Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ALTERATIONS IN THE LONG QT SYNDROME GENES KVLQT1 AND SCN5A AND METHODS FOR DETECTING SAME
Document Type and Number:
WIPO Patent Application WO/2001/024681
Kind Code:
A2
Abstract:
Long QT Syndrome (LQTS) is a cardiovascular disorder characterized by prolongation of the QT interval on electrocardiogram and presence of syncope, seizures and sudden death. Five genes have been implicated in Romano-Ward syndrome, the autosomal dominant form of LQTS. These genes are KVLQT1, HERG, SCN5A, KCNE1 and KCNE2. Mutations in KVLQT1 and KCNE1 also cause the Jervell and Lange-Nielsen syndrome, a form of LQTS associated with deafness, a phenotypic abnormality inherited in an autosomal recessive fashion. Mutational analyses were used to screen 262 unrelated individuals with LQTS for mutations in the five defined genes. A total of 134 mutations were observed of which eighty were novel.

Inventors:
KEATING MARK T
SPLAWSKI IGOR
Application Number:
PCT/US2000/021660
Publication Date:
April 12, 2001
Filing Date:
August 09, 2000
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV UTAH RES FOUND (US)
International Classes:
G01N33/483; A61P9/02; C07C209/84; C07C211/42; C07K14/47; C07K16/18; C12N15/09; C12P21/08; C12Q1/42; C12Q1/68; G01N33/15; G01N33/50; G01N33/53; G01N33/566; (IPC1-7): A61B/
Other References:
FRANQUEZA ET AL.: 'Long QT syndrome-associated mutations in the S4-S5 linker of KvLQT1 potassium channels modify gating and interaction with minK subunits' JOURNAL OF BIOLOGICAL CHEMISTRY vol. 274, no. 30, 23 July 1999, pages 21063 - 21070, XP002960106
NEYROUD ET AL.: 'Heterozygous mutation in the pore of potassium channel gene KcLQT1 causes an apparently normal phenotype in long QT syndrome' EUROPEAN JOURNAL OF HUMAN GENETICS vol. 6, 1998, pages 129 - 133, XP002960104
AN ET AL.: 'Novel LQT-3 mutation affects Na+ channel activity through interactions between alpha and beta 1-subunits' CIRCULATION RESEARCH vol. 83, 1998, pages 141 - 146, XP002960105
See also references of EP 1339867A2
Attorney, Agent or Firm:
Saxe, Stephen A. (Figg Ernst & Manbeck, 555 13th Street N.W. Suite 701 East, Columbia Squar, Washington DC, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:
1. An isolated DNA comprising a sequence of SEQ ID NO : 1 as altered by one or more mutations selected from the group consisting of A332G, G478A, G521A, G535A, G580C, C727T, T742C, T797C, G921+1T, A9222C, G928A, C1046G, C1066T, G1097A, C1172T, C1343G, C1588T, C1697T, C1747T and G1781A.
2. A nucleic acid probe specifically hybridizable to a human mutated KVLQTI and not to wildtype DNA, said mutated KVLQT1 comprising a mutation of SEQ ID NO : 1 selected from the group consisting of A332G, G478A, G521A, G535A, G580C, C727T, T742C, T797C, G921+1T, A9222C, G928A, C1046G, C1066T, G1097A, C1172T, C1343G, C1588T, C1697T, C1747T and G1781A.
3. A method for detecting a mutation in KVLQT1 said mutation selected from the group consisting of A332G, G478A, G521A, G535A, G580C, C727T, T742C, T797C, G921+1T, A9222C, G928A, C1046G, C1066T, G1097A, C1172T, C1343G, C1588T, C1697T, C1747T and G1781A which comprises analyzing a sequence of said gene or RNA from a human sample or analyzing the sequence of cDNA made from mRNA from said sample.
4. The method of claim 3 wherein said mutation is detected by a method selected from the group consisting of : a) hybridizing a probe specific for one of said mutations to RNA isolated from said human sample and detecting the presence of a hybridization product, wherein the presence of said product indicates the presence of said mutation in the sample; b) hybridizing a probe specific for one of said mutations to cDNA made from RNA isolated from said sample and detecting the presence of a hybridization product, wherein the presence of said product indicates the presence of said mutation in the sample; c) hybridizing a probe specific for one of said mutations to genomic DNA isolated from said sample and detecting the presence of a hybridization product, wherein the presence of said product indicates the presence of said mutation in the sample; d) amplifying all or part of said gene in said sample using a set of primers to produce amplified nucleic acids and sequencing the amplified nucleic acids; e) amplifying part of said gene in said sample using a primer specific for one of said mutations and detecting the presence of an amplified product, wherein the presence of said product indicates the presence of said mutation in the sample; f) molecularly cloning all or part of said gene in said sample to produce a cloned nucleic acid and sequencing the cloned nucleic acid; g) amplifying said gene to produce amplified nucleic acids, hybridizing the amplified nucleic acids to a DNA probe specific for one of said mutations and detecting the presence of a hybridization product, wherein the presence of said product indicates the presence of said mutation; h) forming singlestranded DNA from a gene fragment of said gene from said human sample and singlestranded DNA from a corresponding fragment of a wildtype gene, electrophoresing said singlestranded DNAs on a nondenaturing polyacrylamide gel and comparing the mobility of said singlestranded DNAs on said gel to determine if said singlestranded DNA from said sample is shifted relative to wildtype and sequencing said singlestranded DNA having a shift in mobility; i) forming a heteroduplex consisting of a first strand of nucleic acid selected from the group consisting of a genomic DNA fragment isolated from said sample, an RNA fragment isolated from said sample and a cDNA fragment made from mRNA from said sample and a second strand of a nucleic acid consisting of a corresponding human wild type gene fragment, analyzing for the presence of a mismatch in said heteroduplex, and sequencing said first strand of nucleic acid having a mismatch; j) forming singlestranded DNA from said gene of said human sample and from a corresponding fragment of an allele specific for one of said mutations, electrophoresing said singlestranded DNAs on a nondenaturing polyacrylamide gel and comparing the mobility of said singlestranded DNAs on said gel to determine if said singlestranded DNA from said sample is shifted relative to said allele, wherein no shift in electrophoretic mobility of the singlestranded DNA relative to the allele indicates the presence of said mutation in said sample; and k) forming a heteroduplex consisting of a first strand of nucleic acid selected from the group consisting of a genomic DNA fragment of said gene isolated from said sample, an RNA fragment isolated from said sample and a cDNA fragment made from mRNA from said sample and a second strand of a nucleic acid consisting of a corresponding gene allele fragment specific for one of said mutations and analyzing for the presence of a mismatch in said heteroduplex, wherein no mismatch indicates the presence of said mutation.
5. A method according to claim 4 wherein hybridization is performed in situ.
6. An isolated human polypeptide encoded by KVLQT1 comprising a mutation of SEQ ID NO : 2 selected from the group consisting ofYlllC, E160K, R174H, G179S, A194P, R243C, W248R, L266P, V307sp, V310I, S349W, Q356X, R366Q, T391I, P448R, Q530X, S566F, R583C and R594Q.
7. An antibody capable of binding the polypeptide of claim 6 but incapable of binding a wildtype polypeptide.
8. An antibody according to claim 7 wherein said antibody is a monoclonal antibody.
9. A method of assessing a risk in a human subject for long QT syndrome which comprises screening said subject for a mutation in KVLQT1 by comparing the sequence of said KVLQTI or its expression products isolated from a tissue sample of said subject with a wildtype sequence of said KVLQT1 or its expression products, wherein a mutation in the sequence of the subject indicates a risk for long QT syndrome.
10. The method of claim 9 wherein said expression product is selected from mRNA of said gene or a polypeptide encoded by said gene.
11. The method of claim 9 wherein one or more of the following procedures is carried out: (a) observing shifts in electrophoretic mobility of singlestranded DNA from said sample on nondenaturing polyacrylamide gels; (b) hybridizing a probe to genomic DNA isolated from said sample under conditions suitable for hybridization of said probe to said gene; (c) determining hybridization of an allelespecific probe to genomic DNA from said sample; (d) amplifying all or part of said gene from said sample to produce an amplified sequence and sequencing the amplified sequence; (e) determining by nucleic acid amplification the presence of a specific mutant allele in said sample; (f) molecularly cloning all or part of said gene from said sample to produce a cloned sequence and sequencing the cloned sequence; (g) determining whether there is a mismatch between molecules (1) said gene genomic DNA or mRNA isolated from said sample, and (2) a nucleic acid probe complementary to the human wildtype gene DNA, when molecules (1) and (2) are hybridized to each other to form a duplex; (h) amplification of said gene sequences in said sample and hybridization of the amplified sequences to nucleic acid probes which comprise wildtype gene sequences; (i) amplification of said gene sequences in said tissue and hybridization of the amplified sequences to nucleic acid probes which comprise said mutant gene sequences; (j) screening for a deletion mutation; (k) screening for a point mutation; (1) screening for an insertion mutation; (m) determining in situ hybridization of said gene in said sample with one or more nucleic acid probes which comprise said gene sequence or a mutant sequence of said gene; (n) immunoblotting; (o) immunocytochemistry; (p) assaying for binding interactions between said gene protein isolated from said tissue and a binding partner capable of specifically binding the polypeptide expression product of a mutant allele and/or a binding partner for the polypeptide; and (q) assaying for the inhibition of biochemical activity of said binding partner.
12. A nucleic acid probe which hybridizes to the isolated DNA of claim 1 under conditions at which it will not hybridize to wildtype DNA.
13. A method for diagnosing a mutation which causes long QT syndrome comprising hybridizing a probe of claim 12 to a patient's sample of DNA or RNA, the presence of a hybridization signal being indicative of long QT syndrome.
14. A method according to claim 13 wherein the patient's DNA or RNA has been amplified and said amplified DNA or RNA is hybridized with a probe of claim 12.
15. A method according to claim 13 wherein said hybridization is performed in situ.
16. A method according to claim 13 wherein said assay is performed using nucleic acid microchip technology.
17. A method for diagnosing a mutation which causes long QT syndrome comprising amplifying a region of gene or RNA for KVLQT1 and sequencing the amplified gene or RNA wherein long QT syndrome is indicated by any one or more mutations selected from the group consisting of A332G, G478A, G521A, G535A, G580C, C727T, T742C, T797C, G921+1T, A9222C, G928A, C1046G, C1066T, G1097A, C1172T, C1343G, C1588T, C1697T, C1747T and G1781A.
18. A method for diagnosing a mutation which causes long QT syndrome comprising identifying a mismatch between a patient's DNA or RNA and a wildtype DNA or RNA probe wherein said probe hybridizes to a region of DNA or RNA wherein said region comprises a mutation of SEQ ID NO : 1 selected from the group consisting of A332G, G478A, G521A, G535A, G580C, C727T, T742C, T797C, G921+1T, A9222C, G928A, C1046G, C1066T, G1097A, C1172T, C1343G, C1588T, C1697T, C1747T and G1781A.
19. The method of claim 18 wherein the mismatch is identified by an RNase assay.
20. A method for diagnosing long QT syndrome said method consisting of an assay for the presence of mutant KVLQT1 polypeptide in a patient by reacting a patient's sample with an antibody of claim 7, the presence of a positive reaction being indicative of long QT syndrome.
21. The method of claim 20 wherein said assay comprises immunoblotting.
22. The method of claim 20 wherein said assay comprises an immunocytochemical technique.
23. A method for diagnosing long QT syndrome, said method comprising analyzing a KVLQT1 polypeptide, a mutation in said polypeptide being indicative of long QT syndrome wherein said mutation is a mutation selected from the group consisting of Y111C, E160K, R174H, G179S, A194P, R243C, W248R, L266P, V307sp, V310I, S349W, Q356X, R366Q, T391I, P448R, Q530X, S566F, R583C and R594Q.
