Approaches, (and references cited therein) in Human Molecular Genetics (1996), T Strachan and A P Read, BIOS Scientific Publishers Ltd).
Another approach is the administration of "naked DNA" in which the therapeutic gene is directly injected into the bloodstream or muscle tissue.
In situations in which the polypeptides or nucleic acid molecules of the invention are disease-causing agents, the invention provides that they can be used in vaccines to raise antibodies against the disease causing agent. Vaccines according to the invention may either be prophylactic (ie. to prevent infection) or therapeutic (ie. to treat disease after infection). Such vaccines comprise immunising antigen(s), immunogen(s), polypeptide(s), protein(s) or nucleic acid, usually in combination with pharmaceutically-acceptable carriers as described above, which include any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition. Additionally, these carriers may function as immunostimulating agents ("adjuvants"). Furthermore, the antigen or immunogen may be conjugated to a bacterial toxoid, such as a toxoid from diphtheria, tetanus, cholera, H. pylori, and other pathogens.
Since polypeptides may be broken down in the stomach, vaccines comprising polypeptides are preferably administered parenterally (for instance, subcutaneous, intramuscular, intravenous, or intradermal injection). Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti- oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the recipient, and aqueous and non-aqueous sterile suspensions which may include suspending agents or thickening agents.
The vaccine formulations of the invention may be presented in unit-dose or multi-dose containers. For example, sealed ampoules and vials and may be stored in a freeze-dried condition requiring only the addition of the sterile liquid carrier immediately prior to use. The dosage will depend on the specific activity of the vaccine and can be readily determined by routine experimentation.
Genetic delivery of antibodies that bind to polypeptides according to the invention may also be effected, for example, as described in International patent application WO98/55607.
The technology refeπed to as jet injection (see, for example, www.powderject.com) may also be useful in the formulation of vaccine compositions.
A number of suitable methods for vaccination and vaccine delivery systems are described in International patent application WO00/29428.
This invention also relates to the use of nucleic acid molecules according to the present invention as diagnostic reagents. Detection of a mutated form of the gene characterised by the nucleic acid molecules of the invention which is associated with a dysfunction will provide a diagnostic tool that can add to, or define, a diagnosis of a disease, or susceptibility to a disease, which results from under-expression, over-expression or altered spatial or temporal expression of the gene. Individuals carrying mutations in the gene may be detected at the DNA level by a variety of techniques.
Nucleic acid molecules for diagnosis may be obtained from a subject's cells, such as from blood, urine, saliva, tissue biopsy or autopsy material. The genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR, ligase chain reaction (LCR), strand displacement amplification (SDA), or other amplification techniques (see Saiki et al, Nature, 324, 163-166 (1986); Bej, et al, Crit. Rev. Biochem. Molec. Biol., 26, 301-334 (1991); Birkenmeyer et al, J. Virol. Meth., 35, 117-126 (1991); Van Brunt, J., Bio/Technology, 8, 291-294 (1990)) prior to analysis.
In one embodiment, this aspect of the invention provides a method of diagnosing a disease in a patient, comprising assessing the level of expression of a natural gene encoding a polypeptide according to the invention and comparing said level of expression to a control level, wherein a level that is different to said control level is indicative of disease. The method may comprise the steps of: a)contacting a sample of tissue from the patient with a nucleic acid probe under stringent conditions that allow the formation of a hybrid complex between a nucleic acid molecule of the invention and the probe; b)contacting a control sample with said probe under the same conditions used in step a); c)and detecting the presence of hybrid complexes in said samples; wherein detection of levels of the hybrid complex in the patient sample that differ from levels of the hybrid complex in the control sample is indicative of disease.
A further aspect of the invention comprises a diagnostic method comprising the steps of: a)obtaining a tissue sample from a patient being tested for disease; b)isolating a nucleic acid molecule according to the invention from said tissue sample; and c)diagnosing the patient for disease by detecting the presence of a mutation in the nucleic acid molecule which is associated with disease.
To aid the detection of nucleic acid molecules in the above-described methods, an amplification step, for example using PCR, may be included. Deletions and insertions can be detected by a change in the size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to labelled RNA of the invention or alternatively, labelled antisense DNA sequences of the invention. Perfectly-matched sequences can be distinguished from mismatched duplexes by RNase digestion or by assessing differences in melting temperatures. The presence or absence of the mutation in the patient may be detected by contacting DNA with a nucleic acid probe that hybridises to the DNA under stringent conditions to form a hybrid double-stranded molecule, the hybrid double-stranded molecule having an unhybridised portion of the nucleic acid probe strand at any portion coπesponding to a mutation associated with disease; and detecting the presence or absence of an unhybridised portion of the probe strand as an indication of the presence or absence of a disease-associated mutation in the coπesponding portion of the DNA strand.
Such diagnostics are particularly useful for prenatal and even neonatal testing.
Point mutations and other sequence differences between the reference gene and "mutant" genes can be identified by other well-known techniques, such as direct DNA sequencing or single-strand conformational polymorphism, (see Orita et al, Genomics, 5, 874-879 (1989)). For example, a sequencing primer may be used with double-stranded PCR product or a single-stranded template molecule generated by a modified PCR. The sequence determination is performed by conventional procedures with radiolabelled nucleotides or by automatic sequencing procedures with fluorescent-tags. Cloned DNA segments may also be used as probes to detect specific DNA segments. The sensitivity of this method is greatly enhanced when combined with PCR. Further, point mutations and other sequence variations, such as polymorphisms, can be detected as described above, for example, through the use of allele-specific oligonucleotides for PCR amplification of sequences that differ by single nucleotides. DNA sequence differences may also be detected by alterations in the electrophoretic mobility of DNA fragments in gels, with or without denaturing agents, or by direct DNA sequencing (for example, Myers et al, Science (1985) 230:1242). Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and SI protection or the chemical cleavage method (see Cotton et al, Proc. Natl. Acad. Sci. USA (1985) 85: 4397-4401).
In addition to conventional gel electrophoresis and DNA sequencing, mutations such as microdeletions, aneuploidies, translocations, inversions, can also be detected by in situ analysis (see, for example, Keller et al, DNA Probes, 2nd Ed., Stockton Press, New York, N.Y., USA (1993)), that is, DNA or RNA sequences in cells can be analysed for mutations without need for their isolation and/or immobilisation onto a membrane. Fluorescence in situ hybridization (FISH) is presently the most commonly applied method and numerous reviews of FISH have appeared (see, for example, Trachuck et al, Science, 250, 559-562 (1990), and Trask et al, Trends, Genet., 7, 149-154 (1991)).
In another embodiment of the invention, an array of oligonucleotide probes comprising a nucleic acid molecule according to the invention can be constructed to conduct efficient screening of genetic variants, mutations and polymorphisms. Array technology methods are well known and have general applicability and can be used to address a variety of questions in molecular genetics including gene expression, genetic linkage, and genetic variability (see for example: M.Chee et al, Science (1996), Vol 274, pp 610-613).
In one embodiment, the aπay is prepared and used according to the methods described in PCT application WO95/11995 (Chee et al); Lockhart, D. J. et al. (1996) Nat. Biotech. 14: 1675-1680); and Schena, M. et al. (1996) Proc. Natl. Acad. Sci. 93: 10614-10619). Oligonucleotide pairs may range from two to over one million. The oligomers are synthesized at designated areas on a substrate using a light-directed chemical process. The substrate may be paper, nylon or other type of membrane, filter, chip, glass slide or any other suitable solid support. In another aspect, an oligonucleotide may be synthesized on the surface of the substrate by using a chemical coupling procedure and an ink jet application apparatus, as described in PCT application W095/251116 (Baldeschweiler et at). In another aspect, a "gridded" aπay analogous to a dot (or slot) blot may be used to arrange and link cDNA fragments or oligonucleotides to the surface of a substrate using a vacuum system, thermal, UV, mechanical or chemical bonding procedures. An aπay, such as those described above, may be produced by hand or by using available devices (slot blot or dot blot apparatus), materials (any suitable solid support), and machines (including robotic instruments), and may contain 8, 24, 96, 384, 1536 or 6144 oligonucleotides, or any other number between two and over one million which lends itself to the efficient use of commercially-available instrumentation.
In addition to the methods discussed above, diseases may be diagnosed by methods comprising determining, from a sample derived from a subject, an abnormally decreased or increased level of polypeptide or mRNA. Decreased or increased expression can be measured at the RNA level using any of the methods well known in the art for the quantitation of polynucleotides, such as, for example, nucleic acid amplification, for instance PCR, RT-PCR, RNase protection, Northern blotting and other hybridization methods. Assay techmques that can be used to determine levels of a polypeptide of the present invention in a sample derived from a host are well-known to those of skill in the art and are discussed in some detail above (including radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays). This aspect of the invention provides a diagnostic method which comprises the steps of: (a) contacting a ligand as described above with a biological sample under conditions suitable for the formation of a ligand- polypeptide complex; and (b) detecting said complex.
Protocols such as ELISA, RIA, and FACS for measuring polypeptide levels may additionally provide a basis for diagnosing altered or abnormal levels of polypeptide expression. Normal or standard values for polypeptide expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, preferably humans, with antibody to the polypeptide under conditions suitable for complex formation The amount of standard complex formation may be quantified by various methods, such as by photometric means.
Antibodies which specifically bind to a polypeptide of the invention may be used for the diagnosis of conditions or diseases characterised by expression of the polypeptide, or in assays to monitor patients being treated with the polypeptides, nucleic acid molecules, ligands and other compounds of the invention. Antibodies useful for diagnostic purposes may be prepared in the same manner as those described above for therapeutics. Diagnostic assays for the polypeptide include methods that utilise the antibody and a label to detect the polypeptide in human body fluids or extracts of cells or tissues. The antibodies may be used with or without modification, and may be labelled by joining them, either covalently or non-covalently, with a reporter molecule. A wide variety of reporter molecules known in the art may be used, several of which are described above.
Quantities of polypeptide expressed in subject, control and disease samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease. Diagnostic assays may be used to distinguish between absence, presence, and excess expression of polypeptide and to monitor regulation of polypeptide levels during therapeutic intervention. Such assays may also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies, in clinical trials or in monitoring the treatment of an individual patient.
A diagnostic kit of the present invention may comprise: (a) a nucleic acid molecule of the present invention;
(b) a polypeptide of the present invention; or
(c) a ligand of the present invention.
In one aspect of the invention, a diagnostic kit may comprise a first container containing a nucleic acid probe that hybridises under stringent conditions with a nucleic acid molecule according to the invention; a second container containing primers useful for amplifying the nucleic acid molecule; and instructions for using the probe and primers for facilitating the diagnosis of disease. The kit may further comprise a third container holding an agent for digesting unhybridised RNA.
In an alternative aspect of the invention, a diagnostic kit may comprise an aπay of nucleic acid molecules, at least one of which may be a nucleic acid molecule according to the invention.
To detect polypeptide according to the invention, a diagnostic kit may comprise one or more antibodies that bind to a polypeptide according to the invention; and a reagent useful for the detection of a binding reaction between the antibody and the polypeptide. Such kits will be of use in diagnosing a disease or susceptibility to disease in which ion channels are implicated, particularly cardiovascular disease, hypertension, heart arrhythmia, angina, myocardial infarction, tachycardia cerebrovascular disease, thromboembolism, neurological and psychiatric disorders, including stroke, epilepsy, migraine, pain, depression, neuralgia, Alzheimer's disease, brain injury, schizophrenia, Himtington's disease, motor neurone disease, Parkinson's disease, neuropathy, neuromuscular disorder, micturition disorder, impotence, diabetes, dermatitis, pulmonary disease, asthma, cystic fibrosis, mucous membrane disorders including sinusitis, COPD, rhinitis and cough, leukemia, ocular disease, glaucoma, retinopathy and immune disorders. Preferably, the diseases are those in which members of the PKD/REJ family of cation channels are implicated. Examples of such diseases include renal disease, polycystic kidney disease, glomerulnephritis, fertility disorders, infertility, reproductive disorders, polycystic ovary disease, polycystic diseases, endocrine diseases, testosterone deficiency, testosterone-related disorder, testicular cancer, prostate cancer, gastrointestinal disorders, developmental disorders, brain tumour, sexual dysfunction, psychological disorder, alopecia, hirsuitism, diseases related to muscle tone, including vascular muscle disorders such as migraine, pre-eclampsia, hypertension and cardiac aπhythmias; bladder muscle tone disorders, such as urinary incontinence, inability to void, urinary tract infection; gastrointestinal smooth muscle tone disorder, such as Gl motility disorder, gastrointestinal reflux diorder (GERD), reflux oesophagitis, paralytic ileus, irritable bowel syndrome (IBS), diaπhoea including travellers diarrhoea and viral diarrhoea, constipation and malabsorption; skeletal muscle tone disorder, such as spacticity, and muscle fatigue disorders such as myotonias, myasthenia gravis, floppy baby syndrome, ME, frailty of old age; lung disorders such as COPD, asthma, pulmonary hypertension; adrenal disorders, such as adrenal hypertension, Conn's syndrome, Cushing disease and Addison's disease. Various aspects and embodiments of the present invention will now be described in more detail by way of example, with particular reference to the INPIONCH03 and INPIONCH04 polypeptides.