24. A method to screen for drugs which are useful in treating a person with a mutation in KVLQTI wherein said mutation is selected from the group consisting of A332G, G478A, G521A, G535A, G580C, C727T, T742C, T797C, G921+1T, A9222C, G928A, C1046G, C1066T, G1097A, C1172T, C1343G, C1588T, C1697T, C1747T and G1781A, said method comprising: a) placing a first set of cells expressing KVLQT1 with a mutation, wherein said mutation is selected from the group consisting of Y111 C, E160K, R174H, G179S, A194P, R243C, W248R, L266P, V307sp, V310I, S349W, Q356X, R366Q, T391I, P448R, Q530X, S566F, R583C and R594Q, into a bathing solution; b) inducing a first induced K current in the cells of step (a); c) measuring said first induced K+ current ; d) placing a second set of cells expressing wildtype KVLQT1 into a bathing solution e) inducing a second induced K current in the cells of step (d); f) measuring said second induced K current; g) adding a drug to the bathing solution of step (a); h) inducing a third induced K+ current in the cells of step (g); i) measuring said third induced K current ; and j) determining whether the third induced K current is more similar to the second induced K current than is the first induced K current, wherein drugs resulting in a third induced K current which is closer to the second induced K+ current than is the first induced K current are useful in treating said persons. An isolated DNA encoding a KVLQT1 polypeptide of SEQ ID NO : 2 having a mutation selected from the group consisting of 11C, E160K, R174H, G179S, A194P, R243C, W248R, L266P, V307sp, V310I, S349W, Q356X, R366Q, T391I, P448R, Q530X, S566F, R583C and R594Q. An isolated DNA comprising a sequence of SEQ ID NO : 3 as altered by one or more mutations selected from the group consisting of G3340A, C4501G, del48504852, G4868T, G5349A and G5360A. A nucleic acid probe specifically hybridizable to a human mutated SCN5A and not to wildtype DNA, said mutated SCNSA comprising a mutation of SEQ ID NO : 3 selected from the group consisting of G3340A, C4501G, del48504852, G4868T, G5349A and G5360A. A method for detecting a mutation in SCNSA said mutation selected from the group consisting of G3340A, C4501G, del48504852, G4868T, G5349A and G5360A which comprises analyzing a sequence of said gene or RNA from a human sample or analyzing the sequence of cDNA made from mRNA from said sample.
25. 29 The method of claim 28 wherein said mutation is detected by a method selected from the group consisting of : a) hybridizing a probe specific for one of said mutations to RNA isolated from said human sample and detecting the presence of a hybridization product, wherein the presence of said product indicates the presence of said mutation in the sample; b) hybridizing a probe specific for one of said mutations to cDNA made from RNA isolated from said sample and detecting the presence of a hybridization product, wherein the presence of said product indicates the presence of said mutation in the sample; c) hybridizing a probe specific for one of said mutations to genomic DNA isolated from said sample and detecting the presence of a hybridization product, wherein the presence of said product indicates the presence of said mutation in the sample; d) amplifying all or part of said gene in said sample using a set of primers to produce amplified nucleic acids and sequencing the amplified nucleic acids; e) amplifying part of said gene in said sample using a primer specific for one of said mutations and detecting the presence of an amplified product, wherein the presence of said product indicates the presence of said mutation in the sample; f) molecularly cloning all or part of said gene in said sample to produce a cloned nucleic acid and sequencing the cloned nucleic acid; g) amplifying said gene to produce amplified nucleic acids, hybridizing the amplified nucleic acids to a DNA probe specific for one of said mutations and detecting the presence of a hybridization product, wherein the presence of said product indicates the presence of said mutation ; h) forming singlestranded DNA from a gene fragment of said gene from said human sample and singlestranded DNA from a corresponding fragment of a wildtype gene, electrophoresing said singlestranded DNAs on a nondenaturing polyacrylamide gel and comparing the mobility of said singlestranded DNAs on said gel to determine if said singlestranded DNA from said sample is shifted relative to wildtype and sequencing said singlestranded DNA having a shift in mobility; i) forming a heteroduplex consisting of a first strand of nucleic acid selected from the group consisting of a genomic DNA fragment isolated from said sample, an RNA fragment isolated from said sample and a cDNA fragment made from mRNA from said sample and a second strand of a nucleic acid consisting of a corresponding human wild type gene fragment, analyzing for the presence of a mismatch in said heteroduplex, and sequencing said first strand of nucleic acid having a mismatch; j) forming singlestranded DNA from said gene of said human sample and from a corresponding fragment of an allele specific for one of said mutations, electrophoresing said singlestranded DNAs on a nondenaturing polyacrylamide gel and comparing the mobility of said singlestranded DNAs on said gel to determine if said singlestranded DNA from said sample is shifted relative to said allele, wherein no shift in electrophoretic mobility of the singlestranded DNA relative to the allele indicates the presence of said mutation in said sample; and k) forming a heteroduplex consisting of a first strand of nucleic acid selected from the group consisting of a genomic DNA fragment of said gene isolated from said sample, an RNA fragment isolated from said sample and a cDNA fragment made from mRNA from said sample and a second strand of a nucleic acid consisting of a corresponding gene allele fragment specific for one of said mutations and analyzing for the presence of a mismatch in said heteroduplex, wherein no mismatch indicates the presence of said mutation.
26. 30 A method according to claim 29 wherein hybridization is performed in situ.
27. 31 An isolated human polypeptide encoded by SCNSA comprising a mutation of SEQ ID NO : 4 selected from the group consisting of D1114N, L1501V, delF1617, R1623L, E1784K and S1787N.
28. 32 An antibody capable of binding the polypeptide of claim 31 but incapable of binding a wildtype polypeptide.
29. 33 An antibody according to claim 32 wherein said antibody is a monoclonal antibody.
30. 34 A method of assessing a risk in a human subject for long QT syndrome which comprises screening said subject for a mutation in SCN5A by comparing the sequence of said SCNSA or its expression products isolated from a tissue sample of said subject with a wildtype sequence of said SCNSA or its expression products, wherein a mutation in the sequence of the subject indicates a risk for long QT syndrome.
31. 35 The method of claim 34 wherein said expression product is selected from MARNA of said gene or a polypeptide encoded by said gene.
32. 36 The method of claim 34 wherein one or more of the following procedures is carried out: (a) observing shifts in electrophoretic mobility of singlestranded DNA from said sample on nondenaturing polyacrylamide gels; (b) hybridizing a probe to genomic DNA isolated from said sample under conditions suitable for hybridization of said probe to said gene; (c) determining hybridization of an allelespecific probe to genomic DNA from said sample; (d) amplifying all or part of said gene from said sample to produce an amplified sequence and sequencing the amplified sequence; (e) determining by nucleic acid amplification the presence of a specific mutant allele in said sample; (f) molecularly cloning all or part of said gene from said sample to produce a cloned sequence and sequencing the cloned sequence; (g) determining whether there is a mismatch between molecules (1) said gene genomic DNA or mRNA isolated from said sample, and (2) a nucleic acid probe complementary to the human wildtype gene DNA, when molecules (1) and (2) are hybridized to each other to form a duplex ; (h) amplification of said gene sequences in said sample and hybridization of the amplified sequences to nucleic acid probes which comprise wildtype gene sequences; (i) amplification of said gene sequences in said tissue and hybridization of the amplified sequences to nucleic acid probes which comprise said mutant gene sequences ; (j) screening for a deletion mutation; (k) screening for a point mutation; (1) screening for an insertion mutation; (m) determining in situ hybridization of said gene in said sample with one or more nucleic acid probes which comprise said gene sequence or a mutant sequence of said gene; (n) immunoblotting; (o) immunocytochemistry; (p) assaying for binding interactions between said gene protein isolated from said tissue and a binding partner capable of specifically binding the polypeptide expression product of a mutant allele and/or a binding partner for the polypeptide; and (q) assaying for the inhibition of biochemical activity of said binding partner.
33. 37 A nucleic acid probe which hybridizes to the isolated DNA of claim 26 under conditions at which it will not hybridize to wildtype DNA.
34. 38 A method for diagnosing a mutation which causes long QT syndrome comprising hybridizing a probe of claim 37 to a patient's sample of DNA or RNA, the presence of a hybridization signal being indicative of long QT syndrome.
35. 39 A method according to claim 38 wherein the patient's DNA or RNA has been amplified and said amplified DNA or RNA is hybridized with a probe of claim 37.
36. 40 A method according to claim 38 wherein said hybridization is performed in situ.
37. 41 A method according to claim 38 wherein said assay is performed using nucleic acid microchip technology.
38. 42 A method for diagnosing a mutation which causes long QT syndrome comprising amplifying a region of gene or RNA for SCNSA and sequencing the amplified gene or RNA wherein long QT syndrome is indicated by any one or more mutations selected from the group consisting of G3340A, C4501G, del48504852, G4868T, G5349A and G5360A.
39. 43 A method for diagnosing a mutation which causes long QT syndrome comprising identifying a mismatch between a patient's DNA or RNA and a wildtype DNA or RNA probe wherein said probe hybridizes to a region of DNA or RNA wherein said region comprises a mutation of SEQ ID NO : 3 selected from the group consisting of G3340A, C4501G, del48504852, G4868T, G5349A and G5360A.
40. 44 The method of claim 43 wherein the mismatch is identified by an RNase assay.
41. 45 A method for diagnosing long QT syndrome said method consisting of an assay for the presence of mutant SCN5A polypeptide in a patient by reacting a patient's sample with an antibody of claim 32, the presence of a positive reaction being indicative of long QT syndrome.
42. 46 The method of claim 45 wherein said assay comprises immunoblotting.
43. 47 The method of claim 45 wherein said assay comprises an immunocytochemical technique.
44. 48 A method for diagnosing long QT syndrome, said method comprising analyzing a SCN5A polypeptide, a mutation in said polypeptide being indicative of long QT syndrome wherein said mutation is a mutation selected from the group consisting of D1114N, L1501V, delF1617, R1623L, E1784K and S1787N.
45. 49 A method to screen for drugs which are useful in treating a person with a mutation in SCNSA wherein said mutation is selected from the group consisting of G3340A, C4501 G, del48504852, G4868T, G5349A and G5360A, said method comprising: a) placing a first set of cells expressing SCN5A with a mutation, wherein said mutation is selected from the group consisting of D1114N, L1501V, delF1617, R1623L, E1784K and S 1787N, into a bathing solution; b) inducing a first induced Na current in the cells of step (a); c) measuring said first induced Na current; d) placing a second set of cells expressing wildtype SCN5A into a bathing solution; e) inducing a second induced Na+ current in the cells of step (d); f) measuring said second induced Na+ current; g) adding a drug to the bathing solution of step (a); h) inducing a third induced Na+ current in the cells in step (g); i) measuring said third induced Na+ current; and j) determining whether the third induced Na+ current is more similar to the second induced Na+ current than is the first induced Na+ current, wherein drugs resulting in a third induced Na+ current which is closer to the second induced Na+ current than is the first induced Na+ current are useful in treating said persons.
46. 50 An isolated DNA encoding an SCN5A polypeptide of SEQ ID NO : 4 having a mutation selected from the group consisting of D1114N, L1501V, delF1617, R1623L, E1784K and S1787N.
Description:
TITLE OF THE INVENTION ALTERATIONS IN THE LONG QT SYNDROME GENES KVLQT1 AND SCNSA AND METHODS FOR DETECTING SAME This application was made with Government support from NHLBI under Grant Nos.