It will be appreciated that modification of detail may be made without departing from the scope of the invention.
Brief description of the Figures Figure 1: PKD/REJ cation channel family domain/topology diagram highlighting the conserved features.
Figure 2: Top ten results from BLAST against NCBI non-redundant database using SEQ ID NO:56 (INPIONCH03 polypeptide sequence).
Figure 3: Alignment generated by BLAST between SEQ ID NO:56 (INPIONCH03 polypeptide sequence) and the top biochemically annotated hit, the receptor for egg jelly 3 protein (REJ3).
Figure 4: Conserved domain search using the INPIONCH03 query sequence. Figure 5: INPIONCH03 topology prediction using the TMHMM transmembrane domain prediction programme
Figure 6: Top ten results from BLAST against NCBI non-redundant database using SEQ ID NO: 108 (INPIONCH04 polypeptide sequence). Figure 7: Alignment generated by BLAST between SEQ ID NO: 108 (INPIONCH04 polypeptide sequence) and the top biochemically annotated hit, the receptor for egg jelly 3 protein (REJ3).
Figure 8: Conserved domain search using the INPIONCH04 query sequence.
Figure 9: INPIONCH04 topology prediction using the TMHMM transmembrane domain prediction programme
Figure 10: Normalised expression of INPIONCH03 in 22 samples derived from 18 normal human tissues.
Figure 11: Normalised expression of INPIONCH04 in 22 samples derived from 18 normal human tissues.
Examples
Example 1 : INPIONCH03 Protein BLAST Results
The INPIONCH03 polypeptide sequence, shown in SEQ ID NO:56, was used as a BLAST query against the NCBI non-redundant Sequence database. Several of the top ten matches are members of the PKD/REJ family of cation channels. The top ten hits all align to the query sequence with highly significant E-values (8e"39 to 0.0) (Figure 2). Figure 3 shows the alignment of the INPIONCH03 polypeptide query sequence to the sequence of the top biochemically annotated hit, the Strongylocentrotus purpuratus receptor for egg jelly 3 protein (REJ3). These results indicate that the INPIONCH03 polypeptide is a member of the PKD/REJ family of cation channels.
Example 2: INPIONCH03 Conserved Domain Search Results
The INPIONCH03 polypeptide sequence (SEQ ID NO: 56) was analysed against the amino acid sequences of conserved domain in the Pfam and SMART public domain databases.
Figure 4 shows the results of this conserved domain search. The INPIONCH03 query sequence returns matches to the PLAT (or LH2) domain, an ion transport-associated domain and the latrophilin/CL-1 like GPS domain. The results of the conserved domain search therefore confirm that the INPIONCH03 polypeptide is a member of the PKD/REJ family of cation channels, as shown in Example 1 above.
Example 3: INPIONCH03 Topology Information from TMHMM Prediction Programme The INPIONCH03 polypeptide sequence (SEQ ID NO: 56) was analysed using the TMHMM (transmembrane hidden Markov model) programme for prediction of transmembrane domains. The TMHMM programme predicts over 75% of transmembrane topologies coπectly and is therefore a reliable tool for investigating protein topology (Krogh et al, Journal of Molecular Biology, 2001, 305(3):567-580). The results of the TMHMM analysis of INPIONCH03 are shown in Figure 5. Figure 5 shows that the TMHMM confidently predicts that the INPIONCH03 polypeptide contains eleven transmembrane domains, in accordance with other members of the PKD/REJ family of cation channels. In combination with the BLAST results described in Example 1, and the conserved domain search results described in Example 2, this topology prediction therefore provides further evidence that the INPIONCH03 polypeptide is a member of the PKD/REJ family of cation channels. Example 4: INPIONCH04 Protein BLAST Results
The INPIONCH04 polypeptide sequence, shown in SEQ ID NO: 108, was used as a BLAST query against the NCBI non-redundant Sequence database. Several of the top ten matches are members of the PKD/REJ family of cation channels. The top ten hits all align to the query sequence with highly significant E-values (2e' to e" ) (Figure 6). Figure 7 shows the alignment of the INPIONCH04 polypeptide query sequence to the sequence of the top biochemically annotated hit, the Strongylocentrotus purpuratus receptor for egg jelly 3 protein (REJ3). These results indicate that the INPIONCH04 polypeptide is a member of the PKD/REJ family of cation channels.
Example 5: INPIONCH04 Conserved Domain Search Results
The INPIONCH04 polypeptide sequence (SEQ ID NO: 108) was analysed against the amino acid sequences of conserved domain in the Pfam and SMART public domain databases. Figure 8 shows the results of this conserved domain search. The INPIONCH04 query sequence returns matches to the PLAT (or LH2) domain, an ion transport-associated domain and the latrophilin/CL-1 like GPS domain. The results of the conserved domain search therefore confirm that the INPIONCH04 polypeptide is a member of the PKD/REJ family of cation channels, as shown in Example 4 above.
Example 6: INPIONCH04 Topology Information from TMHMM Prediction Programme
The INPIONCH04 polypeptide sequence (SEQ ID NO: 108) was analysed using the TMHMM (transmembrane hidden Markov model) programme for prediction of transmembrane domains. The TMHMM programme predicts over 75% of transmembrane topologies coπectly and is therefore a reliable tool for investigating protein topology (Krogh et al., Journal of Molecular Biology, 2001, 3O5(3):567-580). The results of the TMHMM analysis of INPIONCH04 are shown in Figure 9. Figure 9 shows that the TMHMM confidently predicts that the INPIONCH04 polypeptide contains eleven transmembrane domains, in accordance with other members of the PKD/REJ family of cation channels. In combination with the BLAST results described in Example 4, and the conserved domain search results described in Example 5, this topology prediction therefore provides further evidence that the INPIONCH04 polypeptide is a member of the PKD/REJ family of cation channels.
Example 7: INPIONCH03 and INPIONCH04 Cloning
In order to determine the tissue expression of the proposed cation channel, Taqman RT- PCR quantitation was used. The TaqMan 3'- 5' exonuclease assay signals the formation of PCR amplicons by a process involving the nucleolytic degradation of a double labelled fluorogenic probe that hybridises to the target template at a site between the two primer recognition sequences (cf. U. S. Patent 5,876,930). The ABI Prism 7700 automates the detection and quantitative measurement of these signals, which are stoichiometrically related to the quantities of amplicons produced, during each cycle of amplification. In addition to providing substantial reductions in the time and labour requirements for PCR analyses, this technology permits simplified and potentially highly accurate quantification of target sequences in the reactions.