RO1-HL46401, RO1-HL33843, RO1-HL51618, P50-HL52338 and MO1-RR000064. The federal government may have certain rights in this invention.

BACKGROUND OF THE INVENTION Long QT Syndrome (LQTS) is a cardiovascular disorder characterized by prolongation of the QT interval on electrocardiogram and presence of syncope, seizures and sudden death, usually in young, otherwise healthy individuals (Jervell and Lange-Nielsen, 1957; Romano et al., 1963; Ward, 1964). The clinical features of LQTS result from episodic ventricular tachyarrhythmias, such as torsade de pointes and ventricular fibrillation (Schwartz et al., 1975; Moss et al., 1991). Two inherited forms of LQTS exist. The more common form, Romano- Ward syndrome (RW), is not associated with other phenotypic abnormalities and is inherited as an autosomal dominant trait with variable penetrance (Roman et al., 1963; Ward, 1964). Jervell and Lange-Nielsen syndrome (JLN) is characterized by the presence of deafness, a phenotypic abnormality inherited as an autosomal recessive. trait (Jervell and Lange-Nielsen, 1957). LQTS can also be acquired, usually as a result of pharmacologic therapy.

In previous studies, we mapped LQTS loci to chromosomes 1 lpl5. 5 (LQT1) (Keating et al., 1991), 7 q35-36 (LQT2) (Jiang et al., 1994) and LQT3 to 3p21-24 (Jiang et al., 1994). A fourth locus (LQT4) was mapped to 4q25-27 (Schott et al., 1995). Five genes have been implicated in Romano-Ward syndrome, the autosomal dominant form of LQTS. These genes are KVLQT1 (LQT1) (Wang Q. et al., 1996a), HERG (LQT2) (Curran et al., 1995), SCNSA (LQT3) (Wang et al., 1995a), and two genes located at 21q22-KCNE1 (LQT5) (Splawski et al., 1997a) and KCNE2 (LQTe (Abbott et al., 1999). Mutations in KVLQT1 and KCNE1 also cause the Jervell and Lange-Nielsen syndrome, a form of LQTS associated with deafness, a phenotypic abnormality inherited in an autosomal recessive fashion.

KVLQT1, HERG, KCNE1 and KCNE2 encode potassium channel subunits. Four KVLQT1 a-subunits assemble with minK (p-subunits encoded by KCNE1, stoichiometry is

unknown) to form IKS channels underlying the slowly activating delayed rectifier potassium current in the heart (Sanguinetti et al., 1996a; Barhanin et al., 1996). Four HERG a-subunits assemble with MiRP1 (encoded by KCNE2, stoichiometry unknown) to form IK, channels, which underlie the rapidly activating, delayed rectifier potassium current (Abbott et al., 1999). Mutant subunits lead to reduction of IKS or IKr by a loss-of-function mechanism, often with a dominant- negative effect (Chouabe et al., 1997; Shalaby et al., 1997; Wollnik et al., 1997; Sanguinetti et al., 1996b). SCNSA encodes the cardiac sodium channel that is responsible for INa, the sodium current in the heart (Gellens et al., 1992). LQTS-associated mutations in SCN5A cause a gain-of- function (Bennett et al., 1995; Dumaine et al., 1996). In the heart, reduced IKS or IKr or increased INa leads to prolongation of the cardiac action potential, lengthening of the QT interval and increased risk of arrhythmia. KVLQT1 and KCNEI are also expressed in the inner ear (Neyroud et al., 1997; Vetter et al., 1996). Others and we demonstrated that complete loss of IKS causes the severe cardiac phenotype and deafness in JLN (Neyroud et al., 1997; Splawski et al., 1997b; Tyson et al., 1997; Schulze-Bahr et al., 1997).

Presymptomatic diagnosis of LQTS is currently based on prolongation of the QT interval on electrocardiogram. Genetic studies, however, have shown that diagnosis based solely on electrocardiogram is neither sensitive nor specific (Vincent et al., 1992; Priori et al., 1999).

Genetic screening using mutational analysis can improve presymptomatic diagnosis. However, a comprehensive study identifying and cataloging all LQTS-associated mutations in all five genes has not been achieved. To determine the relative frequency of mutations in each gene, facilitate presymptomatic diagnosis and enable genotype-phenotype studies, we screened a pool of 262 unrelated individuals with LQTS for mutations in the five defined genes. The results of these studies are presented in the Examples below.

The present invention relates to alterations in the KVLQT1, HERG, SCNSA, KCNE1 and KCNE2 genes and methods for detecting such alterations.

The publications and other materials used herein to illuminate the background of the invention or provide additional details respecting the practice, are incorporated by reference, and for convenience are respectively grouped in the appended List of References.

The present invention is directed to alterations in genes and gene products associated with long QT syndrome and to a process for the diagnosis and prevention of LQTS. LQTS is

diagnosed in accordance with the present invention by analyzing the DNA sequence of the KVI6QT1, HERG, SCN5H, KCNE1 or KCNE2 gene of an individual to be tested and comparing the respective DNA sequence to the known DNA sequence of the normal gene. Alternatively, these genes of an individual to be tested can be screened for mutations which cause LQTS.

Prediction of LQTS will enable practitioners to prevent this disorder using existing medical therapy.

SUMMARY OF THE INVENTION The present invention relates to alterations in the KVLQTI, HERG, SCNSA, KCNE1 and KCNE2 genes and methods for detecting such alterations. The alterations in the KVLQT1, HERG, SCN5S, KCNE1 and KCNE2 genes include mutations and polymorphisms. Included among the mutations are frameshift, nonsense, splice, regulatory and missense mutations. Any method which is capable of detecting the alterations described herein can be used. Such methods include, but are not limited to, DNA sequencing, allele-specific probing, mismatch detection, single stranded conformation polymorphism detection and allele-specific PCR amplification.

BRIEF DESCRIPTION OF THE FIGURES Figure 1 is a schematic representation of the predicted topology of KVLQT1 and the locations of LQTS-associated mutations. KVLQT1 consists of six putative transmembrane segments (Sl to S6) and a pore (Pore) region. Each circle represents an amino acid. The approximate location of LQTS-associated mutations identified in our laboratory are shown with filled circles.

Figure 2 is a schematic representation of HERG mutations. HERO consists of six putative transmembrane segments (S 1 to S6) and a pore (Pore) region. Location of LQTS- associated mutations are shown with filled circles.

Figure 3 is a schematic representation of SCN5A and locations of LQTS-associated mutations. SCN5A consists of four domain (DI to DIV), each of which has six putative

transmembrane segments (S 1 to S6) and a pore (Pore) region. Location of LQTS-associated mutations identified in our laboratory are shown with filled circles.

Figure 4 is a schematic representation of minK and locations of LQT-associated mutations. MinK consists of one putative transmembrane domain (Sl). The approximate location of LQTS-associated rnutations identified in our laboratory are shown with filled circles.

Figure 5 is a schematic representation of the predicted topology of MiRP1 and locations of arrhythmia-associated mutations. MiRP1 consists of one putative transmembrane domain (S1). The approximate location of arrhythmia-associated mutations identified in our laboratory are shown with filled circles.

DETAILED DESCRIPTION OF THE INVENTION The present invention relates to alterations in the KVLQT1, HERG, SCN5A, KCNE1 and KCNE2 genes and methods for detecting such alterations. The alterations in the KVlQTl, HERG, SCN5A, KCNE1 and KCNE2 genes include mutations and polymorphisms. Included among the mutations are frameshift, nonsense, splice, regulatory and missense mutations. Any method which is capable of detecting the mutations and polymorphisms described herein can be used. Such methods include, but are not limited to, DNA sequencing, allele-specific probing, mismatch detection, single stranded conformation polymorphism detection and allele-specific PCR amplification.

KVLQT1, HERG, SCNSA, KCNE1 and KCNE2 mutations cause increased risk for LQTS.

Many different mutations occur in KVLQT1, HERG, SCNSA, KCNE1 and KCNE2. In order to detect the presence of alterations in the KVLQT1, HERG, SCN5A, KCNEl and KCNE2 genes, a biological sample such as blood is prepared and analyzed for the presence or absence of a given alteration of KVLQT1, HERG, SCNSA, KCNE1 or KCNE2. In order to detect the increased risk for LQTS or for the lack of such increased risk, a biological sample is prepared and analyzed for the presence or absence of a mutant allele of KVLQT1, HERG, SCNSA, KCNE1 or KCNE2.

Results of these tests and interpretive information are returned to the health care provider for communication to the tested individual. Such diagnoses may be performed by diagnostic

laboratories or, alternatively, diagnostic kits are manufactured and sold to health care providers or to private individuals for self-diagnosis.

The presence of hereditary LQTS may be ascertained by testing any tissue of a human for mutations of the KVLQT1, HERG, SCNSA, KCNEI or KCNE2 gene. For example, a person who has inherited a germline HERG mutation would be prone to develop LQTS. This can be determined by testing DNA from any tissue of the person's body. Most simply, blood can be drawn and DNA extracted from the cells of the blood. In addition, prenatal diagnosis can be accomplished by testing fetal cells, placental cells or amniotic cells for mutations of the KVLQTl, HERG, SCN5A, KCNEI or KCNE2 gene. Alteration of a wild-type KVLQT1, HERG, SCNSA, KCNE1 or KCNE2 allele, whether, for example, by point mutation or deletion, can be detected by any of the means discussed herein.

There are several methods that can be used to detect DNA sequence variation. Direct DNA sequencing, either manual sequencing or automated fluorescent sequencing can detect sequence variation. Another approach is the single-stranded conformation polymorphism assay (SSCP) (Orita et al., 1989). This method does not detect all sequence changes, especially if the DNA fragment size is greater than 200 bp, but can be optimized to detect most DNA sequence variation. The reduced detection sensitivity is a disadvantage, but the increased throughput possible with SSCP makes it an attractive, viable alternative to direct sequencing for mutation detection on a research basis. The fragments which have shifted mobility on SSCP gels are then sequenced to determine the exact nature of the DNA sequence variation. Other approaches based on the detection of mismatches between the two complementary DNA strands include clamped denaturing gel electrophoresis (CDGE) (Sheffield et al., 1991), heteroduplex analysis (HA) (White et al., 1992) and chemical mismatch cleavage (CMC) (Grompe et al., 1989). None of the methods described above will detect large deletions, duplications or insertions, nor will they detect a regulatory mutation which affects transcription or translation of the protein. Other methods which might detect these classes of mutations such as a protein truncation assay or the asymmetric assay, detect only specific types of mutations and would not detect missense mutations. A review of currently available methods of detecting DNA sequence variation can be found in a recent review by Grompe (1993). Once a mutation is known, an allele specific detection approach such as allele specific oligonucleotide (ASO) hybridization can be utilized to rapidly screen large numbers of other samples for that same mutation. Such a technique can

utilize probes which are labeled with gold nanoparticles to yield a visual color result (Elghanian et al., 1997).

A rapid preliminary analysis to detect polymorphisms in DNA sequences can be performed by looking at a series of Southern blots of DNA cut with one or more restriction enzymes, preferably with a large number of restriction enzymes. Each blot contains a series of normal individuals and a series of LQTS cases. Southern blots displaying hybridizing fragments (differing in length from control DNA when probed with sequences near or including the HERG locus) indicate a possible mutation. If restriction enzymes which produce very large restriction fragments are used, then pulsed field gel electrophoresis (PFGE) is employed.

Detection of point mutations may be accomplished by molecular cloning of the KVLQTI, HERG, SCNSA, KCNEI or KCNE2 alleles and sequencing the alleles using techniques well known in the art. Also, the gene or portions of the gene may be amplified, e. g., by PCR or other amplification technique, and the amplified gene or amplified portions of the gene may be sequenced.

There are six well known methods for a more complete, yet still indirect, test for confirming the presence of a susceptibility allele: 1) single stranded conformation analysis (SSCP) (Orita et al., 1989); 2) denaturing gradient gel electrophoresis (DGGE) (Wartell et al., 1990; Sheffield et al., 1989); 3) RNase protection assays (Finkelstein et al., 1990; Kinszler et al., 1991); 4) allele-specific oligonucleotides (ASOs) (Conner et al., 1983); 5) the use of proteins which recognize nucleotide mismatches, such as the E. coli mutS protein (Modrich, 1991); and 6) allele-specific PCR (Ruano and Kidd, 1989). For allele-specific PCR, primers are used which hybridize at their 3'ends to a particular KVLQTI, HERG, SCNSA, KCNEI or KCNE2 mutation.

If the particular mutation is not present, an amplification product is not observed. Amplification Refractory Mutation System (ARMS) can also be used, as disclosed in European Patent Application Publication No. 0332435 and in Newton et al., 1989. Insertions and deletions of genes can also be detected by cloning, sequencing and amplification. In addition, restriction fragment length polymorphism (RFLP) probes for the gene or surrounding marker genes can be used to score alteration of an allele or an insertion in a polymorphic fragment. Such a method is particularly useful for screening relatives of an affected individual for the presence of the mutation found in that individual. Other techniques for detecting insertions and deletions as known in the art can be used.

In the first three methods (SSCP, DGGE and RNase protection assay), a new electrophoretic band appears. SSCP detects a band which migrates differentially because the sequence change causes a difference in single-strand, intramolecular base pairing. RNase protection involves cleavage of the mutant polynucleotide into two or more smaller fragments.

DGGE detects differences in migration rates of mutant sequences compared to wild-type sequences, using a denaturing gradient gel. In an allele-specific oligonucleotide assay, an oligonucleotide is designed which detects a specific sequence, and the assay is performed by detecting the presence or absence of a hybridization signal. In the mutS assay, the protein binds only to sequences that contain a nucleotide mismatch in a heteroduplex between mutant and wild-type sequences.

Mismatches, according to the present invention, are hybridized nucleic acid duplexes in which the two strands are not 100% complementary. Lack of total homology may be due to deletions, insertions, inversions or substitutions. Mismatch detection can be used to detect point mutations in the gene or in its mRNA product. While these techniques are less sensitive than sequencing, they are simpler to perform on a large number of samples. An example of a mismatch cleavage technique is the RNase protection method. In the practice of the present invention, the method involves the use of a labeled riboprobe which is complementary to the human wild-type KVLQT1, HERG, SCNSA, KCNE1 or KCNE2 gene coding sequence. The riboprobe and either mRNA or DNA isolated from the person are annealed (hybridized) together and subsequently digested with the enzyme RNase A which is able to detect some mismatches in a duplex RNA structure. If a mismatch is detected by RNase A, it cleaves at the site of the mismatch. Thus, when the annealed RNA preparation is separated on an electrophoretic gel matrix, if a mismatch has been detected and cleaved by RNase A, an RNA product will be seen which is smaller than the full length duplex RNA for the riboprobe and the mRNA or DNA. The riboprobe need not be the full length of the mRNA or gene but can be a segment of either. If the riboprobe comprises only a segment of the mRNA or gene, it will be desirable to use a number of these probes to screen the whole mRNA sequence for mismatches.

In similar fashion, DNA probes can be used to detect mismatches, through enzymatic or chemical cleavage. See, e. g., Cotton et al., 1988; Shenk et al., 1975; Novack et al., 1986.

Alternatively, mismatches can be detected by shifts in the electrophoretic mobility of mismatched duplexes relative to matched duplexes. See, e. g., Cariello, 1988. With either riboprobes or DNA

probes, the cellular mRNA or DNA which might contain a mutation can be amplified using PCR (see below) before hybridization. Changes in DNA of the KVLQT1, HERG, SCNSA, KCNE1 or KCNE2 gene can also be detected using Southern hybridization, especially if the changes are gross rearrangements, such as deletions and insertions.

DNA sequences of the KVLQTI, HERG, SCNSA, KCNEI or KCNE2 gene which have been amplified by use of PCR may also be screened using allele-specific probes. These probes are nucleic acid oligomers, each of which contains a region of the gene sequence harboring a known mutation. For example, one oligomer may be about 30 nucleotides in length, corresponding to a portion of the gene sequence. By use of a battery of such allele-specific probes, PCR amplification products can be screened to identify the presence of a previously identified mutation in the gene. Hybridization of allele-specific probes with amplified KVLQT1, HERG, SCNSA, KCNEI or KCNE2 sequences can be performed, for example, on a nylon filter.

Hybridization to a particular probe under high stringency hybridization conditions indicates the presence of the same mutation in the tissue as in the allele-specific probe.

The newly developed technique of nucleic acid analysis via microchip technology is also applicable to the present invention. In this technique, literally thousands of distinct oligonucleotide probes are built up in an array on a silicon chip. Nucleic acid to be analyzed is fluorescently labeled and hybridized to the probes on the chip. It is also possible to study nucleic acid-protein interactions using these nucleic acid microchips. Using this technique one can determine the presence of mutations or even sequence the nucleic acid being analyzed or one can measure expression levels of a gene of interest. The method is one of parallel processing of many, even thousands, of probes at once and can tremendously increase the rate of analysis.

Several papers have been published which use this technique. Some of these are Hacia et al., 1996; Shoemaker et al., 1996; Chee et al., 1996; Lockhart et al., 1996; DeRisi et al., 1996; Lipshutz et al., 1995. This method has already been used to screen people for mutations in the breast cancer gene BRCA1 (Hacia et al., 1996). This new technology has been reviewed in a news article in Chemical and Engineering News (Borman, 1996) and been the subject of an editorial (Editorial, Nature Genetics, 1996). Also see Fodor (1997).

The most definitive test for mutations in a candidate locus is to directly compare genomic KVLQTI, HERG, SCNSA, KCNEI or KCNE2 sequences from patients with those from a control

population. Alternatively, one could sequence messenger RNA after amplification, e. g., by PCR, thereby eliminating the necessity of determining the exon structure of the candidate gene.

Mutations from patients falling outside the coding region of KVLQTI, HERG, SCNSA, KCNEI or KCNE2 can be detected by examining the non-coding regions, such as introns and regulatory sequences near or within the genes. An early indication that mutations in noncoding regions are important may come from Northern blot experiments that reveal messenger RNA molecules of abnormal size or abundance in patients as compared to control individuals.

Alteration of KVLQT1, HERG, SCNSA, KCNE1 or KCNE2 mRNA expression can be detected by any techniques known in the art. These include Northern blot analysis, PCR amplification and RNase protection. Diminished mRNA expression indicates an alteration of the wild-type gene. Alteration of wild-type genes can also be detected by screening for alteration of wild-type KVLQT1, HERG, SCN5A, KCNE1 or KCNE2 protein. For example, monoclonal antibodies immunoreactive with HERG can be used to screen a tissue. Lack of cognate antigen would indicate a mutation. Antibodies specific for products of mutant alleles could also be used to detect mutant gene product. Such immunological assays can be done in any convenient formats known in the art. These include Western blots, immunohistochemical assays and ELISA assays. Any means for detecting an altered KVLQT1, HERG, SCN5A, KCNE1 or KCNE2 protein can be used to detect alteration of wild-type KVLQT1, HERG, SCNSA, KCNE1 or KCNE2 genes. Functional assays, such as protein binding determinations, can be used. In addition, assays can be used which detect KVLQT1, HERG, SCN5A, KCNE1 or KCNE2 biochemical function. Finding a mutant KVLQT1, HERG, SCNSA, KCNEI or KCNE2 gene product indicates alteration of a wild-type KVLQT1, HERG, SCNSA, KCNEl or KCNE2 gene.

Mutant KVLQT1, HERG, SCNSA, KCNE1 or KCNE2 genes or gene products can also be detected in other human body samples, such as serum, stool, urine and sputum. The same techniques discussed above for detection of mutant genes or gene products in tissues can be applied to other body samples. By screening such body samples, a simple early diagnosis can be achieved for hereditary LQTS.

Initially, the screening method involves amplification of the relevant KVLQT1, HERG, SCNSA, KCNE1 or KCNE2 sequence. In another preferred embodiment of the invention, the screening method involves a non-PCR based strategy. Such screening methods include two-step label amplification methodologies that are well known in the art. Both PCR and non-PCR based

screening strategies can detect target sequences with a high level of sensitivity. Further details of these methods are briefly presented below and further descriptions can be found in PCT published application WO 96/05306, incorporated herein by reference.

The most popular method used today is target amplification. Here, the target nucleic acid sequence is amplified with polymerases. One particularly preferred method using polymerase- driven amplification is the polymerase chain reaction (PCR). The polymerase chain reaction and other polymerase-driven amplification assays can achieve over a million-fold increase in copy number through the use of polymerase-driven amplification cycles. Once amplified, the resulting nucleic acid can be sequenced or used as a substrate for DNA probes.

When the probes are used to detect the presence of the target sequences, the biological sample to be analyzed, such as blood or serum, may be treated, if desired, to extract the nucleic acids. The sample nucleic acid may be prepared in various ways to facilitate detection of the target sequence; e. g. denaturation, restriction digestion, electrophoresis or dot blotting. The targeted region of the analyte nucleic acid usually must be at least partially single-stranded to form hybrids with the targeting sequence of the probe. If the sequence is naturally single- stranded, denaturation will not be required. However, if the sequence is double-stranded, the sequence will probably need to be denatured. Denaturation can be carried out by various techniques known in the art.

Analyte nucleic acid and probe are incubated under conditions which promote stable hybrid formation of the target sequence in the probe with the putative targeted sequence in the analyte. The region of the probes which is used to bind to the analyte can be made completely complementary to the targeted region of the genes. Therefore, high stringency conditions are desirable in order to prevent false positives. However, conditions of high stringency are used only if the probes are complementary to regions of the chromosome which are unique in the genome. The stringency of hybridization is determined by a number of factors during hybridiza- tion and during the washing procedure, including temperature, ionic strength, base composition, probe length, and concentration of formamide. Under certain circumstances, the formation of higher order hybrids, such as triplexes, quadraplexes, etc., may be desired to provide the means of detecting target sequences.

Detection, if any, of the resulting hybrid is usually accomplished by the use of labeled probes. Alternatively, the probe may be unlabeled, but may be detectable by specific binding

with a ligand which is labeled, either directly or indirectly. Suitable labels, and methods for labeling probes and ligands are known in the art, and include, for example, radioactive labels which may be incorporated by known methods (e. g., nick translation, random priming or kinasing), biotin, fluorescent groups, chemiluminescent groups (e. g., dioxetanes, particularly triggered dioxetanes), enzymes, antibodies and the like. Variations of this basic scheme are known in the art, and include those variations that facilitate separation of the hybrids to be detected from extraneous materials and/or that amplify the signal from the labeled moiety. A number of these variations are well known.