Figure 10 shows normalised expression of INPIONCH03 in 22 samples derived from 18 normal human tissues. Figure 11 shows normalised expression of INPIONCH04 in 22 samples derived from 18 normal human tissues.
Taqman RT-PCR was carried out using 15ng of the indicated cDNA using primers/probes specific for INPIONCH03 and 18s rRNA as described in the detailed description. A standard curve for target and internal control was also carried out, using between 25ng to 0.39ng of cDNA template of a typical tissue sample. Cycle threshold (Ct) determinations, i.e. non-integer calculations of the number of cycles required for reporter dye fluorescence resulting from the synthesis of PCR products to become significantly higher than background fluorescence levels were performed by the instrument for each reaction using default parameters. Using linear regression analysis of the standard curves, the Ct values were used to calculate the amount of actual starting target or 18s cDNA in each test sample.
The levels of target cDNA in each sample were normalised to the level of expression of target in a comparative sample, in the case of INPIONCH03, heart and in the case of
INPIONCH04, testis. The levels of 18s cDNA in each sample were also normalised to the level of expression of 18s in heart and testis, respectively. The expression levels of
INPIONCH03 and INPIONCH04 were then normalised to the expression levels of 18s.
Figures 10 and 11 represent the fold expression of normalised target sequence relative to the level of expression in heart or testis cDNA, which is set arbitrarily to 1. Each sample was quantitated in 2 individual experiments. Figures 10 and 11 show the mean ± SEM for the multiple experiments.
In the case of INPIONCH03, the primer/probe set recognises a sequence spanning a proposed exon-exon boundary within the INPIONCH03 coding sequence. The PCR reaction was also carried out on cDNA made in the absence of reverse transcriptase enzyme. No signal was seen in these reactions (data not shown), indicating that results shown in Figure 10 are not affected by genomic contamination of the RNA sample and that the region of INPIONCH03 detected is present in cDNA.
In the case of INPIONCH04, the primer/probe set recognises a sequence within a proposed exon within the INPIONCH04 coding sequence. The PCR reaction was also carried out on cDNA made in the absence of reverse transcriptase enzyme. A signal was seen in these reactions (data not shown), this was quantified and subtracted from the quantity calculated from the signal seen with cDNA synthesized in the presence of reverse transcriptase. The results shown in Figure 11 are coπected for genomic contamination of the RNA sample and indicate that the region of INPIONCH04 detected is present in cDNA. RNA samples
Human RNA prepared from non-diseased organs was purchased from either Ambion Europe (Huntingdon, UK) or Clontech (BD, Franklin Lakes, NJ).
Oligo Design
Oligonucleotide primers and probes were designed using Primer Express software (Applied Biosystems, Foster City CA) with a GC-content of 40-60%, no G-nucleotide at the 5'-end of the probe, and no more than 4 contiguous Gs.
Each primer and probe was analysed using BLAST (Basic Local Alignment Search Tool, Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. J Mol Biol 1990 Oct 5;215(3):403-10). Results confirmed that each oligonucleotide recognises the target sequence with a specificity >3 bp when compared to other known cDNA's or genomic sequence represented in the Unigene and GoldenPath publicly available databases.
The sequences of the primers and probes used were:
INPIONCH03 Fwd GCTCATCTACTTCCTCTTCCTCCTCTA
INPIONCH03 Probe CGCTTGCCCTGCACCACCATGTA
INPIONCH03 Rev ATAGCCCCACGTCTCTTTACTCAT
INPIONCH04 Fwd CCCTGTTGATGACTGCAATCTACT
INPIONCH04 Probe TGCAAAGAACTCCAATAGATTTTACCTCCACCA
INPIONCH04 Rev TGGTGCGAAAATGTCTTCCA
18s pre-optimised primers and probe were purchased from Applied Biosystems, Foster City, CA.
Probes were covalently conjugated with a fluorescent reporter dye (e.g. 6carboxy- fluorescein [FAM]; Xem = 518nm) and a fluorescent quencher dye (6carboxytetram-ethyl- rhodamine [TAMRA]; Mem = 582nm) at the most 5' and most 3' base, respectively. Primers were obtained from Sigma Genosys, UK and probes were obtained from Eurogentec, Belgium. Primer/probe concentrations were titrated in the range of 50nM to 900nM and optimal concentrations for efficient PCR reactions were determined. Optimal primer and probe concentrations vary in between 1 OOnM and 900nM depending on the target gene that is amplified.
cDNA reaction
cDNA was prepared using components from Applied Biosystems, Foster City CA. 50μl reactions were prepared in 0.5ml RNase free tubes. Reactions contain 500ng total RNA; lx reverse transcriptase buffer; 5.5mM MgC12; ImM dNTP's; 2.5μl random hexamers; 20U RNase inhibitor; and 62.5U reverse transcriptase.
PCR reactions
25μl reactions were prepared in 0.5 ml thin-walled, optical grade PCR 96 well plates (Applied Biosystems, Foster City CA). Reactions contained: lx final concentration of TaqMan Universal Master Mix (a proprietary mixture of AmpliTaq Gold DNA polymerase, AmpEraseX UNG, dNTPs with UTP, passive reference dye and optimised buffer components, Applied Biosystems, Foster City CA); lOOnM Taqman probe; 900nM forward primer; 900nM reverse primer and 15ng of cDNA template.
Performance of Assay
Standard procedures for the operation of the ABI Prism 7700 or similar detection system were used. This includes, for example with the ABI Prism 7700, use of all default program settings with the exception of reaction volume which is changed from 50 to 25 ul. Thermal cycling conditions consist of two min at 50°C, 10 min at 95°C, followed by 40 cycles of 15 sec at 95°C and 1 min at 60 C. Cycle threshold (Ct) determinations, i.e. non-integer calculations of the number of cycles required for reporter dye fluorescence resulting from the synthesis of PCR products to become significantly higher than background fluorescence levels were automatically performed by the instrument for each reaction using default parameters. Assays for target sequences and ribosomal 18s (reference) sequences in the same cDNA samples were performed in separate reaction tubes. Within each experiment, a standard curve was carried out on a typical tissue sample, from 25ng to 0.39ng of cDNA template. From this standard curve, the amount of actual starting target or 18s cDNA in each test sample was determined.
As described above, the levels of target cDNA in each sample were normalised to the level of expression of target in a comparative sample. The levels of internal control cDNA in each sample were normalised to the level of expression of internal control in a comparative sample. The data was then represented as fold expression of normalised target sequence relative to the level of expression in the comparative sample, which is set arbitrarily to 1.
In summary the data provided in this example show that the INPIONCH03 and INPIONCH04 polypeptides are expressed in a variety of human tissues. High levels of INPIONCH03 are expressed in brain, cervix, placenta, bladder, testis, small intestine and heart. The INPIONCH04 protein shows widespread expression, with highest levels in testis and high levels also found in brain, skeletal muscle, ovary, lung and adrenal tissues. Experiments will now be undertaken to verify the full-length transcripts of the INPIONCH03 and INPIONCH04 polypeptides.