As noted above, non-PCR based screening assays are also contemplated in this invention.

This procedure hybridizes a nucleic acid probe (or an analog such as a methyl phosphonate backbone replacing the normal phosphodiester), to the low level DNA target. This probe may have an enzyme covalently linked to the probe, such that the covalent linkage does not interfere with the specificity of the hybridization. This enzyme-probe-conjugate-target nucleic acid complex can then be isolated away from the free probe enzyme conjugate and a substrate is added for enzyme detection. Enzymatic activity is observed as a change in color development or luminescent output resulting in a 103-106 increase in sensitivity. For example, the preparation of oligodeoxynucleotide-alkaline phosphatase conjugates and their use as hybridization probes are well known.

Two-step label amplification methodologies are known in the art. These assays work on the principle that a small ligand (such as digoxigenin, biotin, or the like) is attached to a nucleic acid probe capable of specifically binding the target gene. Allele specific probes are also contemplated within the scope of this example.

In one example, the small ligand attached to the nucleic acid probe is specifically recognized by an antibody-enzyme conjugate. In one embodiment of this example, digoxigenin is attached to the nucleic acid probe. Hybridization is detected by an antibody-alkaline phosphatase conjugate which turns over a chemiluminescent substrate. In a second example, the small ligand is recognized by a second ligand-enzyme conjugate that is capable of specifically complexing to the first ligand. A well known embodiment of this example is the biotin-avidin type of interactions. Methods for labeling nucleic acid probes and their use in biotin-avidin based assays are well known.

It is also contemplated within the scope of this invention that the nucleic acid probe assays of this invention will employ a cocktail of nucleic acid probes capable of detecting the gene or genes. Thus, in one example to detect the presence of KVLQT1 in a cell sample, more than one probe complementary to KVLQT1 is employed and in particular the number of different probes is alternatively 2,3, or 5 different nucleic acid probe sequences. In another example, to detect the presence of mutations in the KVLQT1 gene sequence in a patient, more than one probe complementary to KVLQTI is employed where the cocktail includes probes capable of binding to the allele-specific mutations identified in populations of patients with alterations in KVLQT1.

In this embodiment, any number of probes can be used.

Large amounts of the polynucleotides of the present invention may be produced by replication in a suitable host cell. Natural or synthetic polynucleotide fragments coding for a desired fragment will be incorporated into recombinant polynucleotide constructs, usually DNA constructs, capable of introduction into and replication in a prokaryotic or eukaryotic cell.

Usually the polynucleotide constructs will be suitable for replication in a unicellular host, such as yeast or bacteria, but may also be intended for introduction to (with and without integration within the genome) cultured mammalian or plant or other eukaryotic cell lines. The purification of nucleic acids produced by the methods of the present invention are described, e. g., in Sambrook et al., 1989 or Ausubel et al., 1992.

The polynucleotides of the present invention may also be produced by chemical synthesis, e. g., by the phosphoramidite method described by Beaucage and Caruthers (1981) or the triester method according to Matteucci and Caruthers (1981) and may be performed on commercial, automated oligonucleotide synthesizers. A double-stranded fragment may be obtained from the single-stranded product of chemical synthesis either by synthesizing the complementary strand and annealing the strand together under appropriate conditions or by adding the complementary strand using DNA polymerase with an appropriate primer sequence.

Polynucleotide constructs prepared for introduction into a prokaryotic or eukaryotic host may comprise a replication system recognized by the host, including the intended polynucleotide fragment encoding the desired polypeptide, and will preferably also include transcription and translational initiation regulatory sequences operably linked to the polypeptide encoding segment.

Expression vectors may include, for example, an origin of replication or autonomously replicating sequence (ARS) and expression control sequences, a promoter, an enhancer and

necessary processing information sites, such as ribosome-binding sites, RNA splice sites, polyadenylation sites, transcriptional terminator sequences, and mRNA stabilizing sequences.

Such vectors may be prepared by means of standard recombinant techniques well known in the art and discussed, for example, in Sambrook et al. (1989) or Ausubel et al. (1992).

An appropriate promoter and other necessary vector sequences will be selected so as to be functional in the host, and may include, when appropriate, those naturally associated with the KVLQTI or other gene. Examples of workable combinations of cell lines and expression vectors are described in Sambrook et al. (1989) or Ausubel et al. (1992) ; see also, e. g., Metzger et al.

(1988). Many useful vectors are known in the art and may be obtained from such vendors as Stratagene, New England Biolabs, Promega Biotech, and others. Promoters such as the trp, lac and phage promoters, tRNA promoters and glycolytic enzyme promoters may be used in prokaryotic hosts. Useful yeast promoters include promoter regions for metallothionein, 3- phosphoglycerate kinase or other glycolytic enzymes such as enolase or glyceraldehyde-3- phosphate dehydrogenase, enzymes responsible for maltose and galactose utilization, and others.

Vectors and promoters suitable for use in yeast expression are further described in Hitzeman et al., EP 73,675A. Appropriate non-native mammalian promoters might include the early and late promoters from SV40 (Fiers et al., 1978) or promoters derived from murine Molony leukemia virus, mouse tumor virus, avian sarcoma viruses, adenovirus II, bovine papilloma virus or polyoma. Insect promoters may be derived from baculovirus. In addition, the construct may be joined to an amplifiable gene (e. g., DHFR) so that multiple copies of the gene may be made. For appropriate enhancer and other expression control sequences, see also Enhancers and Eukar,ic Gene Expression, Cold Spring Harbor Press, Cold Spring Harbor, New York (1983). See also, e. g., U. S. Patent Nos. 5,691,198; 5,735,500 ; 5,747,469 and 5,436,146.

While such expression vectors may replicate autonomously, they may also replicate by being inserted into the genome of the host cell, by methods well known in the art.

Expression and cloning vectors will likely contain a selectable marker, a gene encoding a protein necessary for survival or growth of a host cell transformed with the vector. The presence of this gene ensures growth of only those host cells which express the inserts. Typical selection genes encode proteins that a) confer resistance to antibiotics or other toxic substances, e. g. ampicillin, neomycin, methotrexate, etc., b) complement auxotrophic deficiencies, or c) supply critical nutrients not available from complex media, e. g., the gene encoding D-alanine

racemase for Bacilli. The choice of the proper selectable marker will depend on the host cell, and appropriate markers for different hosts are well known in the art.

The vectors containing the nucleic acids of interest can be transcribed in vitro, and the resulting RNA introduced into the host cell by well-known methods, e. g., by injection (see, Kubo et al. (1988)), or the vectors can be introduced directly into host cells by methods well known in the art, which vary depending on the type of cellular host, including electroporation; transfection employing calcium chloride, rubidium chloride calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment ; lipofection; infection (where the vector is an infectious agent, such as a retroviral genome); and other methods. See generally, Sambrook et al. (1989) and Ausubel et al. (1992). The introduction of the polynucleotides into the host cell by any method known in the art, including, inter alia, those described above, will be referred to herein as"transformation."The cells into which have been introduced nucleic acids described above are meant to also include the progeny of such cells.

Large quantities of the nucleic acids and polypeptides of the present invention may be prepared by expressing the KVLQT1 nucleic acid or portions thereof in vectors or other expression vehicles in compatible prokaryotic or eukaryotic host cells. The most commonly used prokaryotic hosts are strains of Escherichia coli, although other prokaryotes, such as Bacillus subtilis or Pseudomonas may also be used.

Mammalian or other eukaryotic host cells, such as those of yeast, filamentous fungi, plant, insect, or amphibian or avian species, may also be useful for production of the proteins of the present invention. Propagation of mammalian cells in culture is per se well known. See, Jakoby and Pastan (eds.) (1979). Examples of commonly used mammalian host cell lines are VERO and HeLa cells, Chinese hamster ovary (CHO) cells, and WI38, BHK, and COS cell lines, although it will be appreciated by the skilled practitioner that other cell lines may be appropriate, e. g., to provide higher expression, desirable glycosylation patterns, or other features. An example of a commonly used insect cell line is SF9.

Clones are selected by using markers depending on the mode of the vector construction.

The marker may be on the same or a different DNA molecule, preferably the same DNA molecule. In prokaryotic hosts, the transformant may be selected, e. g., by resistance to ampicillin, tetracycline or other antibiotics. Production of a particular product based on temperature sensitivity may also serve as an appropriate marker.

Prokaryotic or eukaryotic cells transformed with the polynucleotides of the present invention will be useful not only for the production of the nucleic acids and polypeptides of the present invention, but also, for example, in studying the characteristics of KVLQT1 or other polypeptides.

The probes and primers based on the KVLQT1 or other gene sequences disclosed herein are used to identify homologous KVLQT1 or other gene sequences and proteins in other species.

These gene sequences and proteins are used in the diagnostic/prognostic, therapeutic and drug screening methods described herein for the species from which they have been isolated.

The studies described in the Examples below resulted in the determination of many novel mutations. Previous studies had defined 126 distinct disease causing mutations in the LQTS genes KVLQT1, HERG, SCNSA, KCNEI and KCNE2 (Wang Q. et al., 1996a; Curran et al., 1995; Wang et al., 1995a; Splawski et al., 1997a; Abbott et al., 1999; Chouabe et al., 1997; Wollnik et al., 1997; Neyroud et al., 1997; Splawski et al., 1997b; Tyson et al., 1997; Schulze-Bahr et al., 1997; Priori et al., 1999; Splawski et al., 1998; Wang et al., 1995b; Russell et al., 1996; Neyroud et al., 1998; Neyroud et al., 1999; Donger et al., 1997; Tanaka et al., 1997; Jongbloed et al., 1999; Priori et al., 1998 ; Itoh et al., 1998a; Itoh et al., 1998b; Mohammad-Panah et al., 1999; Saarinen et al., 1998; Ackerman et al., 1998 ; Berthet et al., 1999; Kanters, 1998; van den Berg et al., 1997; Dausse et al., 1996; Benson et al., 1996; Akimoto et al., 1998; Satler et al., 1996; Satler et al., 1998; Makita et al., 1998, An et al., 1998; Schulze-Bahr et al., 1995; Duggal et al., 1998; Chen Q. et al., 1999; Li et al., 1998; Wei et al., 1999; Larsen et al., 1999a ; Bianchi et al., 1999; Ackerman et al., 1999a ; Ackerman et al., 1999b ; Murray et al., 1999; Larsen et al., 1999b ; Yoshida et al., 1999; Wattanasirichaigoon et al., 1999; Bezzina et al., 1999; Hoorntje et al., 1999). The sequence of each wild-type gene has been published. The KVLQT1 can be found in Splawski et al. (1998) and the coding region of the cDNA is shown herein as SEQ ID NO : 1 and the encoded KVLQT1 is shown as SEQ ID NO : 2. SCN5A was reported by Gellens et al. (1992) and its sequence is provided by GenBank Accession No. Nom 000335. The coding sequence of SCN5A is shown herein as SEQ ID NO : 3 and the encoded SCN5A is shown as SEQ ID NO : 4.

Most of the mutations were found in KVLQT1 (Yoshida et al., 1999) and HERG (Itoh et al., 1998b), and fewer in SCN5A (Wang Q. et al., 1996a), KCNEI (Jiang et al., 1994) and KCNE2 (Ward, 1964). These mutations were identified in regions with known intron/exon structure, primarily the transmembrane and pore domains. In this study, we screened 262 individuals with

LQTS for mutations in all known arrhythmia genes. We identified 134 mutations, 80 of which were novel. Together with 43 mutations reported in our previous studies, we have now identified 177 mutations in these 262 LQTS individuals (68%). The failure to identify mutations in 32% of the individuals may result from phenotypic errors, incomplete sensitivity of SSCP or presence of mutations in regulatory sequences. However, it is also clear that additional LQTS genes await discovery (Jiang et al., 1994; Schott et al., 1995).

Missense mutations were most common (72%), followed by frameshift mutations (10%), in-frame deletions, nonsense and splice site mutations (5-7% each). Most mutations resided in intracellular (52%) and transmembrane (30%) domains; 12% were found in pore and 6% in extracellular segments. One hundred one of the 129 distinct LQTS mutations (78%) were identified in single families or individuals. Most of the 177 mutations were found in KVLQT1 (75 or 42%) and HERG (80 or 45%). These two genes accounted for 87% of the identified mutations, while mutations in SCN5A (14 or 8%), KCNE1 (5 or 3%) and KCNE2 (3 or 2%) accounted for the other 13%.

Multiple mutations were found in regions encoding S5, S5/P, P and S6 of KVLQT1 and HERG. The P region of potassium channels forms the outer pore and contains the selectivity filter (Doyle et al., 1998). Transmembrane segment 6, corresponding to the inner helix of KcsA, forms the inner 2/3 of the pore. This structure is supported by the S5 transmembrane segment, corresponding to the outer helix of KcsA, and is conserved from prokaryotes to eukaryotes ( (MacKinnon et al., 1998). Mutations in these regions will likely disrupt potassium transport.

Many mutations were identified in the C-termini of KVLQTI and HERG. Changes in the C- terminus of HERG could lead to anomalies in tetramerization as it has been proposed that the C- terminus of eag, which is related to HERG, is involved in this process (Ludwig et al, 1994).

Multiple mutations were also identified in regions that were different for KVLQT1 and HERG. In KVLQT1, multiple mutations were found in the sequences coding for the S2/S3 and S4/S5 linkers. Coexpression of S2/S3 mutants with wild-type KVLQT1 inXenopus oocytes led to simple loss of function or dominant-negative effect without significantly changing the biophysical properties of IKS channels (Chouabe et al., 1997; Shalaby et al., 1997; Wang et al., 1999). On the other hand, S4/S5 mutations altered the gating properties of the channels and modified KVLQT1 interactions with minK subunits (Wang et al., 1999; Franqueza et al., 1999).

In HERG, more than 20 mutations were identified in the N-terminus. HERG channels lacking this region deactivate faster and mutations in the region had a similar effect (Chen J. et al., 1999).

Mutations in KCNE1 and KCNE2, encoding minK and MiRP1, the respective IKS and IKr -subunits, altered the biophysical properties of the channels (Splawski et al., 1997a; Abbott et al., 1999; Sesti and Goldstein, 1998). A MiRP1 mutant, involved in clarithromyocin-induced arrhythmia, increased channel blockade by the antibiotic (Abbott et al., 1999). Mutations in SCNSA, the sodium channel a-subunit responsible for cardiac INa, destabilized the inactivation gate causing delayed channel inactivation and dispersed reopenings (Bennett et al., 1995 ; Dumaine et al., 1996; Wei et al., 1999; Wang DW et al., 1996). One SCN5A mutant affected the interactions with the sodium channel P-subunit (An et al., 1998).

It is interesting to note that probands with KCNE1 and KCNE2 mutations were older and had shorter QTc than probands with the other genotypes. The significance of these differences is unknown, however, as the number of probands with KCNE1 and KCNE2 genotypes was small.

This catalogue of mutations will facilitate genotype-phenotype analyses. It also has clinical implications for presymptomatic diagnosis and, in some cases, for therapy. Patients with mutations in KVLQT1, HERG, KCNEI and KCNE2, for example, may benefit from potassium therapy (Compton et al., 1996). Sodium channel blockers, on the other hand, might be helpful in patients with SCNSA mutations (Schwartz et al. (1995). The identification of mutations is of importance for ion channel studies as well. The expression of mutant channels in heterologous systems can reveal how structural changes influence the behavior of the channel or how mutations affect processing (Zhou et al., 1998; Furutani et al., 1999). These studies improve our understanding of channel function and provide insights into mechanisms of disease. Finally, mutation identification will contribute to the development of genetic screening for arrhythmia susceptibility.

The present invention is described by reference to the following Examples, which are offered by way of illustration and are not intended to limit the invention in any manner. Standard techniques well known in the art or the techniques specifically described in the Examples were utilized.

Example 1 Ascertainment and Phenol Individuals were ascertained in clinics from North America and Europe. Individuals were evaluated for LQTS based on QTc (the QT interval corrected for heart rate) and for the presence of symptoms. In this study, we focused on the probands. Individuals show prolongation of the QT interval (QTc2460 ms) and/or documented torsade de pointes, ventricular fibrillation, cardiac arrest or aborted sudden death. Informed consent was obtained in accordance with local institutional review board guidelines. Phenotypic data were interpreted without knowledge of genotype. Sequence changes altering coding regions or predicted to affect splicing that were not detected in at least 400 control chromosomes were defined as mutations. No changes except known polymorphisms were detected ina ny of the genes in the control population. This does not exclude the possibility that some mutations are rare variants not associated with disease.

Example 2 Mutational Analyses To determine the spectrum of LQTS mutations, we used SSCP (Single Stand Conformation Polymorphism) and DNA sequence analyses to screen 262 unrelated individuals with LQTS. Seventeen primer pairs were used to screen KVLQT1 (Splawski et al., 1998), twenty-one primer pairs were used for HERG (Splawski et al., 1998) and three primer pairs were used for KCNEI (Splawski et al., 1997a) and KCNE2 (Abbott et al., 1999). Thirty-three primer pairs (Wang Q. et al., 1996b) were used in SSCP analysis to screen all SCNSA exons in 50 individuals with suspected abnormalities in INa. Exons 23-28, in which mutations were previously identified, were screened in all 262 individuals.

Gender, age, QTc and presence of symptoms are summarized in Table 1. The average age at ascertainment was 29 with a corrected QT interval of 492 ms. Seventy-five percent had a history of symptoms and females predominated with an ~ 2 : 1 ratio. Although the numbers were small, corrected QT intervals for individuals harboring KCNE1 and KCNE2 mutations were shorter at 457 ms.

Table 1

Age, QTc, Gender and Presence of Symptoms Genotype Age*, y Gender (F/M) QTc, ms Symptoms (meanSD) (mean~SD) KVLQT1 32 ~ 19 52/23 493 ~ 45 78% HERG 31 i 19 51/29 495 ~ 48 71% SCNSA 32 ~ 24 8/6 51142 55% KCNE1 43 ~ 16 3/2 457 i 25 40% KCNE2 54 i 20 3/0 457 ~ 05 67% unknown 25 ~ 16 56/29 484 ~ 46 81% all 29 ~ 19 173/89 492 ~ 47 75% *-age at ascertainment t-symptoms include syncope, cardiac arrest or sudden death The SSCP analyses revealed many mutations. KVLQT1 mutations associated with LQTS were identified in 52 individuals (Figure 1 and Table 2). Twenty of the mutations were novel. HERG mutations were identified in 68 LQTS individuals (Figure 2 and Table 3). Fifty-two of these mutations were novel. SCNSA mutations were identified in eight cases (Figure 3 and Table 4). Five of the mutations were novel. Three novel KCNE1 mutations were identified (Figure 4 and Table 5) and three mutations were identified in KCNE2 (Figure 5 and Table 6) (Abbott et al., 1999). None of the KVLQTI, HERG, SCN5A, KCNE1 and KCNE2 mutations was observed in 400 control chromosomes.

Table 2 Summary of All KVLQT1 Mutations* Nucleotide Coding Position Exon Number Study Change Effect of families del211-219 del71-73 N-terminus 1 1 Ackerman et al., 1999a A332G # Y111C N-terminus 1 1 This

Nucleotide Coding Position Exon Number Study Changet Effect of . familles del451-452 A150fs/132 S2 2 1 JLN Chen Q. et al., 1999 T470G F157C S2 1 1 Larsen et al., 1999a G477+1A M159sp S2 2 1 JLN, This ; Donger et al., 1997 1 UK G477+5A M159sp S2 1 1 Ackerman et al., 1999b G478A t E160K S2 3 1 This del500-502 F167W/del S2 3 1 Wang Q. et al., 1996a G168 G502A G168R S2 3 7 This ; Splawski et al., 1998; Donger et al., 1997 C520T R174C S2/S3 3 1 Donger et al., 1997 G521A R174H S2/S3 3 1 This G532A A178T S2/S3 3 1 Tanaka et al., 1997 G532C A178P S2/S3 3 1 Wang Q. et al., 1996a G535A G179S S2/S3 3 1 This A551C Y184S S2/S3 3 2 This ; Jongbloed et al., 1999 G565A G189R S2/S3 3 3 Wang Q. et al., 1996a; Jongbloed et al., 1999 insG567-G189fs/94 S2/S3 3 1 (RW + Splawski et al., 1997b 568 JLN) G569A R190Q S2/S3 3 2 Splawski et al., 1998; Donger et al., 1997 del572-576 L191fs/90 S2/S3 3 1 JLN, Tyson et al., 1997; 1 RW Ackerman et al., 1999b 2 (JLN + RW)

Nucleotide Coding Position Exon Number Study Change Effect of families G580C A194P S2/S3 3 1 This C674T S225L S4 4 2 This; Priori et al., 1999 G724A D242N S4/S5 5 1 Itoh et al., 1998b C727T t R243C S4/S5 5 2 This G728A R243H S4/S5 5 1 JLN Saarinen et al., 1998 T742C t W248R S4/S5 5 1 This T749A L250H S4/S5 5 1 Itoh et al., 1998a G760A V254M S4/S5 5 4 This; Wang Q. et al., 1996a; Donger et al., 1997 G781A E261K S4/S5 6 1 Donger et al., 1997 T797C t L266P S5 6 1 This G805A G269S S5 6 1 Ackerman et al., 1999b G806A G269D S5 6 3 This; Donger et al., 1997 C817T L273F S5 6 2 This; Wang Q. et al., 1996a A842G Y281C S5 6 1 Priori et al., 1999 G898A A300T S5/Pore 6 1 Priori et al., 1998 G914C W305S Pore 6 1 JLN Chouabe et al., 1997 G916A G306R Pore 6 1 Wang Q. et al, 1996a del921-V307sp Pore 6 1 Li et al., 1998 (921+2) G921+1T t V307sp Pore 6 1 This A922-2C t V307sp Pore 7 1 This G922-1 C V307sp Pore 7 1 Murray et al., 1999 C926G T309R Pore 7 1 Donger et al., 1997

Nucleotide Coding Position Exon Number Study Change Effect of families G928A V3101 Pore 7 1 This C932T T311I Pore 7 1 Saarinen et al., 1998 C935T T312I Pore 7 2 This; Wang Q. et al., 1996a C939G I313M Pore 7 1 Tanaka et al., 1997 G940A G314S Pore 7 7 Splawski et al., 1998; Russell et al., 1996; Donger et al., 1997; Jongbloed et al., 1999; Itoh et al., 1998b A944C Y315S Pore 7 3 Donger et al., 1997; Jongbloed et al., 1999 A944G Y315C Pore 7 2 Priori et al., 1999; Splawski et al., 1998 G949A D317N Pore 7 2 Wollnik et al., 1997; Saarinen et al., 1998 G954C K318N Pore 7 1 Splawski et al., 1998 C958G P320A Pore 7 1 Donger et al., 1997 G973A G325R S6 7 4 This; Donger et al., 1997; Tanaka et al., 1997 dellO17-delF340 S6 7 2 This; Ackerman et al., 1019 1998 C1022A A341E S6 7 5 This; Wang Q. et al., 1996a; Berthet et al., 1999