Claims

1. A polypeptide, which polypeptide:
(i) comprises the amino acid sequence as recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID
NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52 and/or SEQ ID NO:54; (ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or having an antigenic determinant in common with the polypeptide of (i); or
(iii) is a functional equivalent of (i) or (ii).
2. A polypeptide according to claim 1 which: (i) comprises the amino acid sequence as recited in SEQ ED NO:56;
(ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or having an antigenic determinant in common with the polypeptide of (i); or
(iii) is a functional equivalent of (i) or (ii).
3. A polypeptide according to claim 1 or 2 which:
(i) consists of the amino acid sequence as recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42,
SEQ ID NO:44, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52, SEQ ID NO:54 and/or SEQ ID NO:56;
(ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or having an antigenic determinant in common with the polypeptide of (i); or (iii) is a functional equivalent of (i) or (ii).
4. A polypeptide, which polypeptide:
(i) comprises the amino acid sequence as recited in SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72, SEQ ID NO:74, SEQ ID NO:76, SEQ ID NO:78, SEQ ID NO:80,
SEQ ID NO:82, SEQ ID NO:84, SEQ ID NO:86, SEQ ID NO:88, SEQ ID NO:90, SEQ ID NO:92, SEQ ID NO:94, SEQ ID NO:96, SEQ ID NO:98, SEQ ID NO: 100, SEQ ID NO: 102, SEQ ID NO: 104 and/or SEQ ID NO: 106;
(ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or having an antigenic determinant in common with the polypeptide of (i); or
(iii) is a functional equivalent of (i) or (ii).
5. A polypeptide according to claim 4 which:
(i) comprises the amino acid sequence as recited in SEQ ID NO: 108; (ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or having an antigenic determinant in common with the polypeptide of (i); or
(iii) is a functional equivalent of (i) or (ii).
6. A polypeptide according to claim 4 or 5 which: (i) consists of the amino acid sequence as recited in SEQ ID NO:58, SEQ ID
NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72, SEQ ID NO:74, SEQ ID NO:76, SEQ ID NO:78, SEQ ID NO:80, SEQ ID NO:82, SEQ ID NO:84, SEQ ID NO:86, SEQ ID NO:88, SEQ ID NO:90, SEQ ID NO:92, SEQ ID NO:94, SEQ ID NO:96, SEQ ID NO:98, SEQ ID NO: 100, SEQ ID NO: 102, SEQ ID NO: 104, SEQ ID NO: 106 and/or SEQ ID
NO: 108;
(ii) is a fragment thereof which is a member of the PKD/REJ family of cation channels, or having an antigenic determinant in common with the polypeptide of (i); or (iii) is a functional equivalent of (i) or (ii).
7. A polypeptide which is a functional equivalent according to part (iii) of any of the above claims, characterised in that it is homologous to the amino acid sequence as recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO:12, SEQ ID NO-14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52, SEQ ID NO:54, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72, SEQ ID NO:74, SEQ ID NO:76, SEQ ID NO:78, SEQ ID NO:80, SEQ ID NO:82, SEQ ID NO:84, SEQ ID NO:86, SEQ ID NO:88, SEQ ID NO:90, SEQ ID NO:92, SEQ ID NO:94, SEQ ID NO:96, SEQ ID NO:98, SEQ ID NO: 100, SEQ ID NO: 102, SEQ ID NO: 104, SEQ ID NO: 106 or SEQ ID NO: 108, and is a member of the PKD/REJ family of cation channels.
8. A polypeptide which is a fragment or a functional equivalent as recited in any one of claims 1 to 7, which has greater than 80% sequence identity with the amino acid sequence recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52, SEQ ID NO:54, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72, SEQ ID NO:74, SEQ ID NO:76, SEQ ID NO:78, SEQ ID NO:80, SEQ ID NO:82, SEQ ID NO:84, SEQ ID NO:86, SEQ ID NO:88, SEQ ID NO:90, SEQ ID NO:92, SEQ ID NO:94, SEQ ID NO:96, SEQ ID NO:98, SEQ ID NO: 100, SEQ ID NO: 102, SEQ ID NO: 104, SEQ ID NO: 106 or SEQ ID NO: 108, or with an active fragment thereof, preferably greater than 85%, 90%, 95%, 98% or 99% sequence identity.
9. A polypeptide which is a functional equivalent as recited in any one of claims 1 to 8, which exhibits significant structural homology with a polypeptide having the amino acid sequence recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ DO NO:8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52, SEQ ID NO:54, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72, SEQ ID NO:74, SEQ ID NO:76, SEQ ID NO:78, SEQ ID NO:80, SEQ ID NO:82, SEQ ID NO:84, SEQ ID NO:86, SEQ ID NO:88, SEQ ID NO:90, SEQ ID NO:92, SEQ ID NO:94, SEQ ID NO:96, SEQ ID NO:98, SEQ ID NO: 100, SEQ ID NO: 102, SEQ ID NO: 104, SEQ ID NO:106 or SEQ ID NO:108.
10. A polypeptide which is a fragment as recited in claims 1-6 and claim 8 having an antigenic determinant in common with the polypeptide of part (i) of any one of claim 1 to claim 6 which consists of 7 or more amino acid residues from the amino acid sequence recited in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:22, SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:28, SEQ ID NO:30, SEQ ID NO:32, SEQ ID NO:34, SEQ ID NO:36, SEQ ID NO:38, SEQ ID NO:40, SEQ ID NO:42, SEQ ID NO:44, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:50, SEQ ID NO:52, SEQ ID NO:54, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:60, SEQ ID NO:62, SEQ ID NO:64, SEQ ID NO:68, SEQ ID NO:70, SEQ ID NO:72, SEQ ID NO:74, SEQ ID NO:76, SEQ ID NO:78, SEQ ID NO:80, SEQ ID NO:82, SEQ ID NO:84, SEQ ID NO:86, SEQ ID NO:88, SEQ ID NO:90, SEQ ID NO:92, SEQ ID NO:94, SEQ ID NO:96, SEQ ID NO:98, SEQ ID NO: 100, SEQ ID NO: 102, SEQ ID NO: 104, SEQ ID NO: 106 or SEQ ID NO: 108.
11. A purified nucleic acid molecule which encodes a polypeptide according to any one of the preceding claims.
12. A purified nucleic acid molecule according to claim 11, which comprises the nucleic acid sequence as recited in SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l l, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:25, SEQ ID NO:27, SEQ ID NO:29, SEQ ID NO:31, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:37, SEQ ID NO:39, SEQ ID NO:41, SEQ ID NO:43, SEQ ID NO:45, SEQ ID NO:47, SEQ ID NO:49, SEQ ID NO:51, SEQ ID NO:53, SEQ ID NO:55, SEQ ID NO:57, SEQ ID NO:59, SEQ ID NO:61, SEQ ID NO:63, SEQ ID NO:65, SEQ ID NO:67, SEQ ID NO:69, SEQ ID NO-71, SEQ ID NO:73, SEQ ID NO:75, SEQ ID NO:79, SEQ ID NO:81, SEQ ID NO:83, SEQ ID NO:85, SEQ ID NO:87, , SEQ ID NO:89, SEQ ID NO:91, SEQ ID NO:93, SEQ ID NO:95, SEQ ID NO:97, SEQ ID NO:99, SEQ ID NO:101, SEQ ID NO:103, SEQ ID NO:105 and/or SEQ ID NO:107, or is a redundant equivalent or fragment thereof.
13. A purified nucleic acid molecule according to claim 11 which consists of the nucleic acid sequence as recited in SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l l, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO:21, SEQ ID NO:23, SEQ ID NO:25, SEQ ID
NO:27, SEQ ID NO:29, SEQ ID NO-31, SEQ ID NO:33, SEQ ID NO:35, SEQ ID NO:37, SEQ ID NO:39, SEQ ID NO:41, SEQ ID NO:43, SEQ ID NO:45, SEQ ID NO:47, SEQ ID NO:49, SEQ ID NO:51, SEQ ID NO:53, SEQ ID NO:55, SEQ ID NO:57, SEQ ID NO:59, SEQ ID NO:61, SEQ ID NO:63, SEQ ID NO:65, SEQ ID NO:67, SEQ ID NO:69, SEQ ID NO:71, SEQ ID NO:73, SEQ ID NO:75, SEQ ID
NO:79, SEQ ID NO:81, SEQ ID NO:83, SEQ ID NO:85, SEQ ID NO:87, , SEQ ID NO:89, SEQ ID NO:91, SEQ ID NO:93, SEQ ID NO:95, SEQ ID NO:97, SEQ ID NO:99, SEQ ID NO:101, SEQ ID NO:103, SEQ ID NO:105 and/or SEQ ID NO:107, or is a redundant equivalent or fragment thereof.