Nucleotide Coding Position Exon Number Study Changet Effect of _ families t C1022T A341V S6 7 7 This; Wang Q. et al., 1996a; Russell et al., 1996; Donger et al., 1997; Li et al., 1998 C1024T L342F S6 7 1 Donger et al., 1997 C1031T A344V S6 7 1 Donger et al., 1997 G1032A A344sp S6 7 9 This; Kanters, 1998; Li et al., 1998; Ackerman et al., 1999b ; Murray et al., 1999 G1032C A344sp S6 7 1 Murray et al., 1999 G1033C G345R S6 8 1 van den Berg et al., 1997 G1034A G345E S6 8 1 Wang Q. et al., 1996a C1046G t S349W S6 8 1 This T1058C L353P S6 8 1 Splawski et al., 1998 C1066T f Q356X C-terminus 8 1 This C1096T R366W C-terminus 8 1 Splawski et al., 1998 G1097A t R366Q C-terminus 8 1 This G1097C R366P C-terminus 8 1 Tanaka et al., 1997 G1111A A371T C-terminus 8 1 Donger et al., 1997 T1117C S373P C-terminus 8 1 Jongbloed et al., 1999 C1172T t T391I C-terminus 9 1 This T1174C W392R C-terminus 9 1 Jongbloed et al., 1999 C1343G t P448R C-terminus 10 2 This C1522T R518X C-terminus 12 1 JLN, This; Larsen et al., 1999 3 RW

Nucleotide Coding Position Exon Number Study Changet Effect of families G1573A A525T C-terminus 12 1 Larsen et al., 1999b C1588T t Q530X C-terminus 12 1 JLN, This 1 RW C1615T R539W C-terminus 13 1 Chouabe et al., 1997 del6/ins7 E543fs/107 C-terminus 13 1 JLN Neyroud et al., 1997 C1663T R555C C-terminus 13 3 Donger et al., 1997 C1697Tf S566F C-terminus 14 3 This C1747T R583C C-terminus 15 1 This C1760T T587M C-terminus 15 1 JLN, Donger et al., 1997; 1 RW Itoh et al., 1998b G1772A R591H C-terminus 15 1 Donger et al., 1997 G1781A t R594Q C-terminus 15 3 This dell892-P630fs/13 C-terminus 16 1 JLN Donger et al., 1997 1911 insC1893-P631fs/19 C-terminus 16 1 Donger et al., 1997 1894 *-ins denotes insertion; del denotes deletion; sp denotes the last unaffected amino acid

before the predicted splice mutation; fs denotes the last amino acid unaffected by a frameshift, following fs is the number of amino acids before termination ; X denotes a stop codon occurred. t-denotes novel mutation t-Number of Romano-Ward families unless otherwise indicated (UK-unknown) Table 3

Summary of All HERG Mutations* Nucleotide Coding Position Exon Number Study Change Effect of RW Families C87A t F29L N-terminus 2 1 This A98C N33T N-terminus 2 2 This C132A# C44X N-terminus 2 1 This G140T# G47V N-terminus 2 1 This G157C G53R N-terminus 2 1 This G167A# R56Q N-terminus 2 1 This T196G C66G N-terminus 2 1 This A209G# H70R N-terminus 2 2 This C215A # P72Q N-terminus 2 2 This del221-251 t R73fs/31 N-terminus 2 1 This G232C# A78P N-terminus 2 1 This dupl234-250t A83fs/37 N-terminus 2 1 This C241T t Q81X N-terminus 2 1 This T257G # L86R N-terminus 2 1 This insC422-423f P141fs/2 N-terminus 3 1 This insC453-454t P151fs/N-terminus 3 1 This 179 dupl558-600 L200fs/N-terminus 4 1 Hoorntje et al., 1999 144 insC724-725t P241fs/89 N-terminus 4 1 This del885 t V295fs/63 N-terminus 4 1 This C934T t R312C N-terminus 5 1 This C1039T t P347S N-terminus 5 1 This G1128A# Q376sp N-terminus 5 1 This

Nucleotide Coding Position Exon Number Study Change Effect of RW Families A1129-2G t Q376sp N-terminus 6 1 This dell261 Y420fs/12 si 6 1 Curran et al., 1995 C1283A S428X S1/S2 6 1 Priori et al., 1999 C1307T T436M S1/S2 6 1 Priori et al., 1999 A1408G N470D S2 6 1 Curran et al., 1995 C1421T T474I S2/S3 6 1 Tanaka et al., 1997 C1479G Y493X S2/S3 6 1 Itoh et al., 1998a dell498 1524 del500-S3 6 1 Curran et al., 1995 508 G1592A # R531Q S4 7 1 This C1600T R534C S4 7 1 Itoh et al., 1998a T1655C t L552S S5 7 1 This doIT1671 T556fs/7 S5 7 1 Schulze-Bahr et al., 1995 G1672C A558P S5 7 1 Jongbloed et al., 1999 G1681A A561T S5 7 4 This ; Dausse et al., 1996 C1682T A561V S5 7 4 This; Curran et al., 1995; Priori et al., 1999 G1714C G572R S5/Pore 7 1 Larsen et al., 1999a G1714T G572C S5/Pore 7 1 Splawski et al., 1998 C1744T R582C S5/Pore 7 1 Jongbloed et al., 1999 G1750A t G584S S5/Pore 7 1 This G1755T t W585C S5/Pore 7 1 This A1762G N588D S5/Pore 7 1 Splawski et al., 1998 T1778C t I593T S5/Pore 7 1 This T1778G I593R S5/Pore 7 1 Benson et al, 1996 G1801A G601S S5/Pore 7 1 Akimoto et al., 1998

Nucleotide Coding Position Exon Number Study Change Effect of RW Families G1810A'G604S S5/Pore 7 2 This; Jongbloed et al., 1999 G1825At D609N S5/Pore 7 1 This T1831C Y611H S5/Pore 7 1 Tanalca et al., 1997 T1833 (Aor Y611X S5/Pore 7 1 Schulze-Bahretal., 1995 G) G1834T V612L Pore 7 1 Satler et al., 1998 C1838T T613M Pore 7 4 This; Jongbloed et al., 1999 C1841T A614V Pore 7 6 Priori et al., 1999; Splawski et al., 1998; Tanaka et al.,1997; Satler et al., 1998 C1843G t L615V Pore 7 1 This G1876A t G626S Pore 7 1 This C1881G t F627L Pore 7 1 This G1882A G628S Pore 7 2 This; Curran et al., 1995 A1885G N629D Pore 7 1 Satler et al., 1998 A1886G N629S Pore 7 1 Satler et al., 1998 C1887A N629K Pore 7 1 Yoshida et al., 1999 G1888C V630L Pore 7 1 Tanaka et al., 1997 T1889C V630A Pore 7 1 Splawski et al., 1998 C1894T t P632S Pore 7 1 This A1898G N633S Pore 7 1 Satler et al., 1998 A1912G t K638E S6 7 1 This dell913-1915t delK638 S6 7 1 This

Nucleotide Coding Position Exon Number Study Change Effect of RW Families C1920A F640L S6 7 1 Jongbloed et al., 1999 A1933T t M645L S6 7 1 This de11951-1952 L650fs/2 S6 8 1 Itoh et al., 1998a G2044T t E682X S6/cNBD 8 1 This C2173T Q725X S6/cNBD 9 1 Itoh et al., 1998a insT2218-H739fs/63 S6/cNBD 9 1 This 2219# C2254T # R752W S6/cNBD 9 1 This dupl2356-2386 V796fs/22 cNBD 9 1 Itoh et al., 1998a del2395 t I798fs/10 cNBD 9 1 This G2398+1C L799sp cNBD 9 2 This; Curran et al., 1995 T2414C t F805S cNBD 10 1 This T2414G # F805C cNBD 10 1 This C2453T S818L cNBD 10 1 Berthet et al., 1999 G2464A V822M cNBD 10 2 Berthet et al., 1999; Satler et al., 1996 C2467T# R823W cNBD 10 2 This A2582T N861I C-terminus 10 1 This G2592+1A D864sp C-terminus 10 2 This; Berthet et al., 1999 del2660 K886fs/85 C-terminus 11 1 This C2750T# P917L C-terminus 12 1 This del2762 R920fs/51 C-terminus 12 1 This C2764T# R922W C-terminus 12 1 This insG2775-G925fs/13 C-terminus 12 1 This 2776# del2906# P968fs/4 C-terminus 12 1 This

Nucleotide Coding Position Exon Number Study Change Effect of RW Families del2959-2960t P986fs/C-terminus 12 1 This 130 C3040T R1014X C-terminus 13 2 This del3094f G1031fs/C-terminus 13 1 This 24 insG3107-G1036fs/C-terminus 13 1 Berthet et al., 1999 3108 82 insC3303-P1101fs C-terminus 14 1 This 3304# *-all characters same as in Table 2 Table 4 Summary of All SCN5A Mutations Nucleotide Coding Position Exon Number Study Change Effect of RW Families G3340A D1114N DII/DIII 18 1 This C3911T T1304M DIII/S4 22 1 Wattanasirichaigoon et al., 1999 A3974G N1325S DIII/S4/S5 23 1 Wang et al., 1995b C4501G t L1501V DIII/DIV 26 1 This del4511-dell505-DIII/DIV 26 4 Wang et al., 1995a; Wang et 4519 1507 al., 1995b del4850-delF1617 DIV/S3/S4 28 1 This 4852# G4868A R1623Q DIV/S4 28 2 This; Makita et al., 1998 G4868T R1623L DIV/S4 28 1 This

Nucleotide Coding Position Exon Number Study Change Effect of RW Families G4931A R1644H DIV/S4 28 2 This; Wang et al., 1995b C4934T T1645M DIV/S4 28 1 Wattanasirichaigoon et al., 1999 G5350A E1784K C-terminus 28 2 This; Wei et al., 1999 G5360A# S1787N C-terminus 28 1 This A5369G D1790G C-terminus 28 1 An et al., 1998 insTGA insD 1795 C-terminus 28 1 Bezzina et al., 1999 5385-5386-1796 *-all characters same as in Table 2. Fifty individuals with suspected abnormalities in INa were screened for all SCNSA exons. All individuals were screened for exons 23-28.

Table 5 Summary of All KCNEI Mutations' Nucleotide Coding Position Exon Number of Study Change Effect Families C20T T7I N-terminus 3 1 JLN Schulze- Bahr et al., 1997 G95A R32H N-terminus 3 1 This G139T V47F si 3 1 JLN Bianchi et al., 1999 TG151-L51 H S 1 3 1 JLN Bianchi et 152AT al., 1999 A172C/TG TL58-59PP S1 3 1 JLN Tyson et al., 176-177CT 1997 C221 T S74L C-terminus 3 1 Splawski et al., 1997a

Nucleotide Coding Position Exon Number of Study Change Effect Families G226A D76N C-terminus 3 1 JLN, Splawski et 1 RW, al., 1997a; 1 (JLN + RW) Tyson et al., 1997; Duggal et al., 1998 T259C W87R C-terminus 3 1 Bianchi et al., 1999 C292T R98W C-terminus 3 1 This C379A P127T C-terminus 3 1 This *-all characters same as in Table 2 Table 6

Summary of All KCNE2 Mutations Nucleotide Coding Position Exon Number Study Change Effect of Families C25G Q9E N-terminus 1 1 Abbott et al., 1999 T161T M54T si 1 1 Abbott et al., 1999 T170C I57T S1 1 1 Abbott et al., 1999 Table 7

Mutations by Type Type KVLQTI HERG SCN5A KCNEI KCNE2 Total Missense 59 52 9 5 3 128 Nonsense 6 5 0 0 0 11 AA deletion* 2 2 5 0 0 9 Frameshift 1 16 0 0 0 17 Splice 7 5 0 0 0 12 Total 75 80 14 5 3 177 *-AA denotes amino acid Table 8 Mutations by Position Gene KVLQTI HERG SCNSA KCNEI KCNE2 Protein KVLQT1 HERG SCN5A minK MiRPI Position Total Extracellular 0 7 1 1 1 10 Transmembrane 33 13 5 0 2 53 Pore 9 12 0 N/A N/A 21 Intracellular 33 48 8 4 0 93 Total 75 80 14 5 3 177 While the invention has been disclosed in this patent application by reference to the details of preferred embodiments of the invention, it is to be understood that the disclosure is intended in an illustrative rather than in a limiting sense, as it is contemplated that modifications will readily occur to those skilled in the art, within the spirit of the invention and the scope of the appended claims.

LIST OF REFERENCES Abbott GW, et al. (1999). Cell 97 : 175-187.

Ackerman MJ, et al. (1998). Pediatr. Res. 44: 148-153.

Ackerman MJ, et al. (1999a). N. Engl. J Med 341: 1121-1125.

Ackerman MJ, et al. (1999b). Mayo Clin. Proc. 74: 1088-1094.

Akimoto K, et al. (1998). Hum. Mutat. 1 : S184 S186.

An RH, et al. (1998). Circ. Res. 83: 141-146.

Ausubel FM, et al. (1992). Current Protocols in Molecular Biology, (John Wiley and Sons, New York, New York).

Barhanin J, et al. (1996). Nature 384 : 78-80.

Beaucage SL and Caruthers MH (1981). Tetra. Letts. 22: 1859-1862.

Bennett PB, et al. (1995). Nature 376: 683-685.

Benson DW, et al. (1996). Circulation 93: 1791-1795.

Berthet M, et al. (1999). Circulation 99: 1464-1470.

Bezzina C, et al. (1999). Circ. Res. 85: 1206-1213.

Bianchi L, et al. (1999). Hum. Mol. Genet. 8: 1499-1507.

Borman S (1996). Chemical & EngineeringNews, December 9 issue, pp. 42-43.

Cariello NF (1988). Am. J. Human Genetics 42: 726-734.

Chee M, et al. (1996). Science 274: 610-614.

Chen J, et al. (1999). J Biol. Chem. 274: 10113-10118.

Chen Q, et al. (1999). Circulation 99: 1344-1347.

Chouabe C, et al. (1997). EMBO J. 16: 5472-5479.

Compton SJ, et al. (1996). Circulation 94: 1018-1022.

Conner BJ, et al. (1983). Proc. Natl. Acad. Sci. USA 80: 278-282.

Cotton RG, et al. (1988). Proc. Natl. Acad. Sci. USA 85: 4397-4401.

Curran ME, et al. (1995). Cell 80 : 795-803.

Dausse E, et al. (1996). J ; Mol. Cellg Cardiol. 28: 1609-1615.

DeRisi J, et al. (1996). Nat. Genet. 14: 457-460.

Donger C, et al. (1997). Circulation 96: 2778-2781.

Doyle DA, et al. (1998). Science 280: 69-77.

Duggal P, et al. (1998). Circulation 97: 142-146.

Dumaine R, et al. (1996). Circ. Res. 78: 914-924.

Editorial (1996). Nature Genetics 14: 367-370.

Elghanian R, et al. (1997). Science 277: 1078-1081.

Enhancers and Eukaryotic Gene Expression, Cold Spring Harbor Press, Cold Spring Harbor, New York (1983).

Fiers W, et al. (1978). Nature 273: 113-120.

Finkelstein J, et al. (1990). Genomics 7: 167-172.

Fodor SPA (1997). Science 277: 393-395.

Franqueza L, et al. (1999). J Biol. Chem. 274: 21063-21070.

Furutani M, et al. (1999). Circulation 99: 2290-2294.

Gellens M, et al. (1992). Proc. Natl. Acad. Sci. USA 89 : 554-558.

Grompe M (1993). Nature Genetics 5: 111-117.

Grompe M, et al., (1989). Proc. Natl. Acad. Sci. USA 86 : 5855-5892.

Hacia JG, et al. (1996). Nature Genetics 14: 441-447.

Hoorntje T, et al. (1999). Circulation 100: 1264-1267.

Itoh T, et al. (1998a). Hum. Genet. 102: 435-439.

Itoh T, et al. (1998b). Hum. Genet. 103: 290-294.

Jakoby WB and Pastan IH (eds.) (1979). Cell Culture. Methods in Enzymology volume 58 (Academic Press, Inc., Harcourt Brace Jovanovich (New York)).

Jervell A and Lange-Nielsen F (1957). Am. Heart J. 54: 59-68.

Jiang C, et al. (1994). Nat. Genet. 8: 141-147.

Jongbloed RJ, et al. (1999). Hum. Mutat. 13: 301-310.

Kanters J (1998). J Cardiovasc. Electrophysiol. 9: 620-624.

Keating M, et al. (1991). Science 252 ; 704-706.

Kinszler KW, et al. (1991). Science 251: 1366-1370.

Kubo T, et al. (1988). FEBS Lett. 241: 119.

Larsen LA, et al. (1999a). Hum. Mutat. 13: 318-327.

Larsen LA, et al. (1999b). Eur. J Hum. Genet. 7: 724-728.

Li H, et al. (1998). Circulation 97: 1264-1269.

Lipshutz RJ, et al. (1995). Biotechniques 19: 442-447.

Lockhart DJ, et al. (1996). Nature Biotechnology 14: 1675-1680.

Ludwig J, et al. (1994). EMBO J 13: 4451-4458.

MacKinnon R, et al. (1998). Science 280: 106-109.

Makita N, et al. (1998). FEBSLett. 423: 5-9.

Matteucci MD and Caruthers MH (1981). J Am. Chem. Soc. 103: 3185.

Metzger D, et al. (1988). Nature 334: 31-36.

Modrich P (1991). Ann. Rev. Genet. 25: 229-253.

Mohammad-Panah R, et al. (1999). Am. J. Hum. Genet. 64: 1015-1023.

Moss A, et al. (1991). Circulation 84: 1136-1144.

Murray A, et al. (1999). Circulation 100: 1077-1084.

Newton CR, et al. (1989). Nucl. Acids Res. 17: 2503-2516.

Neyroud N, et al. (1997). Nat. Genet. 15: 186-189.

Neyroud N, et al. (1998). Eur. J. Hum. Genet. 6: 129-133.

Neyroud N, et al. (1999). Circ. Res. 84: 290-297.

Novack DF, et al. (1986). Proc. NatZ. Acad. Sci. USA 83: 586-590.

Orita M, et al. (1989). Proc. Natl. Acad. Sci. USA 86: 2766-2770.

Priori SG, et al. (1998). Circulation 97: 2420-2425.

Priori SG, et al. (1999). Circulation 99: 529-533.

Romano C, et al. (1963). Clin. Pediatr. 45: 656-683.

Ruano G and Kidd KK (1989). Nucl. Acids Res. 17: 8392.

Russell MW, et al. (1996). Hum. Mol. Genet. 5: 1319-1324.

Saarinen K, et al. (1998). Hum. Mutat. 11: 158-165.

Sambrook J, et al. (1989). Molecular Cloning : A Laboratory Manual, 2nd Ed. (Cold Spring Harbor Laboratory, Cold Spring Harbor, New York).

Sanguinetti MC, et al. (1996a). Nature 384: 80-83.

Sanguinetti MC, et al. (1996b). Proc. Natl. Acad. Sci. USA 93 : 2208-2212.

Satler CA, et al. (1996). Am. J. Med. Genet. 65: 27-35.

Satler CA, et al. (1998). Hum. Genet. 102: 265-272.

Schott J, et al. (1995). Am. J : Hum. Genet. 57: 1114-1122.

Schulze-Bahr E, et al. (1995). N. Engl. J Med. 333: 1783-1784.

Schulze-Bahr E, et al. (1997). Nat. Genet. 17: 267-268.

Schwartz PJ, et al. (1975). Am. Heart J. 89: 378-390.

Schwartz PJ, et al. (1995). Circulation 92: 3381-3386.

Sesti F and Goldstein SA (1998). J. Gen. Physiol. 112: 651-663.

Shalaby FY, et al. (1997). Circulation 96: 1733-1736.

Sheffield VC, et al. (1989). Proc. Natl. Acad. Sci. USA 86: 232-236.

Sheffield VC, et al. (1991). Am. J. Hum. Genet. 49: 699-706.

Shenk TE, et al. (1975). Proc. Natl. Acad. Sci. USA 72: 989-993.

Shoemaker DD, et al. (1996). Nature Genetics 14: 450-456.

Splawski I, et al. (1997a). Nat. Genet. 17: 338-340.

Splawski I, et al. (1997b). N. Engl. J. Med. 336: 1562-1567.

Splawski I, et al. (1998). Genomics 51: 86-97.

Tanaka T, et al. (1997). Circulation 95: 565-567.

Tyson J, et al. (1997). Hum. Mol. Genet. 6: 2179-2185. van den Berg MH, et al. (1997). Hum. Genet. 100: 356-361.

Vetter DE, et al. (1996). Neuron 17: 1251-1264.

Vincent GM, et al. (1992). N. Engl. J. Med. 327: 846-852.

Wang DW (1996). Proc. Natl. Acad. Sci. USA 93: 13200-13205.

Wang Q, et al. (1995a). Cell 80: 805-811.

Wang Q, et al. (1995b). Hum. Mol. Genet. 4 : 1603-1607.

Wang Q, et al. (1996a). Nat. Genet. 12: 17-23.

Wang Q, et al. (1996b). Genomics 34: 9-16.

Wang Z, et al. (1999). J Cardiovasc. Electrophysiol. 10: 817-826.

Ward OC (1964). J. Ir. Med. Assoc. 54: 103-106.

Wartell RM, et al. (1990). Nucl. Acids Res. 18: 2699-2705.

Wattanasirichaigoon D, et al. (1999). Am. J. Med. Genet. 86: 470-476.

Wei J, et al. (1999). Circulation 99: 3165-3171.

White MB, et al. (1992). Genomics 12: 301-306.

Wollnik B, et al. (1997). Hum. Mol. Genet. 6: 1943-1949.

Yoshida H, et al. (1999). J Cardiovasc. Electrophysiol. 10: 1262-1270.

Zhou Z, et al. (1998). J : Biol. Chem. 273: 21061-21066.

Hitzeman et al., EP 73,675A.

European Patent Application Publication No. 0332435. U. S. Patent No. 5,436,146 U. S. Patent No. 5,691,198 U. S. Patent No. 5,735,500 U. S. Patent No. 5,747,469