14. A purified nucleic acid molecule which hybridizes under high stringency conditions with a nucleic acid molecule according to any one of claims 11 to 13.
15. A vector comprising a nucleic acid molecule as recited in any one of claims 11 to 14.
16. A host cell transformed with a vector according to claim 15.
17. A ligand which binds specifically to the PKD/REJ cation channel family polypeptide according to any one of claims 1 to 10.
18. A ligand according to claim 17, which is an antibody.
19. A compound that either increases or decreases the level of expression or activity of a polypeptide according to any one of claims 1 to 10.
20. A compound according to claim 19 that binds to a polypeptide according to any one of claims 1 to 10 without inducing any of the biological effects of the polypeptide.
21. A compound according to claim 20, which is a natural or modified substrate, ligand, enzyme, receptor or structural or functional mimetic.
22. A polypeptide according to any one of claims 1 to 10, a nucleic acid molecule according to any one of claims 11 to 14, a vector according to claim 15, a ligand according to claim 17 or claim 18, or a compound according to any one of claims 19 to 21 , for use in therapy or diagnosis of disease.
23. A polypeptide according to any one of claims 1 to 10, a nucleic acid molecule according to any one of claims 11 to 14, a vector according to claim 15, a ligand according to claim 17 or claim 18, or a compound according to any one of claims 19 to 21, for use as a contraceptive agent.
24. A method of diagnosing a disease in a patient, comprising assessing the level of expression of a natural gene encoding a polypeptide according to any one of claims 1 to 10, or assessing the activity of a polypeptide according to any one of claims 1 to 10, in tissue from said patient and comparing said level of expression or activity to a control level, wherein a level that is different to said control level is indicative of disease.
25. A method according to claim 24 that is carried out in vitro.
26. A method according to claim 24 or claim 25, which comprises the steps of: (a) contacting a ligand according to claim 17 or claim 18 with a biological sample under conditions suitable for the formation of a ligand-polypeptide complex; and (b) detecting said complex.
27. A method according to claim 24 or claim 25, comprising the steps of: a) contacting a sample of tissue from the patient with a nucleic acid probe under stringent conditions that allow the formation of a hybrid complex between a nucleic acid molecule according to any one of claims 11 to 14 and the probe; b) contacting a control sample with said probe under the same conditions used in step a); and c) detecting the presence of hybrid complexes in said samples; wherein detection of levels of the hybrid complex in the patient sample that differ from levels of the hybrid complex in the control sample is indicative of disease.
28. A method according to claim 24 or claim 25, comprising: a)contacting a sample of nucleic acid from tissue of the patient with a nucleic acid primer under stringent conditions that allow the formation of a hybrid complex between a nucleic acid molecule according to any one of claims 11 to 14 and the primer; b)contacting a control sample with said primer under the same conditions used in step a); and c)amplifying the sampled nucleic acid; and d)detecting the level of amplified nucleic acid from both patient and control samples; wherein detection of levels of the amplified nucleic acid in the patient sample that differ significantly from levels of the amplified nucleic acid in the control sample is indicative of disease.
29. A method according to claim 24 or claim 25 comprising: a)obtaining a tissue sample from a patient being tested for disease; b)isolating a nucleic acid molecule according to any one of claims 11 to 14 from said tissue sample; and c)diagnosing the patient for disease by detecting the presence of a mutation which is associated with disease in the nucleic acid molecule as an indication of the disease.
30. The method of claim 29, further comprising amplifying the nucleic acid molecule to form an amplified product and detecting the presence or absence of a mutation in the amplified product.
31. The method of claim 29 or claim 30, wherein the presence or absence of the mutation in the patient is detected by contacting said nucleic acid molecule with a nucleic acid probe that hybridises to said nucleic acid molecule under stringent conditions to form a hybrid double-stranded molecule, the hybrid double-stranded molecule having an unhybridised portion of the nucleic acid probe strand at any portion coπesponding to a mutation associated with disease; and detecting the presence or absence of an unhybridised portion of the probe strand as an indication of the presence or absence of a disease-associated mutation.
32. A method according to any one of claims 24 to 31, wherein said disease is selected from the group consisting of cardiovascular disease, hypertension, heart aπhythmia, angina, myocardial infarction, tachycardia cerebrovascular disease, thromboembolism, neurological and psychiatric disorders, including stroke, epilepsy, migraine, pain, depression, neuralgia, Alzheimer's disease, brain injury, schizophrenia, Huntington's disease, motor neurone disease, Parkinson's disease, neuropathy, neuromuscular disorder, micturition disorder, impotence, diabetes, dermatitis, pulmonary disease, asthma, cystic fibrosis, mucous membrane disorders including sinusitis, COPD, rhinitis and cough, leukemia, ocular disease, glaucoma, retinopathy and immune disorders.
33. A method according to any one of claims 24 to 31, wherein said disease is selected from the group consisting of renal disease, polycystic kidney disease, glomerulnephritis, fertility disorders, infertility, reproductive disorders, polycystic ovary disease, polycystic diseases, endocrine diseases, testosterone deficiency, testosterone-related disorder, testicular cancer, prostate cancer, gastrointestinal disorders, developmental disorders, brain tumour, sexual dysfunction, psychological disorder, alopecia, hirsuitism, diseases related to muscle tone, including vascular muscle disorders such as migraine, pre-eclampsia, hypertension and cardiac aπhythmias; bladder muscle tone disorders, such as urinary incontinence, inability to void, urinary tract infection; gastrointestinal smooth muscle tone disorder, such as Gl motility disorder, gastrointestinal reflux diorder (GERD), reflux oesophagitis, paralytic ileus, irritable bowel syndrome (IBS), diarrhoea including travellers diaπhoea and viral diaπhoea, constipation and malabsorption; skeletal muscle tone disorder, such as spacticity, and muscle fatigue disorders such as myotonias, myasthenia gravis, floppy baby syndrome, ME, frailty of old age; lung disorders such as COPD, asthma, pulmonary hypertension; adrenal disorders, such as adrenal hypertension, Conn's syndrome, Cushing disease and Addison's disease.
34. Use of a polypeptide according to any one of claims 1 to 10 as a cation channel.
35. A pharmaceutical composition comprising a polypeptide according to any one of claims 1 to 10, a nucleic acid molecule according to any one of claims 11 to 14, a vector according to claim 15, a host cell according to claim 16, a ligand according to claim 17 or claim 18, or a compound according to any one of claims 19 to 21.
36. A vaccine composition comprising a polypeptide according to any one of claims 1 to 10 or a nucleic acid molecule according to any one of claims 11 to 14.
37. A polypeptide according to any one of claims 1 to 10, a nucleic acid molecule according to any one of claims 11 to 14, a vector according to claim 15, a host cell according to claim 16, a ligand according to claim 17 or claim 18, a compound according to any one of claims 19 to 21, or a pharmaceutical composition according to claim 35, for use in the manufacture of a medicament for the treatment of cardiovascular disease, hypertension, heart arrhythmia, angina, myocardial infarction, tachycardia cerebrovascular disease, thromboembolism, neurological and psychiatric disorders, including stroke, epilepsy, migraine, pain, depression, neuralgia, Alzheimer's disease, brain injury, schizophrenia, Himtington's disease, motor neurone disease, Parkinson's disease, neuropathy, neuromuscular disorder, micturition disorder, impotence, diabetes, dermatitis, pulmonary disease, asthma, cystic fibrosis, mucous membrane disorders including sinusitis, COPD, rhinitis and cough, leukemia, ocular disease, glaucoma, retinopathy and immune disorders.
38. A polypeptide according to any one of claims 1 to 10, a nucleic acid molecule according to any one of claims 11 to 14, a vector according to claim 15, a host cell according to claim 16, a ligand according to claim 17 or claim 18, a compound according to any one of claims 19 to 21, or a pharmaceutical composition according to claim 35, for use in the manufacture of a medicament for the treatment of renal disease, polycystic kidney disease, glomerulnephritis, fertility disorders, infertility, reproductive disorders, polycystic ovary disease, polycystic diseases, endocrine diseases, testosterone deficiency, testosterone-related disorder, testicular cancer, prostate cancer, gastrointestinal disorders, developmental disorders, brain tumour, sexual dysfunction, psychological disorder, alopecia, hirsuitism, diseases related to muscle tone, including vascular muscle disorders such as migraine, pre-eclampsia, hypertension and cardiac arrhythmias; bladder muscle tone disorders, such as urinary incontinence, inability to void, urinary tract infection; gastrointestinal smooth muscle tone disorder, such as Gl motility disorder, gastrointestinal reflux diorder (GERD), reflux oesophagitis, paralytic ileus, irritable bowel syndrome (IBS), diaπhoea including travellers diaπhoea and viral diaπhoea, constipation and malabsorption; skeletal muscle tone disorder, such as spacticity, and muscle fatigue disorders such as myotonias, myasthenia gravis, floppy baby syndrome, ME, frailty of old age; lung disorders such as COPD, asthma, pulmonary hypertension; adrenal disorders, such as adrenal hypertension, Conn's syndrome, Cushing disease and Addison's disease.
39. A polypeptide according to any one of claims 1 to 10, a nucleic acid molecule according to any one of claims 11 to 14, a vector according to claim 15, a host cell according to claim 16, a ligand according to claim 17 or claim 18, a compound according to any one of claims 19 to 21, or a pharmaceutical composition according to claim 35, for use in the manufacture of a contraceptive agent.
40. A method of treating a disease in a patient, comprising administering to the patient a polypeptide according to any one of claims 1 to 10, a nucleic acid molecule according to any one of claims 11 to 14, a vector according to claim 15, a host cell according to claim 16, a ligand according to claim 17 or claim 18, a compound according to any one of claims 19 to 21, or a pharmaceutical composition according to claim 35.
41. A method of preventing pregnancy in a patient, comprising administering to the patient a polypeptide according to any one of claims 1 to 10, a nucleic acid molecule according to any one of claims 11 to 14, a vector according to claim 15, a host cell according to claim 16, a ligand according to claim 17 or claim 18, a compound according to any one of claims 19 to 21, or a pharmaceutical composition according to claim 35.
42. A method according to claim 40, wherein, for diseases in which the expression of the natural gene or the activity of the polypeptide is lower in a diseased patient when compared to the level of expression or activity in a healthy patient, the polypeptide, nucleic acid molecule, vector, ligand, compound or composition administered to the patient is an agonist.
43. A method according to claim 40, wherein, for diseases in which the expression of the natural gene or activity of the polypeptide is higher in a diseased patient when compared to the level of expression or activity in a healthy patient, the polypeptide, nucleic acid molecule, vector, ligand, compound or composition administered to the patient is an antagonist.
44. A method of monitoring the therapeutic treatment of disease in a patient, comprising monitoring over a period of time the level of expression or activity of a polypeptide according to any one of claims 1 to 10, or the level of expression of a nucleic acid molecule according to any one of claims 11 to 14 in tissue from said patient, wherein altering said level of expression or activity over the period of time towards a control level is indicative of regression of said disease.
45. A method for the identification of a compound that is effective in the treatment and/or diagnosis of disease, comprising contacting a polypeptide according to any one of claims 1 to 10, or a nucleic acid molecule according to any one of claims 11 to 14 with one or more compounds suspected of possessing binding affinity for said polypeptide or nucleic acid molecule, and selecting a compound that binds specifically to said nucleic acid molecule or polypeptide.
46. A kit useful for diagnosing disease comprising a first container containing a nucleic acid probe that hybridises under stringent conditions with a nucleic acid molecule according to any one of claims 11 to 14; a second container containing primers useful for amplifying said nucleic acid molecule; and instructions for using the probe and primers for facilitating the diagnosis of disease.
47. The kit of claim 46, further comprising a third container holding an agent for digesting unhybridised RNA.
48. A kit comprising an aπay of nucleic acid molecules, at least one of which is a nucleic acid molecule according to any one of claims 11 to 14.
49. A kit comprising one or more antibodies that bind to a polypeptide as recited in any one of claims 1 to 10; and a reagent useful for the detection of a binding reaction between said antibody and said polypeptide.
50. A transgenic or knockout non-human animal that has been transformed to express higher, lower or absent levels of a polypeptide according to any one of claims 1 to 10.
51. A method for screening for a compound effective to treat disease, by contacting a non- human transgenic animal according to claim 50 with a candidate compound and determining the effect of the compound on the disease of the animal.
52. A method according to any one of claims 40-45 or claim 51, wherein said disease is one of the diseases set forth in claim 32 or claim 33.
PCT/GB2003/001655 2002-04-16 2003-04-16 Cation channel proteins WO2003089469A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003232308A AU2003232308A1 (en) 2002-04-16 2003-04-16 Cation channel proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0208707.0 2002-04-16
GB0208707A GB0208707D0 (en) 2002-04-16 2002-04-16 Proteins

Publications (2)

Publication Number Publication Date
WO2003089469A2 true WO2003089469A2 (en) 2003-10-30
WO2003089469A3 WO2003089469A3 (en) 2004-01-08

Family

ID=9934937

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2003/001655 WO2003089469A2 (en) 2002-04-16 2003-04-16 Cation channel proteins

Country Status (3)

Country Link
AU (1) AU2003232308A1 (en)
GB (1) GB0208707D0 (en)
WO (1) WO2003089469A2 (en)

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
DATABASE GENBANK [Online] 15 September 2001 (2001-09-15), NAGASE T ET AL: "Homo sapiens mRNA for KIAA1879 protein, partial cds." XP002249170 accession no. NCBI Database accession no. AB067466 *
DATABASE GENBANK [Online] 2 November 2001 (2001-11-02), DOE JOINT GENOME INSTITUTE: "Homo sapiens chromosome 16 clone RP11-303E16, complete sequence" XP002249169 accession no. NCBI Database accession no. AC092718 *
DATABASE GENBANK [Online] 7 February 2002 (2002-02-07), NCBI ANNOTATION PROJECT.: "Homo sapiens chromosome 16 working draft sequence segment." XP002249173 accession no. NCBI Database accession no. NT_024792 *
DATABASE GENBANK [Online] 7 February 2002 (2002-02-07), NCBI ANNOTATION PROJECT.: "Homo sapiens similar to KIAA1879 protein (LOC162163), mRNA." XP002249172 accession no. NCBI Database accession no. XM_091397 cited in the application *
DATABASE GENBANK [Online] 7 February 2002 (2002-02-07), NCBI ANNOTATION PROJECT: "KIAA1879 protein ÄHomo sapiens]" XP002249171 accession no. NCBI Database accession no. XP_056635 cited in the application *
HUGHES J ET AL: "Identification of a human homologue of the sea urchin receptor for egg jelly: a polycystic kidney disease-like protein" HUMAN MOLECULAR GENETICS, OXFORD UNIVERSITY PRESS, SURREY, GB, vol. 8, no. 3, March 1999 (1999-03), pages 543-549, XP002120871 ISSN: 0964-6906 cited in the application *
LI AIRONG ET AL: "Identification of two novel polycystic kidney disease-1-like genes in human and mouse genomes." GENOMICS. UNITED STATES JUN 2003, vol. 81, no. 6, June 2003 (2003-06), pages 596-608, XP002249168 ISSN: 0888-7543 *
MENGERINK KATHRYN J ET AL: "suREJ3, a polycystin-1 protein, is cleaved at the GPS domain and localizes to the acrosomal region of sea urchin sperm." JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 277, no. 2, 11 January 2002 (2002-01-11), pages 943-948, XP002249167 January 11, 2002 ISSN: 0021-9258 cited in the application *
NAGASE T ET AL: "PREDICTION OF THE CODING SEQUENCES OF UNIDENTIFIED HUMAN GENES XXI. THE COMPLETE SEQUENCES OF 60 NEW CDNA CLONES FROM BRAIN WHICH CODE FOR LARGE PROTEINS" DNA RESEARCH, UNIVERSAL ACADEMY PRESS, JP, vol. 8, no. 4, 31 August 2001 (2001-08-31), pages 179-187, XP001055485 ISSN: 1340-2838 *
STAYNER C ET AL: "Polycystin channels and kidney disease" TRENDS IN PHARMACOLOGICAL SCIENCES, ELSEVIER TRENDS JOURNAL, CAMBRIDGE, GB, vol. 22, no. 11, 1 November 2001 (2001-11-01), pages 543-546, XP004320222 ISSN: 0165-6147 *

Also Published As

Publication number Publication date
AU2003232308A1 (en) 2003-11-03
AU2003232308A8 (en) 2003-11-03
GB0208707D0 (en) 2002-05-29
WO2003089469A3 (en) 2004-01-08

Similar Documents

Publication Publication Date Title
WO2003054004A2 (en) Secreted proteins
WO2004026904A1 (en) Rfamide-related peptide precursor proteins and rfamide peptides
WO2003099865A1 (en) Cation channel proteins
US20040249123A1 (en) Nuclear hormone receptor ligand binding domain
WO2003089469A2 (en) Cation channel proteins
WO2004101618A2 (en) Progestin-yol002c-cgi-45 receptor-related proteins
US20040132038A1 (en) Nuclear hormone receptor ligand binding domain
US20050026251A1 (en) Transmembrane protein
WO2002029023A1 (en) Nicastrin protein
WO2004003190A1 (en) Polypeptide identified as a cytochrome p450
US20060234218A1 (en) P450 proteins
WO2003066844A2 (en) Cytochrome p450 protein
WO2003072605A1 (en) Nuclear hormone receptor ligand binding domain-containing protein
WO2004009624A2 (en) Three finger toxin fold protein
WO2003072769A1 (en) Cytochrome p450 protein
WO2004081170A2 (en) Progestin receptor-like protein
WO2002046380A2 (en) Protein kinase
WO2002034901A2 (en) Protein kinase
WO2002072805A2 (en) Thymidylate synthase
WO2007060423A1 (en) Signal anchored protein
EP1629008A1 (en) Tnf-like secreted protein
WO2002029062A2 (en) Cytokine proteins
WO2006040594A2 (en) Nek kinase (nima related kinase) proteins

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP