STRUCTURAL MUTATIONS IN TITIN CAUSE DILATED CARDIOMYOPATHY

Abstract
Provided herein are diagnostic markers and methods for identifying a subject having an increased susceptibility for developing or having dilated cardiomyopathy. The method comprises determining if the subject has a mutation in the TTN nucleic as acid or titin polypeptide. Further provided herein are methods of treating subjects having or at risk of having dilated cardiomyopathy.
Description
TECHNOLOGICAL FIELD

The technology described herein relates to determining if a subject is at increased risk of developing dilated cardiomyopathy. The technology described herein further relates to methods and compositions for treating dilated cardiomyopathy.


BACKGROUND

Gene mutations have been implicated as a cause of cardiomyopathy. For example, variations in over 40 genes, most of which encode components of the sarcomere, the cytoskeleton, or the nuclear lamina, have been demonstrated or posited to cause dilated cardiomyopathy (DCM) (Ahmad et al. Annual review of genomics and human genetics 2005; 6:185-216; Dellefave L, Mcnally E M. Current Opinion in Cardiology 2010; 25(3):198-204). Further, while clinical evaluation identifies affected or likely-affected family members in 30 to 50% of DCM cases (Michels et al. N Engl J Med 1992; 326(2):77-82; Baig et al. Journal of the American College of Cardiology 1998; 31(1):195; Mestroni et al. Journal of the American College of Cardiology 1999; 34(1):181-90), implicating a genetic etiology, pathogenic mutations have been found in only 20 to 30% of cases (Zimmerman et al. Genet Med 2010; 12(5):268-78).


TTN, the gene encoding titin, has been implicated in cardiomyopathy, but has been incompletely studied due to technical challenges posed by the monumental size of its coding sequence (˜100 kb). Titin is the largest human protein (˜33,000 amino acids) and the third most abundant striated muscle protein (Trinick et al. J Mol Biol 1984; 180(2):331-56)


SUMMARY

The inventors have discovered inter alia that certain mutations in the TTN gene sequence are associated with an increased risk of having or developing DCM and/or heart failure. Further, the inventors have discovered that certain TTN mutations which result in truncated versions of the titin protein being produced are much more widespread in subjects with DCM as compared to healthy controls as well as control subjects suffering from other forms of cardiomyopathy. Usually, mutations that truncate titin increase the risk of developing DCM and/or heart failure by 100-200 fold. Accordingly, the methods and compositions described herein relate to methods and compositions for determining whether a subject is at increased risk for having or developing DCM as well as for treating a subject determined to have or be at risk of developing DCM.


Accordingly, provided herein is an assay for determining if a subject has an increased risk for developing a dilated cardiomyopathy (DCM) or is in need of treatment to prevent further development of DCM or progression towards DCM. The assay comprises detecting a mutation in the TTN gene, which results in a truncated (shortened) titin polypeptide. Presence of such a mutation in the nucleic acid sample indicates that the subject has an increased risk for developing DCM and/or heart failure.


In some embodiments, detection of the mutation can be by sequencing of a TTN gene derived-nucleic acid. Sequencing of the nucleic acid sample can be carried out using any nucleic acid sequencing known to one of ordinary skill in the art. Exemplary nucleic acid sequencing methods and systems include, but are not limited to, Maxam-Gilbert sequencing, dye-terminator sequencing, Lynx Therapeutics' Massively Parallel Sequencing (MPSS) Polony sequencing, 454 Pyrosequencing, Illumina (Solexa) sequencing, SOLiD™ sequencing, Single Molecule SMART™ sequencing, Single Molecule real time (RNAP) sequencing, Nanaopore DNA sequencing, sequencing by technology from VisiGen Biotechnologies, and the like.


In some embodiments, the mutation is predicted to result in a titin polypeptide lacking or missing part of the pro-band or the A-band region in the individual carrying the mutation.


Also provided herein is a method of treating a subject destined to develop dilated cardiomyopathy and/or heart failure. The method comprises selecting a subject at risk for developing DCM or in need of treatment for DCM or pre-DCM using an assay described herein and administering a treatment for DCM to the subject.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 depicts the spatial distribution of TTN structural mutations. Titin (solid, dark grey lines in left-most portion of the diagram) is the third major filament of the cardiac sarcomere, in addition to the thick and thin (light grey) filaments. The titin protein is linearly depicted with its Z-disk region, I-band region, A-band region, and M-band region demarcated. Titin isoform classes and sequence variants are shown relative to the titin UniProt sequence (Q8WZ42) below. In cardiac tissue, TTN expresses many titin isoforms that span the sarcomere and are classified as N2BA or N2B, as well as the shorter and less abundant novex-3 titin. The exon specific to novex-3 titin is marked (black). The locations of splicing and copy-number mutations (top row of mutation marks) and nonsense and frameshift mutations (middle row of mutation marks, except above the M-band region) identified in DCM subjects, including two frameshift mutations previously reported to be linked to DCM (Table 16), and truncating variants in control and HCM subjects (bottom row of mutation marks) are indicated with vertical bars. Truncation mutations (middle row of mutation marks in the M-band region) previously identified in patients with congenital myopathy (light grey) or limb-girdle muscular dystrophy (dark grey) are also indicated. Overlapping mutations are stacked horizontally and appear as thicker bars.



FIGS. 2A-2D depict the clinical onset and progression in DCM caused by TTN mutations. Kaplan-Meier curves comparing ages at diagnosis (FIG. 2A) and freedom from cardiac transplant, left ventricular assist device implantation, and death (FIG. 2B) of subjects in DCM-A, B, C cohorts with (N=67) and without (N=228) TTN truncating mutations. Kaplan-Meier curves comparing freedom from cardiac transplant, left ventricular assist device implantation, and death among 94 TTN truncation mutation carriers from 19 families (FIG. 5) classified according to mutation type (FIG. 2C) and gender (FIG. 2D).



FIGS. 3A-3B depict myocardial histopathology of subjects with TTN structural mutations. Light microscopy of hematoxylin and eosin stained specimens from the cardiac interventricular septum of subject MEK-111 (FIG. 3A) or MGW-11 (FIG. 3B). Myocyte nuclei with abnormal morphology are indicated (arrowheads).



FIGS. 4A-4B depict Kaplan-Meier curves displaying (FIG. 4A) age of clinical diagnosis and (FIG. 4B) freedom cardiac transplantation, ventricular assist device and/or death among subjects with DCM from group A, group B, and group C.



FIG. 5 depicts pedigrees of families with TTN truncation mutations. Probands (arrow) and family members are from group A or group C of the DCM cohort. Clinical status defined by cardiac evaluations and/or medical records is indicated: black, DCM; white, unaffected; grey, status uncertain due to age≦40 years, and/or confounding cardiac diagnoses; slash, deceased. Genotypes (+, TTN mutation present; −, mutation absent) are indicated.



FIGS. 6A-6D demonstrate that TTN mutation (c.44725+2delT) disrupts normal splicing. The consequences of a single A/T basepair deletion at chr2:179477886 (identified in genomic DNA from subject MIV-14) on splicing was assessed by sequencing RNA from the subject's explanted left ventricular tissue. (FIG. 6A): IGV (Integrated Genomics View) screenshot of ˜950 bp of genomic sequence from subject MIV-14. Eight reads are displayed. Among the 188 50-basepair reads aligned at residue chr2:179477886, 98 reads predicted the A/T deletion (Tables 6 and 8). (FIG. 6B) IGV screenshot of aligned sequences derived from RNA sequence of left ventricular tissue. Reads were aligned using TOPHAT. A BAM file of RNA sequences corresponding to genomic region in A is displayed. Ten of 11 sequences show normal RNA splicing. One read (denoted as exon skip) omitted exon 204. Six other reads that were not aligned by TOPHAT (not shown) also indicate abnormal splicing. (FIG. 6C) IGV screenshot of TTN exons and introns in the 950 bp region flanking chr2:179477886. Note that normal splicing (indicated in panel B) includes exon 204. Primers (denoted pF and pR) designed to correspond to sequences in exon 203 and 205 are: pF:CCATCATGTTCTGGTTTTGTCCAATTCAACCTTACT (SEQ ID NO: 3) and pR: CAATCACAGGATACTGGGTTGAAAGACTGGA (SEQ ID NO:4). (FIG. 6D) MIV-14 RNA was RT-PCR amplified using primer pF and pR and size fractionated by gel electrophoresis (3% agarose). Lanes (left to right) are: 50 bp ladder, control A, control B, and MIV14. PCR products corresponding to WT-splicing and MIV-14 exon skip splicing were analyzed by dideoxy sequencing (data not shown).



FIG. 7 demonstrates confirmation of a large ITN tandem-insertion in MEQ-132. Primer pairs TTN213F and TTN213R (amplimer TTN21), TTN155F and TTN155R (amplimers TTN155), and TTN37+intF1 and TTN_i22_R1 (predicted break-point) were used for PCR amplification of genomic DNA from proband MEQ-132, unaffected relatives (MEQ-130, MEQ-131) and an unrelated control sample (CTL). The lane denoted by ‘−’ contained no genomic DNA. The predicted size of the PCR product was 292 bp.



FIG. 8 depicts a transmission electron micrograph of a cardiac left ventricular free wall specimen, derived at autopsy, from DCM-A subject MGW-11 who carries a TTN nonsense mutation (p.Arg26949X). Sarcomere structure is normal. The myocyte nucleus is highly lobulated.



FIG. 9 is a diagram of an embodiment of a system for performing a method for determining whether a subject has an increased risk of having or developing DCM.



FIG. 10 is a diagram of an embodiment of a comparison module as described herein.



FIG. 11 is a diagram of an embodiment of an operating system and applications for a computing system as described herein.





DETAILED DESCRIPTION

Described herein are compositions and methods of determining if a subject is at increased risk or in need of treatment to prevent onset of DCM and or heart-failure, based upon the inventors' discovery of a number of mutations in the TTN gene, which are predicted to encode truncated titin proteins in the affected individuals and which cause or will eventually cause DCM and or heart failure. This discovery and further characterization of the mutations is the basis for further methods of treating DCM involving administering a variety of treatments to a subject who, without treatment has >95% chance of developing DCM and/or heart failure. Dilated cardiomyopathy (DCM) is a condition in which part of the heart becomes weakened and enlarged by dilation, resulting in inefficient functioning. Heart failure is a condition heart failure in which the heart is unable to maintain an adequate circulation of blood in the tissues of the body or to pump out the venous blood returned to it by the venous circulation.


The resulting decreased heart function can lead to effects on other organs such as the liver and lungs. DCM is the most common form of non-ischemic cardiomyopathy and causes approximately 30% of congestive heart failure cases.


Methods for diagnosing DCM are well known in the art and include, but are not limited to a magnetic resonance image (MRI) to determine whether the subject's heart is enlarged, an electrocardiogram to detect abnormal electrical activity of the heart, an echocardiogram to determine the size and shape of the heart, a radionuclide ventriculogram to examine heart function, or cardiac catheterization of dyes coupled with the use of x-ray examination to assess cardiac structure and function. A diagnosis of DCM can be made when dilation and increased sphericity of, typically, at least the left ventricle and reduced systolic function are observed. Subjects with DCM can be asymptomatic or can exhibit one or more symptoms associated with DCM, which include, but are not limited to shortness of breath, dyspnoea, fatigue, swelling of the ankles and legs, muscle weakness, dystrophy, syncope, arrhythymia, thromboembolism, and/or congestive heart failure. The diagnosis of DCM is reviewed in more detail, for example, in Taylor et al. Orphanet J Rare Dis 2006 1:27; which is incorporated by reference herein in its entirety. In some embodiments, DCM can be idiopathic.


As used herein, a “subject” means a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters. Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon. Patient or subject includes any subset of the foregoing, e.g., all of the above, but excluding one or more groups or species such as humans, primates or rodents. In certain embodiments, the subject is a mammal, e.g., a primate, e.g., a human. The terms, “patient”, “individual” and “subject” are used interchangeably herein. Preferably, the subject is a mammal. The mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples. Mammals other than humans can be advantageously used, for example, as subjects that represent animal models of, for example, DCM. In addition, the methods described herein can be used to treat domesticated animals and/or pets. A subject can be male or female.


A subject can be one who has been previously diagnosed with or identified as suffering from or having DCM or one or more complications related to DCM, and optionally, but need not have already undergone treatment for DCM or the one or more complications related to DCM. A subject can be one who has not been previously diagnosed as having DCM or one or more complications related to DCM. For example, a subject can be one who exhibits one or more risk factors for DCM or one or more complications related to DCM or a subject who does not exhibit risk factors or a subject who is a family member of an individual who has been diagnosed with a cardiac condition.


A subject at risk of having or developing DCM can be identified by any means known in the art, including the present methods described herein. Thus, for example, a subject at risk can be identified by a family history of DCM, signs or symptoms of DCM, or by having one or more risk factors for DCM. Risk factors for DCM include, but are not limited to, prior instances of myocardial infarction, infection with coxsacki B virus or enteroviruses, autoimmune disease, alcohol abuse, chemotherapy (particularly doxorubicin and cobalt), thyroid disease, tachycardia, stimulant use, extrasystole, and pregnancy. A subject in need of treatment for DCM can be a subject having or diagnosed as having DCM, a subject at risk for developing DCM, or a subject displaying signs and/or symptoms of DCM.


In some embodiments, a subject in need of treatment for DCM or at risk of developing DCM can be asymptomatic.


Accordingly, provided herein is a method of treating a subject for dilated cardiomyopathy and/or heart failure. The method comprises selecting a subject at risk for developing DCM or in need of treatment for DCM or pre-DCM using an assay described herein and administering a treatment for DCM to the subject.


In some embodiments, the methods and compositions described herein relate to determining if a subject is at increased risk of having or developing DCM. A subject is at increased risk if they are at least 10% more likely, to develop DCM, a symptom of DCM, or a condition associated with DCM as compared to the average risk or a the risk for a reference of developing DCM, a symptom of DCM, or a condition associated with DCM. The average risk of developing DCM, a symptom of DCM, or a condition associated with DCM can be the likelihood of a subject in a population which does not display risk factors for DCM (e.g. family history of DCM) of developing DCM a symptom of DCM, or a condition associated with DCM. In some embodiments, the reference can be a normal healthy subject with no genetic susceptibility for DCM. For example, a normal healthy subject is not a carrier of any of the TTN mutations described herein or is not diagnosed with any forms DCM or cardiomyopathy. The reference can be also a control sample, a pooled sample of control individuals or a numeric value or range of values based on the same.


In one aspect, the methods and compositions described herein relate to determining the presence of a TTN nucleic acid mutation in a sample obtained from a subject. TTN (NCBI Gene ID: 7273) is a gene encoding the large protein titin, which contributes to the elasticity of muscle tissue. In some embodiments, the methods and compositions described herein relate to determining the presence of a TTN nucleic acid mutation, which results in a truncated (shortened) TITIN polypeptide, in a sample obtained from a subject. Without limitations, sequencing of the nucleic acid sample can be carried out using any nucleic acid sequencing known to one of ordinary skill in the art. Presence of such a mutation can indicate an increased risk of having or developing DCM. TTN nucleic acid mutations, which result in a truncated TITIN polypeptide are also referred to as DCM-risk associated TTN mutations herein.


In some embodiments, the mutation results in a titin polypeptide lacking or missing part of the pro-band or the A-band region, i.e., the truncated TITIN polypeptide lacks part of the A-band, having a wild-type sequence set forth in SEQ ID NO: 725


In some embodiments, the sample can be obtained from a fetus using methods known in the art. This can be helpful in prenetal diagnosis of DCM when a family member has been diagnosed or is at risk of developing DCM or one or more complications related to DCM, or a cardiac condition.


In some embodiments, the presence of one or more of the TTN mutations described herein can indicate an increased risk of having or developing DCM. In some embodiments, the DCM-risk associated TTN mutation can be any of the following mutations: 62476247delG, 12745C>T, 1447014471insCACACTCCATA (SEQ ID NO: 722), 1918319183delG, 2379823810delGTCAAGATATCTG (SEQ ID NO: 723), 3862138622insA, 4433644336delA, 4532245322delT, 49077G>A, 51883C>T, 52408C>T, 5314553146insG, 53347G>T, 5393553935delC, 56367T>A, 56572C>T, 56953C>T, 58678C>T, 59530C>T, 6104661046delC, 6586765867delA, 6705767063de1GCATATGinsTA, 6774567745delT, 7217872179insT, 7272372739delinsAGA, 77065C>T, 79896G>A, 80845C>T, 81046A>T, 81440G>A, 8153681537delCT, 82701C>A, 8497784980de1ATTA, 87953G>A, 88242C>T, 88528G>T, 8917789181de1AAATT, 90241C>T, 9104291042delA, 9153791538insA, 94111A>T, 95522C>A, 30476-1G>A, 34186+1G>T, 35635G>C, 35635+1G>A, 44725+2delT, 48364+1G>T, 50346+3A>G, 54422-5T>A, 54704-1G>A, 55003+1G>A, 62425+5G>A, 63405A>G, 64489+1G>A, 81898+2T>A, 92569+1G>C relative to the wild-type TTN sequence of TTN (i.e. SEQ ID NO: 1, UniProt Q8WZ42.nt). Mutations are are annotated using Human Genome Variation Society guidelines: available on the world wide web at www.hgvs.org/mutnomen.


In some embodiments, a subject at risk for having or developing DCM can have one of the DCM-risk associated TTN mutations. In some embodiments, a subject at risk for having or developing DCM can have two or more of the DCM-risk associated TTN mutations, e.g. two of the DCM-risk associated TTN mutations. In some embodiments, a subject at risk for having or developing DCM can have one or more of the DCM-risk associated TTN mutations and one or more other mutations known to be associated with a risk for DCM. Other mutations known to be associated with a risk for DCM can be mutations relative to the wild-type sequence of DCM or mutations in other DCM-risk associated genes.


In some embodiments, the methods and compositions described herein relate to determining if any of the TTN mutations described herein (e.g. DCM-risk associated TTN mutations) is present in a nucleic acid sample obtained from a subject. Methods of determining the presence of a mutation in a nucleic acid are known to one of ordinary skill in the art. Examples include, but are not limited to, contacting the sample with a probe capable of detecting at least one of the TTN mutations, sequencing the nucleic acid present in the sample. In some embodiments, the nucleic acid can be transformed into one or more detectable targets before determining the presence of a DCM-risk associated TTN mutation in the nucleic acid sample.


As used herein, the term “probe” means any molecule or reagent that can aid in the detection of a mutation in a nucleic acid. As such, a probe is not limited to an oligonucleotide that hybridizes with the nucleic acid of interest. As used herein, the term probe also includes reagents used in new generation nucleic acid segueing technologies. Further, when the probe is a nucleic acid, e.g., an oligonucleotide, it is not necessary that the probe hybridize to a location that includes the mutation site. Thus, when the probe is a nucleic acid, it can hybridize upstream (5′) or downsteam (3′) of the mutation site or a region that incudes the mutation site.


In some embodiments, the probe comprises, at its 3′-terminus, a nucleic acid sequence selected from the group consisting of SEQ ID NO: 4-332 and 342-670.


In some embodiments, the probe is a sequencing primer. As used herein, the term “sequencing primer” refers to an oligonucleotide primer that is used to initiate a sequencing reaction performed on a nucleic acid. The term “sequencing primer” refers to both a forward sequencing primer and to a reverse sequencing primer.


Those skilled in the art will readily recognize that nucleic acid molecules can be double-stranded molecules and that reference to a particular site on one strand refers, as well, to the corresponding site on a complementary strand. In defining a mutation, mutation position, or nucleotide sequence, reference to an adenine, a thymine (uridine), a cytosine, or a guanine at a particular site on one strand of a nucleic acid molecule also defines the thymine (uridine), adenine, guanine, or cytosine (respectively) at the corresponding site on a complementary strand of the nucleic acid molecule. Thus, reference can be made to either strand in order to refer to a particular position, mutation, or nucleotide sequence. Probes and primers can be designed to hybridize to either strand and genotyping methods disclosed herein can generally target either strand. Accordingly, the claims are intended to cover analysis of the opposite strand as well. One of skill in the art can readily determine the reverse complement nucleic acid sequence of the TTN mutations and wild-type sequences described herein.


Identification method of mutations can be of either a positive-type (inclusion of an allele) or a negative-type (exclusion of an allele). Positive-type methods determine the identity of a nucleotide contained in a polymorphic site, whereas negative-type methods determine the identity of a nucleotide not present in a polymorphic site. Thus, a wild-type site can be identified either as wild-type or not mutant. For example, at a biallelic polymorphic site where the wild-type allele contains a cytosine and the mutant allele contains adenine, a site can be positively determined to be either adenine or cytosine or negatively determined to be not adenine (and thus cytosine) or not cytosine (and thus adenine).


One aspect of the invention provides a method for determining an increased risk for developing DCM in a subject, by identifying in a biological sample of the subject the TTN muations described herein. The method comprises (a) contacting a nucleic acid sample obtained from the subject with a probe, wherein the probe is capable of detecting one or more of the DCM-risk associated TTN nucleic acid mutations described above herein; and (b) detecting presence or absence of at least one mutation in the TTN nucleic acid wherein the mutations in the TTN nucleic acid is determined based upon the wild type TTN gene sequence of SEQ ID NO: 1. Detection of the presence of at least one DCM-risk associated TTN mutation is indicative of the subject having an increased risk for having or developing DCM.


In one embodiment, contacting the nucleic acid in the sample involves an allelic discrimination method. In one embodiment, the allelic discrimination method involves use of a first oligonucleotide probe, which anneals with a target portion of the individual's genome. As an illustrative example only, the target portion comprises, for example, the 12745C>T mutation described herein. Because the nucleotide residue at this position differs, for example at the position in the C-allele and the T-allele, the first probe is completely complementary to only one of the two alleles. In some embodiments, a second oligonucleotide probe can also be used which is completely complementary to the target portion of the other of the two alleles. The allelic discrimination method can also involves use of at least one, and preferably a pair of amplification primers for amplifying a reference region, for example, at least a portion of the flanking region including the 12745C>T mutation locus.


The probe in some embodiments is a DNA oligonucleotide having a length in the range from about 20 to about 40 nucleotide residues, preferably from about 20 to about 30 nucleotide residues, and more preferably having a length of about 25 nucleotide residues. In one embodiment, the probe is rendered incapable of extension by a PCR-catalyzing enzyme such as Taq polymerase, for example by having a fluorescent probe attached at one or both ends thereof. Although non-labeled oligonucleotide probes can be used in the kits and methods of the invention, the probes are preferably detectably labeled. Exemplary labels include radionuclides, light-absorbing chemical moieties (e.g. dyes), fluorescent moieties, and the like. Preferably, the label is a fluorescent moiety, such as 6-carboxyfluorescein (FAM), 6-carboxy-4,7,2′,7′-tetrachlorofluoroscein (TET), rhodamine, JOE (2,7-dimethoxy-4,5-dichloro-6-carboxyfluorescein), HEX (hexachloro-6-carboxyfluorescein), or VIC.


In some embodiments, the probe can comprise both a fluorescent label and a fluorescence-quenching moiety such as 6-carboxy-N,N,N′,N′-tetramethylrhodamine (TAMRA), or 4-(4′-dimethlyaminophenylazo)benzoic acid (DABCYL). When the fluorescent label and the fluorescence-quenching moiety are attached to the same oligonucleotide and separated by no more than about 40 nucleotide residues, and preferably by no more than about 30 nucleotide residues, the fluorescent intensity of the fluorescent label is diminished. When one or both of the fluorescent label and the fluorescence-quenching moiety are separated from the oligonucleotide, the intensity of the fluorescent label is no longer diminished. In some embodiments, the probe of the present invention has a fluorescent label attached at or near (i.e. within about 10 nucleotide residues of) one end of the probe and a fluorescence-quenching moiety attached at or near the other end. Degradation of the probe by a PCR-catalyzing enzyme releases at least one of the fluorescent label and the fluorescence-quenching moiety from the probe, thereby discontinuing fluorescence quenching and increasing the detectable intensity of the fluorescent labels. Thus, cleavage of the probe (which, as discussed above, is correlated with complete complementarity of the probe with the target portion) can be detected as an increase in fluorescence of the assay mixture.


If different detectable labels are used, more than one labeled probe can be used, and therefore polymorphisms can be performed in multiplex. For example, the assay mixture can contain a first probe which is completely complementary to the target portion of, for example, the 12745C>T mutation and to which a first label is attached, and a second probe which is completely complementary to the target portion of the 51883C>T DCM risk associated TTN mutation. When two probes are used, the probes are detectably different from each other, having, for example, detectably different size, absorbance, excitation, or emission spectra, radiative emission properties, or the like. For example, a first probe can be completely complementary to the target portion of the polymorphism and have FAM and TAMRA attached at or near opposite ends thereof. The first probe can be used in the method of the present invention together with a second probe which is completely complementary to the target portion of another DCM risk associated TTN mutation and has TET and TAMRA attached at or near opposite ends thereof. Fluorescent enhancement of FAM (i.e. effected by cessation of fluorescence quenching upon degradation of the first probe by Taq polymerase) can be detected at one wavelength (e.g. 518 nanometers), and fluorescent enhancement of TET (i.e. effected by cessation of fluorescence quenching upon degradation of the second probe by Taq polymerase) can be detected at a different wavelength (e.g. 582 nanometers). Using multiplexing methods, more than one mutation described herein can be detected, providing a better diagnosis and more reliable prediction of DCM risk in a subject.


Another allelic discrimination method suitable for use in detection of TTN mutations employs “molecular beacons”. Detailed description of this methodology can be found in Kostrikis et al., Science 1998; 279:1228-1229, which is incorporated herein by reference.


The use of microarrays comprising a multiplicity of sequences, e.g., mutations described herein is becoming increasingly common in the art. Accordingly, a microarray having at least one oligonucleotide probe, as described above, appended thereon, can be used for detecting the presence or absence of a DCM-risk associated TTN mutation.


The polymorphisms of the present invention can be detected directly or indirectly using any of a variety of suitable methods including fluorescent polarization, mass spectroscopy, and the like. Suitable methods comprise direct or indirect sequencing methods, restriction site analysis, hybridization methods, nucleic acid amplification methods, gel migration methods, the use of antibodies that are specific for the proteins encoded by the different alleles of the polymorphism, or by other suitable means. Alternatively, many such methods are well known in the art and are described, for example in T. Maniatis et al., Molecular Cloning, a Laboratory Manual, 2nd Edition, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (1989), J. W. Zyskind et al., Recombinant DNA Laboratory Manual, Academic Press, Inc., New York (1988), and in R. Elles, Molecular Diagnosis of Genetic Diseases, Humana Press, Totowa, N.J. (1996), and Mamotte et al, 2006, Clin Biochem Rev, 27; 63-75) each herein incorporated by reference.


According to the present invention, any approach that detects mutations in a gene can be used, including but not limited to single-strand conformational polymorphism (SSCP) analysis (Orita et al. (1989) Proc. Natl. Acad. Sci. USA 86:2766-2770), heteroduplex analysis (Prior et al. (1995) Hum. Mutat. 5:263-268), oligonucleotide ligation (Nickerson et al. (1990) Proc. Natl. Acad. Sci. USA 87:8923-8927) and hybridization assays (Conner et al. (1983) Proc. Natl. Acad. Sci. USA 80:278-282) and DNA sequence analysis. Traditional Taq polymerase PCR-based strategies, such as PCR-RFLP, allele-specific amplification (ASA) (Ruano and Kidd (1989) Nucleic Acids Res. 17:8392), single-molecule dilution (SMD) (Ruano et al. (1990) Proc. Natl. Acad. Sci. USA 87:6296-6300), and coupled amplification and sequencing (CAS) (Ruano and Kidd (1991) Nucleic Acids Res. 19:6877-6882), are easily performed and highly sensitive methods to determine haplotypes of the present invention (Michalatos-Beloin et al. (1996) Nucleic Acids Res. 24:4841-4843; Barnes (1994) Proc. Natl. Acad. Sci. USA 91:5695-5699; Ruano and Kidd (1991) Nucleic Acids Res. 19:6877-6882).


In some embodiments, the nucleic acid sequences of the gene's allelic variants, or portions thereof, can be the basis for probes or primers, e.g., in methods for determining the identity of the allelic variant of the polymorphic region. Thus, in one embodiment, nucleic acid probes or primers can be used in the methods of the present invention to determine whether a subject is at risk of developing disease such as DCM. One of skill in the art can readily access the nucleic acid sequences flanking or spanning the TTN mutations SNPs described herein by examining SEQ ID NO:1. Thus, a skilled artisan can readily design and optimize primers or probes based on the flanking sequences of the TTN mutations described herein.


One aspect of the invention provides a method for determining an increased risk for developing DCM in a subject, by identifying in a biological sample of the subject the TTN muations described herein. In some embodiments, the method comprises detecting presence or absence of at least one mutation in the TTN nucleic acid wherein the mutations in the TTN nucleic acid is determined based upon the wild type TTN gene sequence of SEQ ID NO:1, and wherein the mutation is selected from the DCM-risk associated TTN mutations described above herein. Detection of the presence of at least one DCM-risk associated TTN mutation is indicative of the subject having an increased risk for having or developing DCM.


In some embodiments, the method comprises: (a) transforming a portion of TTN nucleic acid in sample obtained from the subject into at least one detectable target; and (b) detecting presence or absence of at least one mutation in the TTN nucleic acid wherein the mutations in the TTN nucleic acid is determined based upon the wild type TTN gene sequence of SEQ ID NO:1, and wherein the mutation is selected from the DCM-risk associated TTN mutations described above herein. Detection of the presence of at least one DCM-risk associated TTN mutation is indicative of the subject having an increased risk for having or developing DCM.


As used herein, the term “transforming” or “transformation” refers to changing an object or a substance, e.g., biological sample, nucleic acid or protein, into a substance, which can be differentiated from the first substance. The transformation can be physical, biological or chemical. Exemplary physical transformation includes, but not limited to, pre-treatment of a biological sample, e.g., from whole blood to blood serum by differential centrifugation. A biological/chemical transformation can involve at least one enzyme and/or a chemical reagent in a reaction. For example, a DNA sample can be digested into fragments by one or more restriction enzyme, or an exogenous molecule can be attached to a fragmented DNA sample with a ligase. In some embodiments, a DNA sample can undergo enzymatic replication, e.g., by polymerase chain reaction (PCR).


In some embodiments, restriction enzymes can be utilized to identify variances or a polymorphic site using “restriction fragment length polymorphism” (RFLP) analysis (Lentes et al., Nucleic Acids Res. 16:2359 (1988); and C. K. McQuitty et al., Hum. Genet. 93:225 (1994)). In RFLP, at least one target polynucleotide is digested with at least one restriction enzyme and the resulting restriction fragments are separated based on mobility in a gel. Typically, smaller fragments migrate faster than larger fragments. Consequently, a target polynucleotide that contains a particular restriction enzyme recognition site will be digested into two or more smaller fragments, which will migrate faster than a larger fragment lacking the restriction enzyme site. Knowledge of the nucleotide sequence of the target polynucleotide, the nature of the polymorphic site, and knowledge of restriction enzyme recognition sequences guide the design of such assays. In another embodiment of the present invention, restriction site analysis of particular nucleotide sequence to identify a nucleotide at a polymorphic site is determined by the presence or absence of a restriction enzyme site. A large number of restriction enzymes are known in the art and, taken together, they are capable of recognizing at least one allele of many polymorphisms.


A number of approaches use DNA ligase, an enzyme that can join two adjacent oligonucleotides hybridized to a DNA template. In Oligonucleotide ligaton assay (OLA) the sequence surrounding the mutation site is first amplified and one strand serves as a template for three ligation probes, two of these are ASO (allele-specific oligonucleotides) and a third common probe. Numerous approaches cane be used for the detection of the ligated products, for example the ASOs with differentially labeled with fluorescent of hapten labels and ligated products detected by fluorogenic of colorimetric enzyme-linked immunosorbent assays (To be et al, Nuclic Acid Res, 1996; 24; 3728-32). For electrophorosis-based systems, use of a morbidity modifier taqgs or variation in probe length coupled with floursecence detection enables the multiplex genotyping of several single nucleotide substitutions in a single tube (Baron et al, 1997; Clinical Chem., 43; 1984-6). When used on arrays, ASOs can be spotted at specific locations or addresses on a chip, PCR amplified DNA can then be added and ligation to labeled oligonucleotides at specific addresses on the array measured (Thong et al, Proc Natl Acad Sci 2003; 100; 11559-64).


Allele-specific amplification is also known as amplification refectory mutation system (ARMS) uses allele specific oligonucleotides (ASO)PCR primers and is an well established and known PCR based method for genotyping (Newton et al, J Med Genet, 1991; 28; 248-51). Typically, one of the two oligonucleotide primers used for the PCR binds to the mutation site, and amplification only takes place if the nucleotide of the mutation is present, with a mismatch being refractory to amplification. The resulting PCR Products can be analyzed by any means known to persons skilled in the art. In a variation of the approach, termed mutagenically separated PCR (MS-PCR) the two ARMS primer of different lengths, one specific for the normal gene and one for the mutation are used, to yield PCR procures of different lengths for the normal and mutant alleles (Rust et al, Nucl Acids Res, 1993; 21; 3623-9). Subsequent gel electrophoresis, for example will show at least one of the two allelic products, with normal, mutant or both (heterozygote) genes. A further variation of this forms the basis of the Masscode System™ (www.bioserve.com) which uses small molecular weight tags covalently attached through a photo-cleavable linker to the ARMS primers, with each ARMS primers labeled with a tag of differing weight (Kokoris et al, 2000, 5; 329-40). A catalogue of numerous tags allows simultaneous amplification/genotyping (multiplexing) of 24 different targets in a single PCR reaction. For any one mutation, genotyping is based on comparison of the relative abundance of the two relevant mass tags by mass spectrometry.


Normal or mutant alleles can be genotyped by measuring the binding of allele-specific oligonucleotides (ASO) hybridization probes. In such embodiments, two ASO probes, one complementary to the normal allele and the other to the mutant allele are hybridized to PCR-amplified DNA spanning the mutation site. In some embodiments, the amplified products can be immobilized on a solid surface and hybridization to radiolabelled oligonucleotides such as known as a ‘dot-blot’ assay. In alternative embodiments, the binding of the PCR products containing a quantifiable label (e.g. biotin or fluorescent labels) to a solid phase allele-specific oligonucleotide can be measured. Alternatively, for a reverse hybridixation assay, or “reverse dot-blot” the binding of PCR products containing a quantifiable label (for example but not limited to biotin or fluorescent labels) to a solid phase allele-specific oligonucleotide can be measured. In some embodiments, the use of microarrays comprising hundreds of ASO immobilized onto a solid support surfaces to form an array of ASO can also be used for large scale genotyping of multiple single polymorphisms simultaneously, for example Affymetrix GENECHIP® Mapping 10K Array, which can easily be performed by persons skilled in the art.


Homogenous assays, also called “closed tube” arrays, genomic DNA and all the reagents required for the amplification and genotyping are added simultaneously. Genotyping can be achieved without any post-amplification processing. In some embodiments, one such homogenous assay is the 5′ fluorogenic nuclease assay, also known as the TAQMAN® Assay (Livak et al, Genet Anal, 1999; 14:143-9) and in alternative embodiments Melting curve analyses of FRET probes are used. Such methods are carried out using “real-time” theromcyclers, and utilize two dual-labeled ASO hybridization probes complementary to normal and mutant alleles, where the two probes have different reported labels but a common quencher dye. In such embodiments, the changes in fluorescence characteristics of the probes upon binding to PCR products of target genes during amplification enables “real-time” monitoring of PCR amplification and differences in affinity of the fluorogenic probes for the PCR products of normal and mutant genes enables differentiation of genotypes. The approach uses two dual-labeled ASO hybridization probes complementary to the mutant and normal alleles. The two probes have different fluorescent reported dyes but a common quencher dye. When intact, the probes do not fluoresces due to the proximity of the reporter and quencher dyes. During annealing phase of PCR, two probes compete for hybridization to their target sequences, downstream of the primer sites and are subsequently cleaved by 5′ nuclease activity of Thermophilis aquaticus (Taq) polymerase as the primer is extended, resulting in the separation of the reporter dyes from the quencher. Genotyping is determined by measurement of the fluorescent intensity of the two reporter dyes after PCR amplification. Thus, when intact the probes do not fluoresce due to the proximity of the quencher dyes, whereas during the annealing phase of the PCR the probes compete for hybridization of the target sequences and the separation of one of the probes from the quencher which can be detected.


Melting-curve analysis of FRET hybridization is another approach useful in the method of the invention. Briefly, the reaction includes two oligonucleotide probes which when in close proximity forms a fluorescent complex, where one probe often termed the “mutant sensor” probe is designed to specifically hybridize across the mutation site and the other probe (often referred to as the “anchor probe”) hybridizes to an adjacent site. Fluorescent light is emitted by the “donor” excites the “acceptor” fluorphore creasing a unique fluorogenic complex, which only forms when the probes bind to adjacent sites on the amplified DNA. The “sensor” probe is complementary to either the normal or the mutant allele. Once PCR is complete, heating of the sample through the melting temperatures of the probe yields a fluorescent temperature curve which differs for the mutant and normal allele.


A variation of the FRET hybridization method is the LCGREEN™ method, which obviates the requirement for fluorescent labeled probes altogether. LCGREEN™ is a sensitive highly fluorogenic double-stranded DNA (dsDNA) binding dye that is used to detect the dissociation of unlabelled probes (Liew et al, Clin Chem, 2004; 50; 1156-64 and Zhou et al, Clin Chem, 2005; 51; 1761-2). The method uses unlabeled allele-specific oligonucleotides probes that are perfectly complementary either to the mutant or normal allele, and the mismatch of the ASO/template double strand DNA complex results in a lower melting temperature and an earlier reduction in fluorescent signal form the dsDNA binding dye with increasing temperature.


The OLA can also be used for FRET Probes (Chen et al, 1998; 8:549-56), for example, the PCR/ligation mixture can contain PCR primers, DNA polymerase without 5′ nuclease activity, thermal stable DNA ligase and oligonucleotides for the ligation reaction. The ligation of the allele-specific oligonucleotides have a different acceptor fluorophore and the third ligation oligonucleotide, which binds adjacently to the ASO has a donor fluorophore, and the three ligation oligonucleotides are designed to have a lower melting temperature for the PCR primers to prevent their interference in the PCR amplification. Following PCR, the temperature is lowered to allow ligation to proceed, which results in FRET between the donor and acceptor dyes, and alleles can be disconcerted by comparing the fluorescence emission of the two dyes.


The OLA can also be performed by the use of FRET probes (Chen et al, Genome Res, 1998; 8: 549-56). In such an embodiment, the PCR/ligation mix contains PCR primers, a thermostable DNA polymerase without 5′ exonuclease activity (to prevent the cleavage of ligation probes during the ligation phase), a thermostable DNA ligase as well as the oligonucleotides for the ligation reaction. The ligation of the ASO each have a different acceptor fluorophore and the third ligation oligonucleotide which binds adjacently to the ASO has a donor fluorophore. The three ligation oligonucleotides are designed to habe a lower melting temperature than the annealing temperature for the PCR primers prevent their interference in PCR amplification. Following PCR, the temperature is lowered to allow ligation to proceed. Ligation results in FRET between donor and acceptor dyes, and alleles can be discerned by comparing the fluorescence emission of the two dyes.


Further, variations of the homogenous PCR- and hybridization based techniques to detect polymorphisms are also encompassed in the present invention. For example, the use of Molecular Beacons (Tyagi et al, Nat Biotech 1998; 16; 49-53) and SCORPION® Probes (Thelwell et al, Nucleic Acid Res 2000; 28; 3752-61). Molecular Beacons are comprised of oligonucleotides that have fluorescent reporter and dyes at their 5′ and 3′ ends, with the central portion of the oligonucleotide hybridizing across the target sequence, but the 5′ and 3′ flanking regions are complementary to each other. When not hybridized to their target sequence, the 5′ and 3′ flanking regions hybridize to form a stem-loop structure, and there is little fluorescence because of the proximity of the reported and the quencher dyes. However, upon hybridization to their target sequence, the dyes are separated and there is a large increase in the fluorescence. Mismatched probe-target hybrids dissociate at substantially lower temperatures than exactly matched complementary hybrids. There are a number of variations of the “molecular Beacon” approach. In some embodiments, such a variation includes use of SCORPION® Probes which are similar but incorporate a PCR primer sequence as part of the probe (Thelwell et al, Nucleic Acid Res 2000; 28; 3752-61). In another variation, ‘duplex’ format gives a better fluorescent signal (Solinas et al, Nucleic Acid Res, 2001, 29; E96).


In another embodiment, polymorphisms can be detected by genotyping using a homogenous or real-time analysis on whole blood samples, without the need for DNA extraction or real-time PCR. Such a method is compatible with FRET and TAQMAN® (Castley et al, Clin Chem, 2005; 51; 2025-30) enabling extremely rapid screening for the particular polymorphism of interest.


In FP, the degree to which the emitted light remains polarized in a particular plane is proportional to the speed at which the molecules rotate and tumble in solution. Under constand pressure, temperature and viscosity, FP is directly related to the molecular weight of a fluorescent species. Therefore, when a small fluorescent molecule is incorporated into a larger molecule, there is an increase in FR FP can be used in for genotyping of polymorphisms of interest (Chen et al, Genome Res, 1999; 9: 492-8 and Latif et al, Genome Res, 2001; 11; 436-40). FP can be utilized in 5′ nuclease assay (as described above), where the oligonucleotide probe is digested to a lower molecule weight species, for example is amenable to analysis by FP, but with the added benefit of not requiring a quencher. For example, Perlkin-Elmers AcycloPrime™-FP SNP Detection Kit can be used as a FP minisequencing method. Following PCR amplification, unincorporated primers and nucleotides are degraded enzymatially, the enzymes heat inactivated and a miniseqencing reaction using DNA polymerase and fluorescent-labelled dideoxynucleotides performed. FP is then measured, typically in a 96- to 386-well plate format on a FP-plate reader.


One aspect of the invention provides a method for determining an increased risk for developing DCM in a subject, by identifying in a biological sample of the subject the TTN muations described herein. The method comprises (a) sequencing at least a portion of a TTN nucleic acid in a sample obtained from the subject; (b) comparing the sequence obtained in step (a) with the wildtype TTN sequence of SEQ ID NO: 1 to determine the presence or absence of a TTN mutation; and wherein the mutation is selected from the DCM-risk associated TTN mutations described above herein. Detection of the presence of at least one DCM-risk associated TTN mutation is indicative of the subject having an increased risk for having or developing DCM.


Single base-extension or minisequencing involves annealing an oligonucleotide primer to the single strand of a PCR product and the addition of a single dideoxynucleotide by thermal DNA polymerase. The oligonucleotide is designed to be one base short of the mutation site. The dideoxynucleotide incorporated is complementary to the base at the mutation site. Approaches cans uses different fluorescent tags or haptens for each of the four different dideoxynucleotides (Pastinen et al, Clin Chem 1996, 42; 1391-7). The dideoxynucleotide differ in molecular weight and this is the basis for single-base extension methods utilizing mass-spectrometry, and genotyping based on the mass of the extended oligonucleotide primer, can be used, for example matrix-assisted laser adsorption/ionization time-of flight mass spectrometry or MALDI-TOF (Li et al, Electrophorosis, 1999, 20; 1258-65), which is quantitative and can be used to calculate the relative allele abundance making the approach suitable for other applications such as gene dosage studies (for example for estimation of allele frequencies on pooled DNA samples).


Minisequencing or Microsequencing by MALDI-TOF can be performed by means known by persons skilled in the art. In a variation of the MALDI-TOF technique, some embodiments can use the Sequenom's Mass Array Technology (www.sequenom.com) (Sauser et al, Nucleic Acid Res, 2000, 28;E13 and Sauser et al, Nucleic Acid Res 2000, 28: E100). and also the GOOD Assay (Sauer S et al, Nucleic Acid Res, 2000; 28, E13 and Sauer et al, Nucleic Acid Res, 2000; 28:E100).


In some embodiments, variations of MALDI-TOF can be performed for analysis of variances in the genes associated with mutations described herein. For example, MALDI and electrospray ioinization (ESI) (Sauer S. Clin Chem Acta, 2006; 363; 93-105) is also useful with the methods of the present invention.


In some embodiments, the primer extension reaction and analysis is performed using PYROSEQUENCING™ (Uppsala, Sweden) which essentially is sequencing by synthesis. A sequencing primer, designed directly next to the nucleic acid differing between the disease-causing mutation and the normal allele is first hybridized to a single stranded, PCR amplified DNA template from the individual, and incubated with the enzymes, DNA polymerase, ATP sulfurylase, luciferase and apyrase, and the substrates, adenosine 5′ phosphosulfate (APS) and luciferin. One of four deoxynucleotide triphosphates (dNTP), for example, corresponding to the nucleotide present in the mutation or polymorphism, is then added to the reaction. DNA polymerase catalyzes the incorporation of the dNTP into the standard DNA strand. Each incorporation event is accompanied by release of pyrophosphate (PPi) in a quantity equimolar to the amount of incorporated nucleotide. Consequently, ATP sulfurylase converts PPi to ATP in the presence of adenosine 5′ phosphosulfate. This ATP drives the luciferase-mediated conversion of luciferin to oxyluciferin that generates visible light in amounts that are proportional to the amount of ATP. The light produced in the luciferase-catalyzed reaction is detected by a charge coupled device (CCD) camera and seen as a peak in a PYROGRAM™. Each light signal is proportional to the number of nucleotides incorporated and allows a clear determination of the presence or absence of, for example, the mutation or polymorphism. Thereafter, apyrase, a nucleotide degrading enzyme, continuously degrades unincorporated dNTPs and excess ATP. When degradation is complete, another dNTP is added which corresponds to the dNTP present in for example the selected SNP. Addition of dNTPs is performed one at a time. Deoxyadenosine alfa-thio triphosphate (dATPS) is used as a substitute for the natural deoxyadenosine triphosphate (dATP) since it is efficiently used by the DNA polymerase, but not recognized by the luciferase. For detailed information about reaction conditions for the PYROSEQUENCING, see, e.g. U.S. Pat. No. 6,210,891, which is incorporated herein by reference in its entirety.


Other techniques known to persons skilled in the art are also incorporated for use with the present invention, for example see Kwok, Hum Mut 2002; 9; 315-323 and Kwok, Annu Rev Genomic Hum Genetics, 2001; 2; 235-58 for reviews, which are incorporated herein in their entirety by reference. Examples of other techniques to detect variances and/or polymorphisms are the INVADER® Assay (Gut et al, Hum Mutat, 2001; 17:475-92, Shi et al, Clin Chem, 2001, 47,164-92, and Olivier et al, Mutat Res, 2005; 573:103-110), the method utilizing FLAP endonucleases (U.S. Pat. No. 6,706,476) and the SNPlex genoptyping systems (Tobler et al, J. Biomol Tech, 2005; 16; 398-406.


In one embodiment, a long-range PCR (LR-PCR) is used to detect mutations or polymorphisms of the present invention. LR-PCR products are genotyped for mutations or polymorphisms using any genotyping methods known to one skilled in the art, and haplotypes inferred using mathematical approaches (e.g., Clark's algorithm (Clark (1990) Mol. Biol. Evol. 7:111-122).


For example, methods including complementary DNA (cDNA) arrays (Shalon et al., Genome Research 6(7):639-45, 1996; Bernard et al., Nucleic Acids Research 24(8):1435-42, 1996), solid-phase mini-sequencing technique (U.S. Pat. No. 6,013,431, Suomalainen et al. Mol. Biotechnol. Jun; 15(2):123-31, 2000), ion-pair high-performance liquid chromatography (Doris et al. J. Chromatogr. A can 8; 806(1):47-60, 1998), and 5′ nuclease assay or real-time RT-PCR (Holland et al. Proc Natl Acad Sci USA 88: 7276-7280, 1991), or primer extension methods described in the U.S. Pat. No. 6,355,433, can be used.


Molecular beacons also contain fluorescent and quenching dyes, but FRET only occurs when the quenching dye is directly adjacent to the fluorescent dye. Molecular beacons are designed to adopt a hairpin structure while free in solution, bringing the fluorescent dye and quencher in close proximity. Therefore, for example, two different molecular beacons are designed, one recognizing the mutation or polymorphism and the other the corresponding wildtype allele. When the molecular beacons hybridize to the nucleic acids, the fluorescent dye and quencher are separated, FRET does not occur, and the fluorescent dye emits light upon irradiation. Unlike TaqMan probes, molecular beacons are designed to remain intact during the amplification reaction, and must rebind to target in every cycle for signal measurement. TaqMan probes and molecular beacons allow multiple DNA species to be measured in the same sample (multiplex PCR), since fluorescent dyes with different emission spectra can be attached to the different probes, e.g. different dyes are used in making the probes for different DCM-risk associated mutations. Multiplex PCR also allows internal controls to be co-amplified and permits allele discrimination in single-tube assays. (Ambion Inc, Austin, Tex., TechNotes 8(1)-February 2001, Real-time PCR goes prime time).


Another method to detect mutations or polymorphisms is by using fluorescence tagged dNTP/ddNTPs. In addition to use of the fluorescent label in the solid phase mini-sequencing method, a standard nucleic acid sequencing gel can be used to detect the fluorescent label incorporated into the PCR amplification product. A sequencing primer is designed to anneal next to the base differentiating the mutations and wildtype alleles. A primer extension reaction is performed using chain terminating dideoxyribonucleoside triphosphates (ddNTPs) labeled with a fluorescent dye, one label attached to the ddNTP to be added to the standard nucleic acid and another to the ddNTP to be added to the target nucleic acid.


Alternatively, an INVADER® assay can be used (Third Wave Technologies, Inc (Madison, Wis.)). This assay is generally based upon a structure-specific nuclease activity of a variety of enzymes, which are used to cleave a target-dependent cleavage structure, thereby indicating the presence of specific nucleic acid sequences or specific variations thereof in a sample (see, e.g. U.S. Pat. No. 6,458,535). For example, an INVADER® operating system (OS), provides a method for detecting and quantifying DNA and RNA. The INVADER® OS is based on a “perfect match” enzyme-substrate reaction. The INVADER® OS uses proprietary CLEAVASE® enzymes (Third Wave Technologies, Inc (Madison, Wis.)), which recognize and cut only the specific structure formed during the INVADER® process which structure differs between the different alleles selected for detection, i.e. the wildtype TTN sequence and the DCM-risk associated mutations. Unlike the PCR-based methods, the INVADER® OS relies on linear amplification of the signal generated by the INVADER® process, rather than on exponential amplification of the target.


In the INVADER® process, two short DNA probes hybridize to the target to form a structure recognized by the CLEAVASE® enzyme. The enzyme then cuts one of the probes to release a short DNA “flap.” Each released flap binds to a fluorescently-labeled probe and forms another cleavage structure. When the CLEAVASE® enzyme cuts the labeled probe, the probe emits a detectable fluorescence signal.


Mutations or polymophisms can also be detected using allele-specific hybridization followed by a MALDI-TOF-MS detection of the different hybridization products. In the preferred embodiment, the detection of the enhanced or amplified nucleic acids representing the different alleles is performed using matrix-assisted laser desorption ionization/time-of-flight (MALDI-TOF) mass spectrometric (MS) analysis described in the Examples below. This method differentiates the alleles based on their different mass and can be applied to analyze the products from the various above-described primer-extension methods or the INVADER® process.


In one embodiment, a haplotyping method can be used for the purpose of the invention. A halotyping method is a physical separation of alleles by cloning, followed by sequencing. Other methods of haplotyping include, but are not limited to monoallelic mutation analysis (MAMA) (Papadopoulos et al. (1995) Nature Genet. 11:99-102) and carbon nanotube probes (Woolley et al. (2000) Nature Biotech. 18:760-763). U.S. Patent Application No. US 2002/0081598 also discloses a useful haplotying method which involves the use of PCR amplification.


Computational algorithms such as expectation-maximization (EM), subtraction and PHASE are useful methods for statistical estimation of haplotypes (see, e.g., Clark, A.G. Inference of haplotypes from PCR-amplified samples of diploid populations. Mol Biol Evol 7, 111-22. (1990); Stephens, M., Smith, N.J. & Donnelly, P. A new statistical method for haplotype reconstruction from population data. Am J Hum Genet. 68, 978-89. (2001); Templeton, A. R., Sing, C. F., Kessling, A. & Humphries, S. A cladistic analysis of phenotype associations with haplotypes inferred from restriction endonuclease mapping. II. The analysis of natural populations. Genetics 120, 1145-54. (1988)).


Other methods for genetic screening can be used within the scope of the present invention, for example, to detect mutations in genomic DNA, cDNA and/or RNA samples. Methods commonly used, or newly developed or methods yet unknown are encompassed for used in the present invention. Examples of newly discovered methods include for example, but are not limited to; SNP mapping (Davis et al, Methods Mol Biology, 2006; 351; 75-92); Nanogen Nano Chip, (keen-Kim et al, 2006; Expert Rev Mol Diagnostic, 6; 287-294); Rolling circle amplification (RCA) combined with circularable oligonucleotide probes (c-probes) for the detection of nucleic acids (Zhang et al, 2006: 363; 61-70), luminex XMAP system for detecting multiple SNPs in a single reaction vessel (Dunbar S A, Clin Chim Acta, 2006; 363; 71-82; Dunbar et al, Methods Mol Med, 2005; 114:147-1471) and enzymatic mutation detection methods (Yeung et al, Biotechniques, 2005; 38; 749-758).


Methods used to detect point mutations include denaturing gradient gel electrophoresis (“DGGE”), restriction fragment length polymorphism analysis (“RFLP”), chemical or enzymatic cleavage methods, direct sequencing of target regions amplified by PCR (see above), single strand conformation polymorphism analysis (“SSCP”) and other methods well known in the art.


One method of screening for point mutations is based on RNase cleavage of base pair mismatches in RNA/DNA or RNA/RNA heteroduplexes. As used herein, the term “mismatch” is defined as a region of one or more unpaired or mispaired nucleotides in a double-stranded RNA/RNA, RNA/DNA or DNA/DNA molecule. This definition thus includes mismatches due to insertion/deletion mutations, as well as single or multiple base point mutations.


In such embodiments, protection from cleavage agents (such as a nuclease, hydroxylamine or osmium tetroxide and with piperidine) can be used to detect mismatched bases in RNA/RNA DNA/DNA, or RNA/DNA heteroduplexes (see, e.g., Myers et al. (1985) Science 230:1242). In general, the technique of “mismatch cleavage” starts by providing heteroduplexes formed by hybridizing a control nucleic acid, which is optionally labeled, e.g., RNA or DNA, comprising a nucleotide sequence of the allelic variant of the gene of interest with a sample nucleic acid, e.g., RNA or DNA, obtained from a tissue sample. The double-stranded duplexes are treated with an agent which cleaves single-stranded regions of the duplex such as duplexes formed based on basepair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with S1 nuclease to enzymatically digest the mismatched regions. In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine whether the control and sample nucleic acids have an identical nucleotide sequence or in which nucleotides they are different. See, for example, U.S. Pat. No. 6,455,249, Cotton et al. (1988) Proc. Natl. Acad. Sci. USA 85:4397; Saleeba et al. (1992) Methods Enzy. 217:286-295. In another embodiment, the control or sample nucleic acid is labeled for detection.


U.S. Pat. No. 4,946,773 describes an RNaseA mismatch cleavage assay that involves annealing single-stranded DNA or RNA test samples to an RNA probe, and subsequent treatment of the nucleic acid duplexes with RNaseA. For the detection of mismatches, the single-stranded products of the RNaseA treatment, electrophoretically separated according to size, are compared to similarly treated control duplexes. Samples containing smaller fragments (cleavage products) not seen in the control duplex are scored as positive.


Other investigators have described the use of RNaseI in mismatch assays. The use of RNaseI for mismatch detection is described in literature from Promega Biotech. Promega markets a kit containing RNaseI that is reported to cleave three out of four known mismatches.


In other embodiments, alterations in electrophoretic mobility is used to identify the particular allelic variant. For example, single strand conformation polymorphism (SSCP) can be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids (Orita et al. (1989) Proc Natl. Acad. Sol USA 86:2766; Cotton (1993) Mutat. Res. 285:125-144 and Hayashi (1992) Genet Anal Tech Appl 9:73-79). Single-stranded DNA fragments of sample and control nucleic acids are denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments can be labeled or detected with labeled probes. The sensitivity of the assay can be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In another preferred embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al. (1991) Trends Genet. 7:5).


Gel Migration Single strand conformational polymorphism (SSCP; M. Orita et al., Genomics 5:8 74-8 79 (1989); Huinphfies et al., In: Molecular Diagnosis of Genetic Diseases, R. Elles, ed. pp 321-340 (1996)) and temperature gradient gel electrophoresis (TGGE; R. M. Wartell et al., Nucl. Acids Res. 18:2699-2706 (1990)) are examples of suitable gel migration-based methods for determining the identity of a polymorphic site. In SSCP, a single strand of DNA will adopt a conformation that is uniquely dependent of its sequence composition. This conformation is usually different, if even a single base is changed. Thus, certain embodiments of the present invention, SSCP can be utilized to identify polymorphic sites, as wherein amplified products (or restriction fragments thereof of the target polynucleotide are denatured, then run on a non-denaturing gel. Alterations in the mobility of the resultant products are thus indicative of a base change. Suitable controls and knowledge of the “normal” migration patterns of the wild-type alleles can be used to identify polymorphic variants.


In yet another embodiment, the identity of the allelic variant is obtained by analyzing the movement of a nucleic acid comprising the polymorphic region in polyacrylamide gels containing a gradient of denaturant, which is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al. (1985) Nature 313:495). When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for, example by adding a GC clamp of approximately 40 bp of high-melting GC rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing agent gradient to identify differences in the mobility of control and sample DNA (Rosenbaum and Reissner (1987) Biophys Chem 265:1275).


Others have described using the MutS protein or other DNA-repair enzymes for detection of single-base mismatches. Alternative methods for detection of deletion, insertion or substitution mutations that can be used in the practice of the present invention are disclosed in U.S. Pat. Nos. 5,849,483, 5,851,770, 5,866,337, 5,925,525 and 5,928,870, each of which is incorporated herein by reference in its entirety. Several methods have been developed to screen polymorphisms and some examples are listed below. The reference of Kwok and Chen (2003) and Kwok (2001) provide overviews of some of these methods, both of these references are specifically incorporated by reference.


Examples of identifying polymorphisms and applying that information in a way that yields useful information regarding patients can be found, for example, in U.S. Pat. No. 6,472,157; U.S. Patent Application Publications 20020016293, 20030099960, 20040203034; WO 0180896, all of which are hereby incorporated by reference.


In another embodiment, multiplex PCR procedures using allele-specific primers can be used to simultaneously amplify multiple regions of a target nucleic acid (PCT Application WO89/10414), enabling amplification only if a particular allele is present in a sample. Other embodiments using alternative primer-guided nucleotide incorporation procedures for assaying polymorphic sites in DNA can be used, and have been described (Komher, J. S. et al., Nucl. Acids. Res. 17:7779-7784 (1989); Sokolov, B. P., Nucl. Acids Res. 18:3671 (1990); Syvanen, A.-C., et al., Genomics 8:684-692 (1990); Kuppuswamy, M. N. et al., Proc. Nat. Acad. Sci. (U.S.A) 88:1143-1147 (1991); Bajaj et al. (U.S. Pat. No. 5,846,710); Prezant, T. R. et al., Hum Mutat. 1: 159-164 (1992); Ugozzoli, L. et al., GATA 9:107-112 47 (1992); Nyr6n, P. et al., Anal. Biochem. 208:171-175 (1993)).


Other known nucleic acid amplification procedures include transcription-based amplification systems (Malek, L. T. et al., U.S. Pat. No. 5,130,238; Davey, C. et al., European Patent Application 329,822; Schuster et al.) U.S. Pat. No. 5,169,766; Miller, H. I. et al., PCT-Application WO89/06700; Kwoh, D. et al., Proc. NatI. Acad. Sci. (U.S.A) 86:1173 Z1989); Gingeras, T. R. et al., PCT Application WO88/10315)), or isothermal amplification methods (Walker, G. T. et al., Proc. Natl. 4cad Sci. (U.S.A) 89:392-396 (1992)) can also be used.


Another method to determine genetic variation is using “gene chips.” Probes can be affixed to surfaces for use as “gene chips.” Such gene chips can be used to detect genetic variations by a number of techniques known to one of skill in the art. In one technique, oligonucleotides are arrayed on a gene chip for determining the DNA sequence of a by the sequencing by hybridization approach, such as that outlined in U.S. Pat. Nos. 6,025,136 and 6,018,041. The probes of the present invention also can be used for fluorescent detection of a genetic sequence. Such techniques have been described, for example, in U.S. Pat. Nos. 5,968,740 and 5,858,659. A probe also can be affixed to an electrode surface for the electrochemical detection of nucleic acid sequences such as described by Kayyem et al. U.S. Pat. No. 5,952,172 and by Kelley, S. O. et al. (1999) Nucleic Acids Res. 27:4830-4837.


Any oligonucleotide-based diagnostic may be utilized to determine whether a sample includes the presence or absence of a polymorphic variant in a sample. For example, primer extension methods, ligase sequence determination methods (e.g., U.S. Pat. Nos. 5,679,524 and 5,952,174, and WO 01/27326), mismatch sequence determination methods (e.g., U.S. Pat. Nos. 5,851,770; 5,958,692; 6,110,684; and 6,183,958), microarray sequence determination methods, restriction fragment length polymorphism (RFLP), single strand conformation polymorphism detection (SSCP) (e.g., U.S. Pat. Nos. 5,891,625 and 6,013,499), PCR-based assays (e.g., TAQMAN™ PCR System (Applied Biosystems)), and nucleotide sequencing methods may be used. Oligonucleotide extension methods typically involve providing a pair of oligonucleotide primers in a polymerase chain reaction (PCR) or in other nucleic acid amplification methods for the purpose of amplifying a region from the nucleic acid sample that comprises the polymorphic variation. One oligonucleotide primer is complementary to a region 3′ or downstream of the polymorphism and the other is complementary to a region 5′ or upstream of the polymorphism. A PCR primer pair may be used in methods disclosed in U.S. Pat. Nos. 4,683,195; 4,683,202, 4,965,188; 5,656,493; 5,998,143; 6,140,054; WO 01/27327; and WO 01/27329 for example. PCR primer pairs may also be used in any commercially available machines that perform PCR, such as any of the GENEAMP™, systems available from Applied Biosystems. Also, those of ordinary skill in the art will be able to design oligonucleotide primers based upon the nucleotide sequences set forth in SEQ ID NO:1.


Determination of the presence or absence of a TTN mutation described herein can also involve the use of an extension oligonucleotide that hybridizes to the amplified fragment adjacent to the polymorphic variation. An adjacent fragment refers to the 3′ end of the extension oligonucleotide being often 1 nucleotide from the 5′ end of the polymorphic site, and sometimes 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides from the 5′ end of the polymorphic site, in the nucleic acid when the extension oligonucleotide is hybridized to the nucleic acid. The extension oligonucleotide then is extended by one or more nucleotides, and the number and/or type of nucleotides that are added to the extension oligonucleotide determine whether the polymorphic variant is present. Oligonucleotide extension methods are disclosed, for example, in U.S. Pat. Nos. 4,656,127; 4,851,331; 5,679,524; 5,834,189; 5,876,934; 5,908,755; 5,912,118; 5,976,802; 5,981,186; 6,004,744; 6,013,431; 6,017,702; 6,046,005; 6,087,095; 6,210,891; and WO 01/20039. Oligonucleotide extension methods using mass spectrometry are described, for example, in U.S. Pat. Nos. 5,547,835; 5,605,798; 5,691,141; 5,849,542; 5,869,242; 5,928,906; 6,043,031; and 6,194,144. Multiple extension oligonucleotides may be utilized in one reaction, which is referred to as multiplexing.


In some embodiments, the methods and assays described herein can comprise administering a treatment for DCM if the subject is determined to be at risk for DCM, e.g. if a sample obtained from the subject is determined to comprise at least one of the DCM-risk associated TTN mutations described herein. Methods of treating DCM are known to one of skill in the art and include, but are not limited to the use of angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers, beta blockers, diuretics, aldosterone antagonists, digoxin (Lanoxin), blood thinning medications, biventricular pacemakers, implantable cardioverter-defibrillators (ICDs), heart pumps (left ventricular assist devices, or LVADs), heart transplant, salt restriction, digitalis, alcohol restriction, reverse remodeling, conenzyme Q10, and any combinations thereof. A subject determined to have an increased DCM risk can further be given life-style advice, dietary advice, follow-up scheduling advice or agents that may assist in preventing or slowing down symptons or development of DCM.


In some embodiments, a subject treated according to the methods described herein is asymptomatic. In some embodiments, a subject treated according to the methods described herein has idiopathic DCM.


In some embodiments, the methods described herein relating to treating DCM in a subject can comprise administering a polypeptide comprising titin (e.g. a polypeptide comprising the sequence of SEQ ID NO:2). In some embodiments, a polypeptide comprising the sequence of SEQ ID NO:2 is administered. In some embodiments, a nucleic acid encoding the sequence of SEQ ID NO: 2 is administered.


Subjects determined to have one or more of the DCM-risk associated TTN mutations described herein can be suffering from a lack of full-length, or fully functional titin. Accordingly, in some embodiments, a subject determined to have one or more of the DCM-risk associated TTN mutations described herein can be administered a polypeptide comprising titin (e.g. a polypeptide having the sequence of SEQ ID NO:2). In some embodiments, a polypeptide comprising the sequence of SEQ ID NO:2 is administered. In some embodiments, a nucleic acid encoding the sequence of SEQ ID NO: 2 is administered.


As used herein, the terms “treatment” and “treating,” with respect to treatment of DCM, means preventing the progression of the disease, or altering the course of the disorder (for example, but not limited to, slowing the progression of the disorder), or reversing a symptom of the disorder or reducing one or more symptoms and/or one or more biochemical markers in a subject, preventing one or more symptoms from worsening or progressing, promoting recovery or improving prognosis. For example, in the case of DCM treatment, therapeutic treatment can refer to reducing the myocardial enlargement in a subject. Measurable lessening includes any statistically significant decline in a measurable marker or symptom, such as measuring the size of the subject's heart or the performance of the subject's heart after treatment.


In some embodiments, a nucleic acid encoding a polypeptide comprising the amino acid sequence of SEQ ID NO: 2 is administered to a subject. In some embodiments, the nucleic acid encoding the polypeptide of SEQ ID NO: 2 is operably linked to a vector. In some embodiments, the nucleic acid encoding the polypeptide of SEQ ID NO: 2 is a recombinant gene.


Gene therapy has the advantage of potentially long-term therapeutic benefit with only one, or perhaps a limited number, of administrations. These methods allow clinicians to introduce DNA coding for a gene of interest directly into a patient (in vivo gene therapy) or into cells isolated from a patient or a donor (ex vivo gene therapy). Therapeutic proteins produced by transduced cells after gene therapy may be maintained at a relatively constant level in the myocardial tissue of a subject, as compared to a protein that is administered directly, which will typically vary greatly in concentration between the time right after administration of a first dose and the time immediately before the succeeding dose.


Administration of gene therapy vectors can be performed by intravenous, intramuscular, intraarterial, intraventricular, intracardiac, intradermal, intraperitoneal, subcutaneous, subcuticular, and/or intraarticular administration with known techniques.


Further, regulatable genetic constructs using small molecule inducers have been developed that might be included in vectors to be used in gene therapy embodiments of the present invention. Rivera et al. (1996) Nat. Med. 2:1028-32; No et al. (1996) Proc. Natl. Acad. Sci. USA, 93:3346-51; Gossen and Bujard (1992) Proc. Natl. Acad. Sci. USA 89:5547-51; the GeneSwitch® system (Valentis, Inc., Burlingame, Calif.). These systems are based on the use of engineered transcription factors whose activity is controlled by a small molecule drug, and a transgene whose expression is driven by the regulated transcription factor. One such system, based on induction by rapamycin (referred to herein as the “dimerizer system”), involves formation of a functional transcription factor from two synthetic fusion proteins dependent upon addition of rapamycin. Rivera et al. (1996) Nat. Med. 2:1028-32; Pollock et al. (2000) Proc. Natl. Acad. Sci. USA 97:13221-26. The dimerizer system is a component of the ARGENT Transcription Technology platform of ARIAD Pharmaceuticals, Inc. (Cambridge, Mass.). See U.S. Pat. Nos. 6,043,082 and 6,649,595; Rivera et al. (1999) Proc. Natl. Acad. Sci. USA 96:8657-62.


DNA may be introduced into a patient's cells in several ways. There are transfection methods, including chemical methods such as calcium phosphate precipitation and liposome-mediated transfection, and physical methods such as electroporation. In general, transfection methods are not suitable for in vivo gene delivery. Genes can be delivered using “naked” DNA in plasmid form. There are also methods that use recombinant viruses. Current viral-mediated gene delivery methods employ retrovirus, adenovirus, herpes virus, pox virus, and adeno-associated virus (AAV) vectors. Of the more than one hundred gene therapy trials conducted, more than 95% used viral-mediated gene delivery. C. P. Hodgson, Bio/Technology 13, 222-225 (1995).


In one embodiment, the recombinant titin encoding gene is operably linked to a vector. In general, as used herein, the term “vector” refers to any genetic element, such as a plasmid, phage, transposon, cosmid, chromosome, virus, virion, etc., that is capable of replication when associated with the proper control elements and that can transfer gene sequences to cells. Thus, the term includes cloning and expression vehicles, as well as viral vectors. By “recombinant vector” is meant a vector that includes a heterologous nucleic acid sequence, or “transgene,” that is capable of expression in vivo.


In additional embodiments, it can be desirable to fuse the gene of interest to immunoglobulin molecules, for example the Fc portion of a mouse IgG2a with a noncytolytic mutation, to provide for sustained expression. Such a technique has been shown to provide for sustained expression of cytokines, especially when combined with electroporation. See e.g. Jiang et al. (2003) J. Biochem. 133:423-27; Adachi et al. (2002) Gene Ther. 9:577-83.


It should be understood that the vectors delivered by the methods of the present invention be combined with other suitable compositions and therapies for DCM.


Plasmid-Directed Gene Delivery

The recombinant titin encoding gene can be delivered using non-viral plasmid-based nucleic acid delivery systems, as described in U.S. Pat. Nos. 6,413,942, 6,214,804, 5,580,859, 5,589,466, 5,763,270 and 5,693,622, all incorporated herein by reference in their entireties. Plasmids will include the gene of interest operably linked to control elements that direct the expression of the gene in a target cell, which control elements are well known in the art. Plasmid DNA can be guided by a nuclear localization signal or like modification.


Alternatively, plasmid vectors encoding the gene of interest can be packaged in liposomes prior to delivery to a subject or to cells, as described in U.S. Pat. Nos. 5,580,859, 5,549,127, 5,264,618, 5,703,055, all incorporated herein by reference in their entireties. For a review of the use of liposomes as carriers for delivery of nucleic acids, see, Hug and Sleight (1991) Biochim. Biophys. Acta. 1097:1-17; Straubinger et al. (1983) in Methods of Enzymology Vol. 101, pp. 512-27; de Lima et al. (2003) Current Medicinal Chemistry, Volume 10(14): 1221-31. The DNA can also be delivered in cochleate lipid compositions similar to those described by Papahadjopoulos et al. (1975) Biochem. Biophys. Acta. 394:483-491. See also U.S. Pat. Nos. 4,663,161 and 4,871,488, incorporated herein by reference in their entireties. In one embodiment, the plasmid vector is complexed with Lipofectamine 2000 at a ratio of 3 μL1 of Lipofectamine per μg of DNA. Wang et al. (2005) Mol. Therapy 12(2):314-320.


Biolistic delivery systems employing particulate carriers such as gold and tungsten may also be used to deliver genes of interest. The particles are coated with the gene to be delivered and accelerated to high velocity, generally under reduced pressure, using a gun powder discharge from a “gene gun.” See, e.g., U.S. Pat. Nos. 4,945,050, 5,036,006, 5,100,792, 5,179,022, 5,371,015, and 5,478,744, all incorporated herein by reference in their entireties.


A wide variety of other methods can be used to deliver the vectors. Such methods include DEAE dextran-mediated transfection, calcium phosphate precipitation, polylysine- or polyornithine-mediated transfection, or precipitation using other insoluble inorganic salts, such as strontium phosphate, aluminum silicates including bentonite and kaolin, chromic oxide, magnesium silicate, talc, and the like. Other useful methods of transfection include electroporation, sonoporation, protoplast fusion, peptoid delivery, or microinjection. See, e.g., Sambrook et al (1989) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratories, New York, for a discussion of techniques for transforming cells of interest; and Felgner, P. L. (1990) Advanced Drug Delivery Reviews 5:163-87, for a review of delivery systems useful for gene transfer. Exemplary methods of delivering DNA using electroporation are described in U.S. Pat. Nos. 6,132,419; 6,451,002, 6,418,341, 6,233,483, U.S. Patent Publication No. 2002/0146831, and International Publication No. WO/0045823, all of which are incorporated herein by reference in their entireties.


Plasmid vectors can also be introduced directly into the heart by injection, as described herein in greater detail with regard to protein administration. Plasmid DNA can be complexed with cationic agents such as polyethyleneimine (PEI) or Lipofectamine 2000 to facilitate uptake. See, e.g., Wang et al. (2005) Mol. Therapy 12(2):314-320. In one embodiment, a plasmid vector encoding titin is complexed with PEI (25 kDa, Sigma-Aldrich, San Diego, Calif.) in a 5% glucose solution at a N/P ratio of approximately 15, where N represents PEI nitrogen and P represents DNA phosphate.


Retroviral Gene Delivery

Retroviruses provide a convenient platform for gene delivery. A selected gene can be inserted into a vector and packaged in retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to cells of the subject either in vivo or ex vivo. A number of retroviral systems have been described. See, e.g., U.S. Pat. No. 5,219,740; Miller and Rosman (1989) BioTechniques 7:980-90; Miller, A. D. (1990) Human Gene Therapy 1:5-14; Scarpa et al. (1991) Virology 180:849-52; Burns et al. (1993) Proc. Natl. Acad. Sci. USA 90:8033-37; Boris-Lawrie and Temin (1993) Curr. Opin. Genet. Develop. 3:102-09.


Replication-defective murine retroviral vectors are widely used gene transfer vectors. Murine leukemia retroviruses include a single stranded RNA molecule complexed with a nuclear core protein and polymerase (pol) enzymes, encased by a protein core (gag), and surrounded by a glycoprotein envelope (env) that determines host range. The genomic structure of retroviruses includes gag, pol, and env genes and 5′ and 3′ long terminal repeats (LTRs). Retroviral vector systems exploit the fact that a minimal vector containing the 5′ and 3′ LTRs and the packaging signal are sufficient to allow vector packaging, infection and integration into target cells, provided that the viral structural proteins are supplied in trans in the packaging cell line. Fundamental advantages of retroviral vectors for gene transfer include efficient infection and gene expression in most cell types, precise single copy vector integration into target cell chromosomal DNA and ease of manipulation of the retroviral genome.


Adenoviral Gene Delivery

In one embodiment of the subject invention, a nucleotide sequence encoding titin is inserted into an adenovirus-based expression vector Unlike retroviruses, which integrate into the host genome, adenoviruses persist extrachromosomally thus minimizing the risks associated with insertional mutagenesis (Haj-Ahmad and Graham (1986) J. Virol. 57:267-74; Bett et al. (1993) J. Virol. 67:5911-21; Mittereder et al. (1994) Human Gene Therapy 5:717-29; Seth et al. (1994) J. Virol. 68:933-40; Barr et al. (1994) Gene Therapy 1:51-58; Berkner, K. L. (1988) BioTechniques 6:616-29; and Rich et al. (1993) Human Gene Therapy 4:461-76).


The adenovirus genome is a linear double-stranded DNA molecule of approximately 36,000 base pairs with the 55-kDa terminal protein covalently bound to the 5′ terminus of each strand. Adenoviral (“Ad”) DNA contains identical Inverted Terminal Repeats (“ITRs”) of about 100 base pairs with the exact length depending on the serotype. The viral origins of replication are located within the ITRs exactly at the genome ends.


Adenoviral vectors have several advantages in gene therapy. They infect a wide variety of cells, have a broad host-range, exhibit high efficiencies of infectivity, direct expression of heterologous genes at high levels, and achieve long-term expression of those genes in vivo. The virus is fully infective as a cell-free virion so injection of producer cell lines is not necessary. With regard to safety, adenovirus is not associated with severe human pathology, and the recombinant vectors derived from the virus can be rendered replication defective by deletions in the early-region 1 (“E1”) of the viral genome. Adenovirus can also be produced in large quantities with relative ease. For all these reasons vectors derived from human adenoviruses, in which at least the E1 region has been deleted and replaced by a gene of interest, have been used extensively for gene therapy experiments in the pre-clinical and clinical phase.


Adenoviral vectors for use with the present invention can be derived from any of the various adenoviral serotypes, including, without limitation, any of the over 40 serotype strains of adenovirus, such as serotypes 2, 5, 12, 40, and 41. The adenoviral vectors used herein are replication-deficient and contain the gene of interest under the control of a suitable promoter, such as any of the promoters discussed below with reference to adeno-associated virus.


Other recombinant adenoviruses of various serotypes, and comprising different promoter systems, can be created by those skilled in the art. See, e.g., U.S. Pat. No. 6,306,652, incorporated herein by reference in its entirety.


Moreover, “minimal” adenovirus vectors as described in U.S. Pat. No. 6,306,652 will find use with the present invention. Such vectors retain at least a portion of the viral genome required for encapsidation (the encapsidation signal), as well as at least one copy of at least a functional part or a derivative of the ITR. Packaging of the minimal adenovirus vector can be achieved by co-infection with a helper virus or, alternatively, with a packaging-deficient replicating helper system.


Other useful adenovirus-based vectors for delivery of titin gene include the “gutless” (helper-dependent) adenovirus in which the vast majority of the viral genome has been removed. Wu et al. (2001) Anesthes. 94:1119-32. Such “gutless” adenoviral vectors produce essentially no viral proteins, thus allowing gene therapy to persist for over a year after a single administration. Parks (2000) Clin. Genet. 58:1-11; Tsai et al. (2000) Curr. Opin. Mol. Ther. 2:515-23. In addition, removal of the viral genome creates space that can be used to insert control sequences that provide for regulation of transgene expression by systemically administered drugs (Burcin et al. (1999) Proc. Natl. Acad. Sci. USA 96:355-60), adding both safety and control of virally driven protein expression. These and other recombinant adenoviruses will find use with the present methods.


Adeno Associated Virus (AAV) Gene Delivery

One viral system that has been used for gene delivery is AAV. AAV is a parvovirus which belongs to the genus Dependovirus. AAV has several attractive features not found in other viruses. First, AAV can infect a wide range of host cells, including non-dividing cells. Second, AAV can infect cells from different species. Third, AAV has not been associated with any human or animal disease and does not appear to alter the biological properties of the host cell upon integration. Indeed, it is estimated that 80-85% of the human population has been exposed to the virus. Finally, AAV is stable at a wide range of physical and chemical conditions, facilitating production, storage and transportation.


The AAV genome is a linear single-stranded DNA molecule containing approximately 4681 nucleotides. The AAV genome generally comprises an internal non-repeating genome flanked on each end by inverted terminal repeats (ITRs). The ITRs are approximately 145 base pairs (bp) in length. The ITRs have multiple functions, including serving as origins of DNA replication and as packaging signals for the viral genome.


The internal non-repeated portion of the genome includes two large open reading frames, known as the AAV replication (rep) and capsid (cap) genes. The rep and cap genes code for viral proteins that allow the virus to replicate and package the viral genome into a virion. In particular, a family of at least four viral proteins is expressed from the AAV rep region, Rep 78, Rep 68, Rep 52, and Rep 40, named according to their apparent molecular weight. The AAV cap region encodes at least three proteins, VP1, VP2, and VP3.


AAV is a helper-dependent virus; that is, it requires co-infection with a helper virus (e.g., adenovirus, herpesvirus or vaccinia) in order to form AAV virions in the wild. In the absence of co-infection with a helper virus, AAV establishes a latent state in which the viral genome inserts into a host cell chromosome, but infectious virions are not produced. Subsequent infection by a helper virus rescues the integrated genome, allowing it to replicate and package its genome into infectious AAV virions. While AAV can infect cells from different species, the helper virus must be of the same species as the host cell. Thus, for example, human AAV will replicate in canine cells co-infected with a canine adenovirus.


Adeno-associated virus (AAV) has been used with success in gene therapy. AAV has been engineered to deliver genes of interest by deleting the internal nonrepeating portion of the AAV genome (i.e., the rep and cap genes) and inserting a heterologous gene (in this case, the gene encoding the anti-inflammatory cytokine) between the ITRs. The heterologous gene is typically functionally linked to a heterologous promoter (constitutive, cell-specific, or inducible) capable of driving gene expression in the patient's target cells under appropriate conditions.


Recombinant AAV virions comprising a titin gene can be produced using a variety of art-recognized techniques. In one embodiment, a rAAV vector construct is packaged into rAAV virions in cells co-transfected with wild-type AAV and a helper virus, such as adenovirus. See, e.g., U.S. Pat. No. 5,139,941.


Alternatively, plasmids can be used to supply the necessary replicative functions from AAV and/or a helper virus. In one embodiment of the present invention, rAAV virions are produced using a plasmid to supply necessary AAV replicative functions (the “AAV helper functions”). See e.g., U.S. Pat. Nos. 5,622,856 and 5,139,941, both incorporated herein by reference in their entireties. In another embodiment, a triple transfection method is used to produce rAAV virions. The triple transfection method is described in detail in U.S. Pat. Nos. 6,001,650 and 6,004,797, which are incorporated by reference herein in their entireties. The triple transduction method is advantageous because it does not require the use of an infectious helper virus during rAAV production, enabling production of a stock of rAAV virions essentially free of contaminating helper virus. This is accomplished by use of three vectors for rAAV virion production: an AAV helper function vector, an accessory function vector, and a rAAV expression vector. One of skill in the art will appreciate, however, that the nucleic acid sequences encoded by these vectors can be provided on two or more vectors in various combinations. Vectors and cell lines necessary for preparing helper virus-free rAAV stocks are commercially available as the AAV Helper-Free System (Catalog No. 240071) (Stratagene, La Jolla, Calif.).


The AAV helper function vector encodes AAV helper function sequences (i.e., rep and cap) that function in trans for productive rAAV replication and encapsidation. Preferably, the AAV helper function vector supports efficient rAAV virion production without generating any detectable replication competent AAV virions (i.e., AAV virions containing functional rep and cap genes). An example of such a vector, pHLP19, is described in U.S. Pat. No. 6,001,650. The rep and cap genes of the AAV helper function vector can be derived from any of the known AAV serotypes. For example, the AAV helper function vector may have a rep gene derived from AAV-2 and a cap gene derived from AAV-6. One of skill in the art will recognize that other rep and cap gene combinations are possible, the defining feature being the ability to support rAAV virion production.


The accessory function vector encodes nucleotide sequences for non-AAV-derived viral and/or cellular functions upon which AAV is dependent for replication (the “accessory functions”). The accessory functions include those functions required for AAV replication, including, without limitation, genes involved in activation of AAV gene transcription, stage specific AAV mRNA splicing, AAV DNA replication, synthesis of cap expression products, and AAV capsid assembly. Viral-based accessory functions can be derived from any of the well-known helper viruses such as adenovirus, herpesvirus (other than herpes simplex virus type-1), and vaccinia virus. In one embodiment, the accessory function plasmid pLadeno5 can be used. See U.S. Pat. No. 6,004,797. This plasmid provides a complete set of adenovirus accessory functions for AAV vector production, but lacks the components necessary to form replication-competent adenovirus.


Unlike stocks of rAAV vectors prepared using infectious helper virus, stocks prepared using an accessory function vector (e.g. the triple transfection method) do not contain contaminating helper virus because no helper virus is added during rAAV production. Even after purification, for example by CsCl density gradient centrifugation, rAAV stocks prepared using helper virus still remain contaminated with some level of residual helper virus. When adenovirus is used as the helper virus in preparing a stock of rAAV virions, contaminating adenovirus can be inactivated by heating to temperatures of approximately 60° C. for 20 minutes or more. This treatment effectively inactivates only the helper virus since AAV is extremely heat stable, while the helper adenovirus is heat labile. Although heat inactivating of rAAV stocks may render much of the contaminating adenovirus non-infectious, it does not physically remove the helper virus proteins from the stock. Such contaminating viral protein can elicit undesired immune responses in subjects and are to be avoided if possible. Contaminating adenovirus particles and proteins in rAAV stocks can be avoided by use of the accessory function vectors disclosed herein.


Recombinant AAV Expression Vectors

Recombinant AAV expression vectors can be constructed using standard techniques of molecular biology. rAAV vectors comprise a transgene of interest (e.g. a gene encoding titin) flanked by AAV ITRs at both ends. rAAV vectors are also constructed to contain transcription control elements operably linked to the transgene sequence, including a transcriptional initiation region and a transcriptional termination region. The control elements are selected to be functional in a mammalian target cell.


The nucleotide sequences of AAV ITR regions are known. See, e.g., Kotin (1994) Human Gene Therapy 5:793-801; Berns “Parvoviridae and their Replication” in Fundamental Virology, 2nd Edition, (B. N. Fields and D. M. Knipe, eds.) for the AAV-2 sequence. AAV ITRs used in the vectors of the invention need not have a wild-type nucleotide sequence, and may be altered, e.g., by the insertion, deletion or substitution of nucleotides. Additionally, AAV ITRs may be derived from any of several AAV serotypes, including without limitation, AAV-1, AAV-2, AAV-3, AAV-4, AAV-5, AAV-6, AAV-7 and AAV-8, etc. Furthermore, 5′ and 3′ ITRs which flank a selected nucleotide sequence in an AAV expression vector need not necessarily be identical or derived from the same AAV serotype or isolate, so long as they function as intended, i.e., to allow for excision and rescue of the sequence of interest from a host cell genome or vector, and to allow integration of the DNA molecule into the recipient cell genome when AAV Rep gene products are present in the cell.


Suitable transgenes for delivery in AAV vectors will be less than about 5 kilobases (kb) in size. In one embodiment, a complete titin gene can be delivered with AAV vectors. The selected polynucleotide sequence is operably linked to control elements that direct the transcription thereof in the subject in vivo. Such control elements can comprise control sequences normally associated with the selected gene. Alternatively, heterologous control sequences can be employed. Useful heterologous control sequences generally include those derived from sequences encoding mammalian or viral genes. Examples include, but are not limited to, neuron-specific enolase promoter, a GFAP promoter, the SV40 early promoter, mouse mammary tumor virus LTR promoter; adenovirus major late promoter (Ad MLP); a herpes simplex virus (HSV) promoter, a cytomegalovirus (CMV) promoter such as the CMV immediate early promoter region (CMVIE), a rous sarcoma virus (RSV) promoter, synthetic promoters, hybrid promoters, and the like. In addition, sequences derived from nonviral genes, such as the murine metallothionein gene, will also find use herein. Such promoter sequences are commercially available from, e.g., Stratagene (San Diego, Calif.).


The AAV expression vector harboring a transgene of interest bounded by AAV ITRs can be constructed by directly inserting the selected sequence(s) into an AAV genome that has had the major AAV open reading frames (“ORFs”) excised. Other portions of the AAV genome can also be deleted, so long as enough of the ITRs remain to provide replication and packaging functions. Such constructs can be designed using techniques well known in the art. See, e.g., U.S. Pat. Nos. 5,173,414 and 5,139,941; International Publication Nos. WO 92/01070 and WO 93/03769; Lebkowski et al. (1988) Molec. Cell. Biol. 8:3988-96; Vincent et al. (1990) Vaccines 90 (Cold Spring Harbor Laboratory Press); Carter (1992) Current Opinion in Biotechnology 3:533-39; Muzyczka (1992) Current Topics in Microbiol. and Immunol. 158:97-129; Kotin (1994) Human Gene Therapy 5:793-801; Shelling and Smith (1994) Gene Therapy 1:165-69; and Zhou et al. (1994) J. Exp. Med. 179:1867-75.


AAV ITR-containing DNA fragments can be ligated at both ends of a selected transgene using standard techniques, such as those described in Sambrook et al., supra. For example, ligations can be accomplished in 20 mM Tris-Cl pH 7.5, 10 mM MgCl2, 10 mM DTT, 33 μg/ml BSA, 10 mM-50 mM NaCl, and either 40 μM ATP, 0.01-0.02 (Weiss) units T4 DNA ligase at 0° C. (for “sticky end” ligation) or 1 mM ATP, 0.3-0.6 (Weiss) units T4 DNA ligase at 14° C. (for “blunt end” ligation). Intermolecular “sticky end” ligations are usually performed at 30-100 μg/ml total DNA concentrations (5-100 nM total end concentration).


Suitable host cells for producing rAAV virions of the present invention from rAAV expression vectors include microorganisms, yeast cells, insect cells, and mammalian cells. Such host cells are preferably capable of growth in suspension culture, a bioreactor, or the like. The term “host cell” includes the progeny of the original cell that has been transfected with an rAAV virion. Cells from the stable human cell line, 293 (readily available through the American Type Culture Collection under Accession Number ATCC CRL1573) are preferred in the practice of the present invention. The human cell line 293 is a human embryonic kidney cell line that has been transformed with adenovirus type-5 DNA fragments (Graham et al. (1977) J. Gen. Virol. 36:59), and expresses the adenoviral E1a and E1b genes (Aiello et al. (1979) Virology 94:460). The 293 cell line is readily transfected, and provides a particularly convenient platform in which to produce rAAV virions.


Other Viral Vectors for Gene Delivery

Additional viral vectors useful for delivering the nucleic acid molecules of interest include those derived from the pox family of viruses, including vaccinia virus and avian poxvirus. By way of example, vaccinia virus recombinants expressing a gene of interest can be constructed as follows. DNA carrying the gene is inserted into an appropriate vector adjacent to a vaccinia promoter and flanking vaccinia DNA sequences, such as the sequence encoding thymidine kinase (TK). This vector is then used to transfect cells that are simultaneously infected with vaccinia. Homologous recombination serves to insert the vaccinia promoter and the gene into the viral genome. The resulting TK-recombinant can be selected by culturing the cells in the presence of 5-bromodeoxyuridine and picking viral plaques resistant thereto.


Alternatively, avipoxviruses, such as the fowlpox and canarypox viruses, can be used to deliver the genes. Recombinant avipox viruses expressing immunogens from mammalian pathogens are known to confer protective immunity when administered to non-avian species. The use of avipox vectors in human and other mammalian species is advantageous with regard to safety because members of the avipox genus can only productively replicate in susceptible avian species. Methods for producing recombinant avipoxviruses are known in the art and employ genetic recombination, as described above with respect to the production of vaccinia viruses. See, e.g., WO 91/12882; WO 89/03429; and WO 92/03545.


Molecular conjugate vectors, such as the adenovirus chimeric vectors, can also be used for gene delivery. Michael et al. (1993) J. Biol. Chem. 268:6866-69 and Wagner et al. (1992) Proc. Natl. Acad. Sci. USA 89:6099-6103. Members of the Alphavirus genus, for example the Sindbis and Semliki Forest viruses, may also be used as viral vectors for delivering the TTN gene or a fragment thereof. See, e.g., Dubensky et al. (1996) J. Virol. 70:508-19; WO 95/07995; WO 96/17072.


As used herein, the term “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.


As used herein, the term “pharmaceutically acceptable carrier” means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (i) sugars, such as lactose, glucose and sucrose; (ii) starches, such as corn starch and potato starch; (iii) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (iv) powdered tragacanth; (v) malt; (vi) gelatin; (vii) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (viii) excipients, such as cocoa butter and suppository waxes; (ix) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (x) glycols, such as propylene glycol; (xi) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol (PEG); (xii) esters, such as ethyl oleate and ethyl laurate; (xiii) agar; (xiv) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (xv) alginic acid; (xvi) pyrogen-free water; (xvii) isotonic saline; (xviii) Ringer's solution; (xix) ethyl alcohol; (xx) pH buffered solutions; (xxi) polyesters, polycarbonates and/or polyanhydrides; (xxii) bulking agents, such as polypeptides and amino acids (xxiii) serum component, such as serum albumin, HDL and LDL; (xxiv) C2-C12 alchols, such as ethanol; and (xxv) other non-toxic compatible substances employed in pharmaceutical formulations. Wetting agents, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants can also be present in the formulation.


The term “administer” or “administration” as used herein refers to the placement of a composition into a subject by a method or route which results in at least partial localization of the composition at a desired site such that desired effect is produced, such as intracranially to brain or specific areas of brain. Stereotactic means can be used to guide intracranial administration if desired. Routes of administration suitable for the methods of the invention include both local and systemic administration. Generally, local administration results in more of the composition being delivered to a specific location as compared to the entire body of the subject, whereas, systemic administration can result in delivery to essentially the entire body of the subject. However, it is envisioned that chemotropic property of NSCs can guide the cells to a specific location with a tissue injury, e.g., brain, even with systemic administration.


A composition described herein can be administered by any appropriate route known in the art including, but not limited to, oral or parenteral routes, including intravenous, intramuscular, subcutaneous, transdermal, and nasal administration.


Exemplary modes of administration include, but are not limited to, injection, infusion, instillation, inhalation, or ingestion. “Injection” includes, without limitation, intravenous, intramuscular, intraarterial, intraventricular, intracardiac, intradermal, intraperitoneal, subcutaneous, subcuticular, and intraarticular injection and infusion.


Kits for determining if a subject is at increased risk of developing DCM will include at least one reagent specific for detecting for the presence or absence of the TTN mutations described herein and instructions for observing that the subject is at increased risk of developing DCM if the presence of at least one of the TTN mutations described herein is detected. The kit may optionally include a nucleic acid for detection of the gene of interest.


In some embodiments, the invention also provides assays to identify a subject with an increased risk for developing DCM. In one embodiment, the assay comprises or consists essentially of a system for transforming and identifying at least one TTN mutation described herein in a biological sample of a subject, and a system for computing the likelihood of the subject getting DCM on the basis of comparison of the identified TNN mutation against the DCM-risk associated TTN mutations described herein. If the computing or comparison system, which can be a computer implemented system, indicates that at least one of the TTN mutations described herein is present in the sample, the subject from which the sample is collected can be diagnosed with increased risk for having or developing DCM.


Embodiments of the invention also provide for systems (and computer readable media for causing computer systems) to perform a method for determining presence or absence of TTN mutations associated with an increased risk of a subject for developing DCM. In one embodiment, provided herein is a system comprising: (a) a determination module configured to identify and detect at least one TTN mutation as described herein in a biological sample of a subject; (b) a storage module configured to store output data from the determination module; (c) a computing module adapted to identify from the output data at least one of DCM risk associated TTN mutations is present in the output data stored on the storage module; and (d) a display module for displaying if any of the DCM risk associated TTN mutations was identified or not, and/or displaying the detected TTN mutations.


Embodiments of the invention can be described through functional modules, which are defined by computer executable instructions recorded on computer readable media and which cause a computer to perform method steps when executed. The modules are segregated by function for the sake of clarity. However, it should be understood that the modules/systems need not correspond to discreet blocks of code and the described functions can be carried out by the execution of various code portions stored on various media and executed at various times. Furthermore, it should be appreciated that the modules can perform other functions, thus the modules are not limited to having any particular functions or set of functions.


In one embodiment, provided herein is a system comprising: (a) at least one memory containing at least one computer program adapted to control the operation of the computer system to implement a method that includes (i) a determination module configured to identify and/or detect the presence or absence of DCM-risk associated mutation in a sample obtained from a subject or the sequence of a nucleic acid comprising a titin-encoding DNA or RNA in a sample obtained from a subject; (ii) a storage module configured to store output data from the determination module; (iii) a computing module adapted to identify from the output data whether the nucleic acid sequence of the sample obtained from the subject differs from SEQ ID NO:1 or comprises a DCM-risk associated mutation as described herein and (iv) a display module for displaying whether the subject has an increased risk of having or developing DCM and/or is in need of treatment for DCM and/or displaying the presence or absence of a DCM-risk associated mutation as described herein (b) at least one processor for executing the computer program (see FIG. 9).


The computer readable storage media can be any available tangible media that can be accessed by a computer. Computer readable storage media includes volatile and nonvolatile, removable and non-removable tangible media implemented in any method or technology for storage of information such as computer readable instructions, data structures, program modules or other data. Computer readable storage media includes, but is not limited to, RAM (random access memory), ROM (read only memory), EPROM (eraseable programmable read only memory), EEPROM (electrically eraseable programmable read only memory), flash memory or other memory technology, CD-ROM (compact disc read only memory), DVDs (digital versatile disks) or other optical storage media, magnetic cassettes, magnetic tape, magnetic disk storage or other magnetic storage media, other types of volatile and non-volatile memory, and any other tangible medium which can be used to store the desired information and which can accessed by a computer including and any suitable combination of the foregoing.


Computer-readable data embodied on one or more computer-readable media may define instructions, for example, as part of one or more programs that, as a result of being executed by a computer, instruct the computer to perform one or more of the functions described herein, and/or various embodiments, variations and combinations thereof. Such instructions may be written in any of a plurality of programming languages, for example, Java, J#, Visual Basic, C, C#, C++, Fortran, Pascal, Eiffel, Basic, COBOL assembly language, and the like, or any of a variety of combinations thereof. The computer-readable media on which such instructions are embodied may reside on one or more of the components of either of a system, or a computer readable storage medium described herein, may be distributed across one or more of such components.


The computer-readable media may be transportable such that the instructions stored thereon can be loaded onto any computer resource to implement the aspects of the present invention discussed herein. In addition, it should be appreciated that the instructions stored on the computer-readable medium, described above, are not limited to instructions embodied as part of an application program running on a host computer. Rather, the instructions may be embodied as any type of computer code (e.g., software or microcode) that can be employed to program a computer to implement aspects of the present invention. The computer executable instructions may be written in a suitable computer language or combination of several languages. Basic computational biology methods are known to those of ordinary skill in the art and are described in, for example, Setubal and Meidanis et al., Introduction to Computational Biology Methods (PWS Publishing Company, Boston, 1997); Salzberg, Searles, Kasif, (Ed.), Computational Methods in Molecular Biology, (Elsevier, Amsterdam, 1998); Rashidi and Buehler, Bioinformatics Basics: Application in Biological Science and Medicine (CRC Press, London, 2000) and Ouelette and Bzevanis Bioinformatics: A Practical Guide for Analysis of Gene and Proteins (Wiley & Sons, Inc., 2nd ed., 2001).


The functional modules of certain embodiments of the invention include at minimum a determination module, a storage module, a computing module, and a display module. The functional modules can be executed on one, or multiple, computers, or by using one, or multiple, computer networks. The determination module has computer executable instructions to provide e.g., allelic variance etc in computer readable form.


The determination module can comprise any system for detecting a signal elicited from the SNPs described herein in a biological sample. In some embodiments, such systems can include an instrument, e.g., for genotyping such as Pyrosequencer described earlier. In another embodiment, the determination module can comprise multiple units for different functions, such as amplication and hybridization. In one embodiment, the determination module can be configured to perform the genotyping methods described in the Examples, including restriction enzyme digestion, ligation, PCR, purification, labeling, incubation and hybridization.


In some embodiments, the determination module can be further configured to identify and detect the presence of at least one additional DCM risk associated TTN mutation as described above herein.


The information determined in the determination system can be read by the storage module. As used herein the “storage module” is intended to include any suitable computing or processing apparatus or other device configured or adapted for storing data or information. Examples of electronic apparatus suitable for use with the present invention include stand-alone computing apparatus, data telecommunications networks, including local area networks (LAN), wide area networks (WAN), Internet, Intranet, and Extranet, and local and distributed computer processing systems. Storage modules also include, but are not limited to: magnetic storage media, such as floppy discs, hard disc storage media, magnetic tape, optical storage media such as CD-ROM, DVD, electronic storage media such as RAM, ROM, EPROM, EEPROM and the like, general hard disks and hybrids of these categories such as magnetic/optical storage media. The storage module is adapted or configured for having recorded thereon, for example, sample name and TTN mutations, and frequency of each TTN mutation. Such information may be provided in digital form that can be transmitted and read electronically, e.g., via the Internet, on diskette, via USB (universal serial bus) or via any other suitable mode of communication.


As used herein, “stored” refers to a process for encoding information on the storage module. Those skilled in the art can readily adopt any of the presently known methods for recording information on known media to generate manufactures comprising expression level information.


In one embodiment of any of the systems described herein, the storage module stores the output data from the determination module. In additional embodiments, the storage module stores the reference information such as DCM risk associated TTN mutations at the allels described herein, and/or the wild-type sequence in subjects who do not have symptoms associated with DCM.


The “computing module” can use a variety of available software programs and formats for computing the presence or absence of at least one TTN mutation described herein and identifying the presence or absence of at least one of DCM risk associated mutation described herein. Genotyping algorithms are well established in the art. A skilled artisan is readily able to determine the appropriate genotyping algorithms based on the size and quality of the sample. Genotyping algorithms, e.g., DM or BRLMM, and statistics tools for data analysis described in Examples can be implemented in the computing module of the invention. In one embodiment, the computing module further comprises a comparison module, which compares the genotype determined at the TTN mutations described herein with the DCM-risk associated TTN mutations and/or wide-type TTN sequence and the comparison module can generate an output indicating undetermined risk for AD. In various embodiments, the comparison module can be configured using existing commercially-available or freely-available software for comparison purpose, and may be optimized for particular data comparisons that are conducted. (see FIG. 10)


The computing and/or comparison module, or any other module of the invention, can include an operating system (e.g., UNIX) on which runs a relational database management system, a World Wide Web application, and a World Wide Web server. World Wide Web application includes the executable code necessary for generation of database language statements (e.g., Structured Query Language (SQL) statements). Generally, the executables will include embedded SQL statements. In addition, the World Wide Web application may include a configuration file which contains pointers and addresses to the various software entities that comprise the server as well as the various external and internal databases which must be accessed to service user requests. The Configuration file also directs requests for server resources to the appropriate hardware—as may be necessary should the server be distributed over two or more separate computers. In one embodiment, the World Wide Web server supports a TCP/IP protocol. Local networks such as this are sometimes referred to as “Intranets.” An advantage of such Intranets is that they allow easy communication with public domain databases residing on the World Wide Web (e.g., the GenBank or Swiss Pro World Wide Web site). Thus, in a particular preferred embodiment of the present invention, users can directly access data (via Hypertext links for example) residing on Internet databases using a HTML interface provided by Web browsers and Web servers (FIG. 11).


The computing and/or comparison module provides a computer readable comparison result that can be processed in computer readable form by predefined criteria, or criteria defined by a user, to provide a content-based in part on the comparison result that may be stored and output as requested by a user using an output module, e.g., a display module.


In some embodiments, the content displayed on the display module can be a genotype identified in the biological sample of the subject together with a reference sequence. For example, the reference sequence can be a DCM-risk associated TTN mutation or a wild-type TTN sequence. In some embodiments, the content displayed on the display module can be a numerical value indicating the probability of developing DCM. In such embodiments, the probability can be expressed in percentages or a fraction of developing DCM. For example, higher percentage or a fraction closer to 1 indicates a higher likelihood of a subject going to to be affected by DCM. In some embodiments, the content displayed on the display module can be single word or phrases to quanlitatively indicate the likelihood of a subject going to be affected with DCM. For example, a word “unlikely” can be used to indicate a lower risk for DCM, while “likely” can be used to indicate a high risk for DCM.


In one embodiment of the invention, the content based on the computing and/or comparison result is displayed on a computer monitor. In one embodiment of the invention, the content based on the computing and/or comparison result is displayed through printable media. The display module can be any suitable device configured to receive from a computer and display computer readable information to a user. Non-limiting examples include, for example, general-purpose computers such as those based on Intel PENTIUM-type processor, Motorola PowerPC, Sun UltraSPARC, Hewlett-Packard PA-RISC processors, any of a variety of processors available from Advanced Micro Devices (AMD) of Sunnyvale, Calif., or any other type of processor, visual display devices such as flat panel displays, cathode ray tubes and the like, as well as computer printers of various types.


In one embodiment, a World Wide Web browser is used for providing a user interface for display of the content based on the computing/comparison result. It should be understood that other modules of the invention can be adapted to have a web browser interface. Through the Web browser, a user can construct requests for retrieving data from the computing/comparison module. Thus, the user will typically point and click to user interface elements such as buttons, pull down menus, scroll bars and the like conventionally employed in graphical user interfaces.


Systems and computer readable media described herein are merely illustrative embodiments of the invention for identifying at least one TTN mutation described herein in a subject and determining a risk of the subject for developing DCM, and therefore are not intended to limit the scope of the invention. Variations of the systems and computer readable media described herein are possible and are intended to fall within the scope of the invention.


The modules of the machine, or those used in the computer readable medium, may assume numerous configurations. For example, function may be provided on a single machine or distributed over multiple machines.


Methods and compositions described herein relating to determining if a subject is at increased risk of having or developing DCM can relate to determining the presence of a TTN mutation in a sample obtained from the subject. In some embodiments, the sample is a nucleic acid sample. A sample obtained from a subject can be a biological sample. “Biological sample” as used herein denotes a sample taken or isolated from a biological organism, e.g., tissue cell culture supernatant, cell lysate, a homogenate of a tissue sample from a subject or a fluid sample from a subject. Exemplary biological samples include, but are not limited to, blood, sputum, urine, spinal fluid, pleural fluid, nipple aspirates, lymph fluid, the external sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, milk, feces, sperm, cells or cell cultures, serum, leukocyte fractions, smears, tissue samples of all kinds, embryos, etc and mixtures or combinations thereof. The term “biological sample” also includes untreated or pretreated (or pre-processed) biological samples.


A “biological sample” can contain cells from subject, but the term can also refer to non-cellular biological material, such as non-cellular fractions of blood, saliva, or urine, that can be used to determine the presence of a TTN mutation as described herein. In some embodiments, the sample is from a resection, biopsy, or core needle biopsy. In addition, fine needle aspirate samples can be used. Samples can be either paraffin-embedded or frozen tissue.


The sample can be obtained by removing a sample of cells from a subject, but can also be accomplished by using previously isolated cells (e.g. isolated by another person). In addition, the biological sample can be freshly collected or a previously collected sample. Furthermore, the biological sample can be utilized for the detection of the presence and/or quantitative level of a biomolecule of interest. Representative biomolecules include, but are not limited to, DNA, RNA, mRNA, polypeptides, and derivatives and fragments thereof. In some embodiments, the biological sample can be used for TTN mutation determination for diagnosis of a disease or a disorder, e.g., DCM, using the methods, assays and systems of the invention.


In some embodiments, biological sample is a biological fluid. Examples of biological fluids include, but are not limited to, saliva, bone marrow, blood, serum, plasma, urine, sputum, cerebrospinal fluid, an aspirate, tears, and any combinations thereof.


In some embodiments, the biological sample is an untreated biological sample. As used herein, the phrase “untreated biological sample” refers to a biological sample that has not had any prior sample pre-treatment except for dilution and/or suspension in a solution. Exemplary methods for treating a biological sample include, but are not limited to, centrifugation, filtration, sonication, homogenization, heating, freezing and thawing, and any combinations thereof.


In some embodiments, the biological sample is a frozen biological sample, e.g., a frozen tissue or fluid sample such as urine, blood, serum or plasma. The frozen sample can be thawed before employing methods, assays and systems of the invention. After thawing, a frozen sample can be centrifuged before being subjected to methods, assays and systems of the invention.


In some embodiments, the biological fluid sample can be treated with at least one chemical reagent, such as a nuclease inhibitor. In some embodiments, the biological fluid sample is a clarified biological fluid sample, for example, by centrifugation and collection of a supernatant comprising the clarified biological fluid sample.


In some embodiments, a biological sample is a pre-processed biological sample, for example, supernatant or filtrate resulting from a treatment selected from the group consisting of centrifugation, filtration, sonication, homogenization, lysis, thawing, amplification, purification, restriction enzyme digestion ligation and any combinations thereof. In some embodiments, a biological sample can be a nucleic acid product amplified after polymerase chain reaction (PCR). The term “nucleic acid” used herein refers to DNA, RNA, or mRNA.


In some embodiments, the biological sample can be treated with a chemical and/or biological reagent. Chemical and/or biological reagents can be employed to protect and/or maintain the stability of the sample, including biomolecules (e.g., nucleic acid and protein) therein, during processing. One exemplary reagent is a protease inhibitor, which is generally used to protect or maintain the stability of protein during processing. In addition, or alternatively, chemical and/or biological reagents can be employed to release nucleic acid or protein from the sample.


The skilled artisan is well aware of methods and processes appropriate for pre-processing of biological samples required for determination of TTN mutations as described herein.


Nucleic acid and ribonucleic acid (RNA) molecules can be isolated from a particular biological sample using any of a number of procedures, which are well-known in the art, the particular isolation procedure chosen being appropriate for the particular biological sample. For example, freeze-thaw and alkaline lysis procedures can be useful for obtaining nucleic acid molecules from solid materials; heat and alkaline lysis procedures can be useful for obtaining nucleic acid molecules from urine; and proteinase K extraction can be used to obtain nucleic acid from blood (Roiff, A et al. PCR: Clinical Diagnostics and Research, Springer (1994)).


The invention can be further illustrated by any of the following numbered paragraphs:

  • 1. An assay for determining if a subject has an increased risk for developing a dilated cardiomyopathy (DCM) or increased risk for heart failure or is in need of treatment for DCM, the assay comprising:
    • (i) sequencing at least a portion of TTN gene in a sample from a human subject to detect a TTN nucleic acid which results in a truncated TITIN polypeptide; and
    • (ii) detecting if any such mutation is present in the nucleic acid sample,
    • wherein detection of the presence of at least one said mutation is indicative of the subject having an increased risk for developing DCM.
  • 2. The assay of paragraph 1, wherein said nucleic acid sequencing is dideoxy sequencing, Maxam-Gilbert sequencing, dye-terminator sequencing, Lynx Therapeutics' Massively Parallel Sequencing (MPSS) Polony sequencing, 454 Pyrosequencing, Illumina (Solexa) sequencing, Single Molecule real time (RNAP) sequencing, Nanaopore DNA sequencing, or sequencing by technology from VisiGen Biotechnologies.
  • 3. The assay of paragraph 1 or 2, wherein the truncated TITIN polypeptide lacks part of the A-band, having a wild-type sequence set forth in SEQ ID NO: 725.
  • 4. The assay of any of paragraphs 1-3, wherein said TTN nucleic acid mutation is selected from the group consisting of 62476247delG, 12745C>T, 1447014471insCACACTCCATA (SEQ ID NO: 722), 1918319183delG, 2379823810de1GTCAAGATATCTG (SEQ ID NO: 723), 3862138622insA, 4433644336delA, 4532245322de11, 49077G>A, 51883C>T, 52408C>T, 5314553146insG, 53347G>T, 5393553935delC, 56367T>A, 56572C>T, 56953C>T, 58678C>T, 59530C>T, 6104661046delC, 6586765867delA, 6705767063de1GCATATGinsTA, 6774567745delT, 7217872179insT, 7272372739delinsAGA, 77065C>T, 79896G>A, 80845C>T, 81046A>T, 81440G>A, 8153681537delCT, 82701C>A, 8497784980de1ATTA, 87953G>A, 88242C>T, 88528G>T, 8917789181de1AAATT, 90241C>T, 9104291042delA, 9153791538insA, 94111A>T, 95522C>A, 30476-1G>A, 34186+1G>T, 35635G>C, 35635+1G>A, 44725+2delT, 48364+1G>T, 50346+3A>G, 54422-5T>A, 54704-1G>A, 55003+1G>A, 62425+5G>A, 63405A>G, 64489+1G>A, 81898+2T>A, 92569+1G>C, and any combination thereof, wherein the mutation location is determined based upon the wildtype TTN sequence having a nucleic acid sequence set forth in SEQ ID NO: 1.
  • 5. The assay of any of paragraphs 1-4, wherein said sequencing comprises contacting the nucleic acid sample with a probe or primer.
  • 6. The assay of any of paragraphs 1-5, wherein the subject is one who exhibits one or more risk factors for DCM or one or more complications related to DCM or a subject who does not exhibit risk factors or a subject who is a family member of an individual who has been diagnosed with a cardiac condition.
  • 7. The assay of any of paragraphs 1-6, wherein the subject is asymptomatic.
  • 8. The assay of any of paragraphs 1-7, wherein dilated cardiomyopathy is idiopathic dilated cardiomyopathy.
  • 9. A method of treating a subject for dilated cardiomyopathy, comprising:
    • (i) selecting a subject at risk for developing DCM or in need of treatment for DCM using an assay of paragraph 1; and
    • (ii) administering a treatment for DCM to the subject.
  • 10. The method of paragraph 9, wherein the treatment is selected from the group consisting of angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers, beta blockers, diuretics, aldosterone antagonists, digoxin (Lanoxin), blood thinning medications, biventricular pacemakers, implantable cardioverter-defibrillators (ICDs), heart pumps (left ventricular assist devices, or LVADs), heart transplant, gene therapy, calcium channel blockers, tissue growth factor inhibitors, and any combinations thereof.
  • 11. An in vitro assay comprising:
    • (iii) contacting a nucleic acid sample obtained from a subject with a probe, wherein the probe is capable of detecting a mutation resulting in a truncated TITIN polypeptide;
    • (iv) detecting the presence of the mutation in the nucleic acid sample,
    • wherein detection of the presence of at least one said mutation is indicative of the subject having an increased risk for developing DCM.
  • 12. The assay of paragraph 11, wherein the probe is a sequencing primer.
  • 13. An in vitro assay for determining if a subject has an increased risk for developing a dilated cardiomyopathy (DCM), the assay comprising:
    • (i) transforming a portion of a titin (TTN) nucleic acid in a sample obtained from the subject into at least one detectable target;
    • (ii) detecting presence or absence of at least one mutation in the TTN nucleic acid, wherein the at least one mutation in the TTN nucleic acid results in a truncated TITIN polypeptide
    • wherein detection of the presence of at least one of the mutation is indicative of the subject having an increased risk for developing DCM.
  • 14. An in vitro assay for determining if a subject is need of treatment for DCM, the method comprising:
    • (i) sequencing at least a portion of TTN gene in a sample from a human subject;
    • (ii) comparing the sequence obtained in step (i) with wild type TTN gene sequence having a nucleic acid sequence set forth in SEQ ID NO: 1; and
    • (iii) determining if the variation is one or more which results in a truncated (shortened) TITIN polypeptide; and
    • wherein detection of at least one of the variations is indicative of subject is in need of treatment for DCM.
  • 15. The in vitro assay of paragraph 14, further comprising administering a treatment for DCM to the subject, if presence of at least one the variations is detected.
  • 16. The in vitro assay of any of paragraphs 11-15, wherein the truncated TITIN polypeptide lacks part of the A-band, having a wild-type sequence set forth in SEQ ID NO: 725.
  • 17. The in vitro assay of any of paragraphs 11-16, wherein the mutation is selected from the group consisting of 62476247delG, 12745C>T, 1447014471insCACACTCCATA (SEQ ID NO: 722), 1918319183delG, 2379823810de1GTCAAGATATCTG (SEQ ID NO: 723), 3862138622insA, 4433644336delA, 4532245322delT, 49077G>A, 51883C>T, 52408C>T, 5314553146insG, 53347G>T, 5393553935delC, 56367T>A, 56572C>T, 56953C>T, 58678C>T, 59530C>T, 6104661046delC, 6586765867delA, 6705767063de1GCATATGinsTA, 6774567745delT, 7217872179insT, 7272372739delinsAGA, 77065C>T, 79896G>A, 80845C>T, 81046A>T, 81440G>A, 8153681537delCT, 82701C>A, 8497784980de1ATTA, 87953G>A, 88242C>T, 88528G>T, 8917789181de1AAATT, 90241C>T, 9104291042delA, 9153791538insA, 94111A>T, 95522C>A, 30476-1G>A, 34186+1G>T, 35635G>C, 35635+1G>A, 44725+2delT, 48364+1G>T, 50346+3A>G, 54422-5T>A, 54704-1G>A, 55003+1G>A, 62425+5G>A, 63405A>G, 64489+1G>A, 81898+2T>A, 92569+1G>C, and any combination thereof, wherein the mutation location is determined based upon the wildtype TTN sequence having a nucleic acid sequence set forth in SEQ ID NO: 1.
  • 18. The in vitro assay of any of paragraphs 11-17, wherein the step of detecting the mutation is performed by nucleic acid sequencing.
  • 19. The in vitro assay of paragraph 18, wherein said nucleic acid sequencing is Maxam-Gilbert sequencing, dye-terminator sequencing, Lynx Therapeutics' Massively Parallel Sequencing (MPSS) Polony sequencing, 454 Pyrosequencing, Illumina (Solexa) sequencing, Single Molecule real time (RNAP) sequencing, Nanaopore DNA sequencing, or sequencing by technology from VisiGen Biotechnologies.
  • 20. The in vitro assay of any of paragraph 11-19, wherein detection of the mutation is by a computer implemented system.
  • 21. The in vitro assay of any of paragraphs 11-20, further comprising the step of displaying the result on a display module.
  • 22. The in vitro assay of any of paragraphs 11-21, wherein dilated cardiomyopathy is idiopathic dilated cardiomyopathy or heart failure or any other related cardiomyopathy.
  • 23. The in vitro assay of any of paragraphs 11-22, wherein the subject is one who exhibits one or more risk factors for DCM or one or more complications related to DCM or a subject who does not exhibit risk factors or a subject who is a family member of an individual who has been diagnosed with a cardiac condition.
  • 24. The in vitro assay of any of paragraphs 11-23, wherein the subject is asymptomatic.
  • 25. A method of treating a subject for dilated cardiomyopathy, comprising:
    • (iii) selecting a subject at risk for developing DCM or in need of treatment for DCM using an in vitro assay of any of paragraphs 11-24; and
    • (iv) administering a treatment for DCM to the subject.
  • 26. The method of paragraph 25, wherein the treatment is selected from the group consisting of angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers, beta blockers, diuretics, aldosterone antagonists, digoxin (Lanoxin), blood thinning medications, biventricular pacemakers, implantable cardioverter-defibrillators (ICDs), heart pumps (left ventricular assist devices, or LVADs), heart transplant, gene therapy, calcium channel blockers, tissue growth factor inhibitors, and any combinations thereof.
  • 27. The method of paragraph 26, wherein the treatment comprises administering to said subject a polypeptide comprising amino acid SEQ ID NO: 2 or a nucleic acid encoding a polypeptide comprising amino acid SEQ ID NO: 2.
  • 28. The method of paragraph 27, wherein the nucleic acid encoding the polypeptide of amino acid SEQ ID NO: 2 is operably linked to a vector.
  • 29. The method any of paragraphs 25-28, wherein the subject is one who exhibits one or more risk factors for DCM or one or more complications related to DCM or a subject who does not exhibit risk factors or a subject who is a family member of an individual who has been diagnosed with a cardiac condition.
  • 30. The method of any of paragraphs 25-29, wherein the subject is asymptomatic.
  • 31. A method comprising administering a treatment for dilated cardiomyopathy to a subject determined to have a mutation in TTN gene which results in a truncated TITIN polypeptide.
  • 32. The method of paragraph 31, wherein the truncated TITIN polypeptide lacks part of the A-band, having a wild-type sequence set forth in SEQ ID NO: 725.
  • 33. The method of paragraph 31 or 32, wherein the mutation is selected from the group consisting of 62476247delG, 12745C>T, 1447014471insCACACTCCATA (SEQ ID NO: 722), 1918319183delG, 2379823810de1GTCAAGATATCTG (SEQ ID NO: 723), 3862138622insA, 4433644336delA, 4532245322delT, 49077G>A, 51883C>T, 52408C>T, 5314553146insG, 53347G>T, 5393553935delC, 56367T>A, 56572C>T, 56953C>T, 58678C>T, 59530C>T, 6104661046delC, 6586765867delA, 6705767063de1GCATATGinsTA, 6774567745delT, 7217872179insT, 7272372739delinsAGA, 77065C>T, 79896G>A, 80845C>T, 81046A>T, 81440G>A, 8153681537delCT, 82701C>A, 8497784980de1ATTA, 87953G>A, 88242C>T, 88528G>T, 8917789181de1AAATT, 90241C>T, 9104291042delA, 9153791538insA, 94111A>T, 95522C>A, 30476-1G>A, 34186+1G>T, 35635G>C, 35635+1G>A, 44725+2delT, 48364+1G>T, 50346+3A>G, 54422-5T>A, 54704-1G>A, 55003+1G>A, 62425+5G>A, 63405A>G, 64489+1G>A, 81898+2T>A, 92569+1G>C, and any combination thereof, wherein the mutation location is determined based upon the wildtype TTN sequence having a nucleic acid sequence set forth in SEQ ID NO: 1.
  • 34. The method any of paragraphs 31-33, wherein detection of the mutation is by nucleic acid sequencing.
  • 35. The assay of paragraph 34, wherein said nucleic acid sequencing is Maxam-Gilbert sequencing, dye-terminator sequencing, Lynx Therapeutics' Massively Parallel Sequencing (MPSS) Polony sequencing, 454 Pyrosequencing, Illumina (Solexa) sequencing, SOLiD™ sequencing, Single Molecule SMART™ sequencing, Single Molecule real time (RNAP) sequencing, Nanaopore DNA sequencing, or sequencing by technology from VisiGen Biotechnologies.
  • 36. The assay of any of paragraph 31-35, wherein detection of the mutation is by a computer implemented system.
  • 37. The assay of any of paragraphs 31-36, further comprising the step of displaying the detection of the mutation on a display module.
  • 38. The assay of any of paragraphs 31-37, wherein dilated cardiomyopathy is idiopathic dilated cardiomyopathy.
  • 39. The assay of any of paragraphs 31-38, wherein the subject is one who exhibits one or more risk factors for DCM or one or more complications related to DCM or a subject who does not exhibit risk factors or a subject who is a family member of an individual who has been diagnosed with a cardiac condition.
  • 40. The assay of any of paragraphs 31-39, wherein the subject is asymptomatic.
  • 41. A computer implemented system for determining presence or absence of nucleic acid mutation associated with an increased risk of a subject for developing DCM, the system comprising:
    • (i) a determination module configured to identify and detect at least one nucleic acid mutation in TTN gene of SEQ ID NO: 1, wherein the nucleic acid mutation is selected from the group consisting of 62476247delG, 12745C>T, 1447014471insCACACTCCATA (SEQ ID NO: 722), 1918319183delG, 2379823810de1GTCAAGATATCTG (SEQ ID NO: 723), 3862138622insA, 4433644336delA, 4532245322delT, 49077G>A, 51883C>T, 52408C>T, 5314553146insG, 53347G>T, 5393553935delC, 56367T>A, 56572C>T, 56953C>T, 58678C>T, 59530C>T, 6104661046delC, 6586765867delA, 6705767063de1GCATATGinsTA, 6774567745delT, 7217872179insT, 7272372739delinsAGA, 77065C>T, 79896G>A, 80845C>T, 81046A>T, 81440G>A, 8153681537delCT, 82701C>A, 8497784980de1ATTA, 87953G>A, 88242C>T, 88528G>T, 8917789181de1AAATT, 90241C>T, 9104291042delA, 9153791538insA, 94111A>T, 95522C>A, 30476-1G>A, 34186+1G>T, 35635G>C, 35635+1G>A, 44725+2delT, 48364+1G>T, 50346+3A>G, 54422-5T>A, 54704-1G>A, 55003+1G>A, 62425+5G>A, 63405A>G, 64489+1G>A, 81898+2T>A, 92569+1G>C, and any combination thereof;
    • (ii) a storage module configured to store output data from the determination module;
    • (iii) a computing module adapted to identify from the output data at least one of DCM risk associated mutation is present in the output data stored on the storage module; and
    • (iv) a display module for displaying if any of the DCM risk associated mutation was identified or not, and/or displaying the detected mutation.
  • 42. The computer implemented system of paragraph 41, wherein the determination module comprises a system that transforms any nucleic acid or mutation therein into a detectable molecule.
  • 43. The computer implemented system of paragraph 42, further comprising a system that detects the detectable molecule.
  • 44. The computer implemented system of any of paragraphs 41-43, wherein the determination module comprises a system for contacting a nucleic acid sample obtained from a subject with a probe, wherein the probe is capable of detecting the TTN nucleic acid mutation.


Unless stated otherwise, or implicit from context, the following terms and phrases include the meanings provided below. Unless explicitly stated otherwise, or apparent from context, the terms and phrases below do not exclude the meaning that the term or phrase has acquired in the art to which it pertains. The definitions are provided to aid in describing particular embodiments, and are not intended to limit the claimed invention, because the scope of the invention is limited only by the claims. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular.


As used herein the term “comprising” or “comprises” is used in reference to compositions, methods, and respective component(s) thereof, that are useful to the invention, yet open to the inclusion of unspecified elements, whether useful or not.


The singular terms “a,” “an,” and “the” include plural referents unless context clearly indicates otherwise. Similarly, the word “or” is intended to include “and” unless the context clearly indicates otherwise.


As used herein, the term “about” means 2.5% of the value being referred to. For example, about 10 means from 7.5 to 12.5.


Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of this disclosure, suitable methods and materials are described below. The term “comprises” means “includes.” The abbreviation, “e.g.” is derived from the Latin exempli gratia, and is used herein to indicate a non-limiting example. Thus, the abbreviation “e.g.” is synonymous with the term “for example.”


The terms “decrease”, “reduced”, “reduction”, “decrease” or “inhibit” are all used herein generally to mean a decrease by a statistically significant amount. However, for avoidance of doubt, ““reduced”, “reduction” or “decrease” or “inhibit” means a decrease by at least 10% as compared to a reference level. In some embodiments, decrease can be at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% decrease (e.g. absent level as compared to a reference sample), or any decrease between 10-100% as compared to a reference level.


The terms “increased”,“increase” or “enhance” or “activate” are all used herein to generally mean an increase by a statically significant amount; for the avoidance of any doubt, the terms “increased”, “increase” or “enhance” or “activate” means an increase of at least 10% as compared to a reference level. In some embodiments, increase can be at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.


The term “statistically significant” or “significantly” refers to statistical significance and generally means at least two standard deviation (2SD) away from a reference level. The term refers to statistical evidence that there is a difference. It is defined as the probability of making a decision to reject the null hypothesis when the null hypothesis is actually true.


The disclosure is further illustrated by the following examples which should not be construed as limiting. The examples are illustrative only, and are not intended to limit, in any manner, any of the aspects described herein. The following examples do not in any way limit the invention.


EXAMPLES
Example 1
Truncation of Titin Causing Dilated Cardiomyopathy

Dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM) arise from mutations in many genes. The TTN gene, which encodes the sarcomere protein titin, has been insufficiently interrogated for cardiomyopathy mutations because of its enormous size.


TTN was analyzed in 312 DCM subjects, 231 HCM subjects, and 249 control subjects using next-generation or dideoxy sequencing. Deleterious variants were evaluated for co-segregation in families and assessed clinical characteristics.


Seventy two mutations were identified (25 nonsense, 23 frameshift, 23 splicing, and one large tandem insertion) that altered full-length titin. Among subjects studied by next-generation sequencing, TTN mutations were strongly enriched in DCM (54 of 203; 27%) versus HCM (3 of 231; 1.3%; P=3×10−16) or control subjects (7 of 249; 2.8%; P=9×10−14). TTN mutations co-segregated with DCM in families (combined LOD score=11.1) with high (>95%) penetrance after the age of 40 years. DCM mutations were overrepresented in the A-band but absent from the Z-disk and M-band regions of titin (P≦0.01). Overall, cardiac outcomes were similar in subjects with and without TTN mutations, but adverse events occurred earlier in male than female mutation carriers (P=4×10−5).


TTN truncating mutations are a common cause of DCM, occurring in approximately 25% of familial and 18% of sporadic idiopathic DCM. Incorporation of next-generation sequencing approaches that detect TTN truncations into DCM genetic testing should substantially increase test sensitivity, thereby enabling earlier diagnosis and therapeutic intervention for many DCM subjects. Defining the functional impact of TTN truncating mutations should improve understanding of DCM pathophysiology.


Gene mutation is an important cause of cardiomyopathy. Mutations in eight sarcomere protein genes cause hypertrophic cardiomyopathy (HCM) and are detected in 40 to 70% of HCM patients1, 2. Variations in over 40 genes, most of which encode components of the sarcomere, the cytoskeleton, or the nuclear lamina, have been demonstrated or posited to cause dilated cardiomyopathy (DCM)3, 4. Clinical evaluation identifies affected or likely-affected family members in 30 to 50% of DCM cases5-7, implicating a genetic etiology, but pathogenic mutations have been found in only 20 to 30% of cases8.


TTN, the gene encoding titin, has been implicated in cardiomyopathy, but has been incompletely studied due to technical challenges posed by the monumental size of its coding sequence (˜100 kb). TTN mutations have been definitively linked to DCM in 3 families9-11, but not to HCM. Additionally, TTN mutations have been implicated in congenital myopathies involving cardiac and skeletal muscle, hereditary myopathy with early respiratory failure, tibial muscular dystrophy, and limb-girdle muscular dystrophy12-15.


Titin is the largest human protein (˜33,000 amino acids) and the third most abundant striated muscle protein16. Two titin molecules together span the sarcomere (˜2 μLB) and are anchored at the Z-line and M-line (FIG. 1)17. Titin is necessary for sarcomere assembly18, 19, provides the majority of passive force20, 21 and modulates active contractile force22, 23. There are many different isoforms of titin, which in the heart are classified predominantly as N2B and N2BA (FIG. 1)24. TTN also encodes a separate cardiac isoform, novex-3 titin, which is only 5,600 amino acid, lacks the A-band and M-band segments of titin25 and is less abundant in cardiac tissue than full-length titin.


Filter-based hybridization capture followed by next-generation sequencing26 or traditional dideoxy sequencing was undertaken to assess the contribution of TTN mutations to cardiomyopathies, analyzing 312 subjects with idiopathic DCM, 231 subjects with HCM, and 249 control subjects.


Materials and Methods

Subjects:


Studies were performed according to institutional guidelines and human tissue act UK guidelines or local ethics committee approval. Idiopathic DCM subjects were studied from three cohorts (Table 5): 92 subjects recruited at Brigham and Women's Hospital (BWH) (DCM-A); 71 subjects recruited during cardiac transplant evaluation at the Royal Brompton and Harefield NHS Trust (DCM-B); and 149 subjects prospectively recruited in Colorado or Italy into a Familial Dilated Cardiomyopathy Registry (DCM-C). DCM-A and DCM-C cohorts were enriched for familial disease. HCM subjects (N=231) were recruited at BWH or the Mayo Clinic. DCM and HCM subjects were diagnosed using published criteria27, 28. Control subjects (N=249) without cardiomyopathy were recruited from multiple sites. No subjects within cohorts had a known familial relationship.


Dna Sequencing And Genotyping

Genomic DNA isolated from DCM-A, DCM-B, HCM, and control subjects were used to construct DNA libraries. DNA libraries were then enriched for TTN using filter-based hybridization capture26 with minor modifications (Tables 3, 4) and studied by single- or paired-end sequencing using an Illumina Genome Analyzer II or HiSeq29. TTN sequence was assessed in DCM-C subjects by traditional Sanger dideoxy sequencing, performed by the University of Washington, Department of Genome Sciences.


Dna Sequence Analyses:


Next-generation sequence data was analyzed using a custom pipeline integrating existing tools, including NOVALIGN™ (http://www.novocraft.com) and the GENOME ANALYSIS TOOLKIT™30, and Perl (using Bio-Samtools) and R31 scripts. Primary analyses of TTN variations were performed among subjects studied by the same approach (Table 1) to control for differences in variant detection. The amino acid positions of titin variants were identified using the UniProt titin sequence (Q8WZ42) and mutations were reported using Human Genome Variation Society nomenclature (Table 6). Variant confirmation and genotyping was performed by polymerase-chain reaction amplification followed by dideoxy sequencing, restriction digestion, gel electrophoresis26 and/or RNA sequencing of cardiac tissue32.


Statistical Analyses:


Association and cross-cohort analyses were performed using Fisher's exact tests, exact conditional tests of independence, or goodness of fit tests unless otherwise specified. The uniformity of the spatial distribution of mutations was assessed using a chi-square goodness of fit test, incorporating the size of each region. The clinical characteristics of subsets of each cohort were compared using two-tailed, unpaired t-tests. Kaplan-Meier curves were computed using software survfit and compared using coxph in R31.


Two-point logarithm of the odds (LOD) scores were calculated in 19 DCM families (FIG. 5; Table 15) using FASTLINK™ software, computed at theta=0, phenocopy rate=0.00533, and indicated disease penetrances34. An indeterminate status was assigned to family members≦40 years old without clinical criteria for DCM27 and to family members with confounding cardiac diagnoses.


Subject Cohorts:


Study subjects with dilated cardiomyopathy (DCM) were recruited form 3 independent groups. Group A was recruited from among DCM patients clinically evaluated at the Brigham and Women's Cardiovascular Genetics Center and subjects referred directly for research genetic evaluation. Group B was recruited from UK patients undergoing evaluation for cardiac transplantation with sufficient DNA samples for genetic analyses. Group C was recruited from subjects prospectively recruited in Colorado or Italy into a Familial Dilated Cardiomyopathy Registry. Ischemic heart disease was assessed in subjects from Groups A and C non-invasive studies and/or cardiac catheterization. All subjects in cohort DCM-B had coronary arteriography. Subjects who were found to have significant coronary artery disease or who were diagnosed with DCM in infancy were excluded from study. Groups A and C were enriched for subjects with a known family history of DCM. Concurrent genetic studies of other previously identified DCM genes1-10 in subjects recruited from the Brigham and Women's Cardiovascular Genetics Center, led to the identification of likely pathogenic mutations in 40 subjects. These 40 group A subjects did not undergo TTN sequencing or phenotypic analyses reported here. However, to avoid untended inflation of the frequency of TTN truncating mutations in DCM, the size of group A was increased by 40 subjects for comparisons of mutation frequencies.


The hypertrophic cardiomyopathy (HCM) cohort consisted of 136 subjects recruited from the Brigham and Women's Cardiovascular Center, and subjects referred directly for research genetic studies and 100 HCM subjects who underwent septal myectomy at the Mayo Clinic, Rochester, Minn.


DCM and HCM subjects were diagnosed according to standard guidelines11, 12 without knowledge of genotype. Control samples (n=249) are from de-identified subjects, recruited from multiple sites, with no known history of idiopathic cardiomyopathy. There were no significant differences in the frequencies of TTN truncating variants amongst control samples from different sites.


All studies were performed in accordance with institutional guidelines and with the approval of the local ethics committees. Subjects in DCM groups A and C, all HCM, and all control subjects provided written informed consent. Within the proband cohorts, no subjects had a known familial relationship.


Segregation Analyses:


For linkage studies, unknown affection status was assigned to family members of age≦40 years who lacked criteria for DCM and/or had confounding clinical diagnoses. Logarithm of the odds scores were calculated using FASTLINK (available on the world wide web at http://www.ncbi.nlm.nih.gov/CBBresearch/Schaffer/fastlink.html)13


Kaplan-Meier curves (FIG. 2D) of freedom from cardiac transplant, left ventricular assist device implantation, and death in male and female mutation carriers remained significantly different when family was used as a covariate (P=1.4×10−5).


DNA Sequencing:


Targets for filter-based hybridization capture were defined as TTN exons in the UCSC hg18 refGene table (NM003319, NM133437, NM133378, NM133432, NM133379) or the hg18 known Gene table (uc002 umr.1, uc002 ums.1, uc010frc.1, uc010frd.1, uc010fre.1, uc002 umz.1, uc002unb.1)+/−10 bp and genomic intervals in the UCSC hg18 phastConsElements28wayPlacMammal and hg18 phastConsElements28way tables that were within 2 kb of TTN and had conservation scores>=350. Target regions within 180 bp of one another were merged and primers were designed using EXONPRIMER™ (available on the world wide web at http://ihg.gsf.de/ihg/ExonPrimer.html) or PRIMER3™ (available on the world wide web at http://frodo.wi.mit.edu/primer3) with a 28 bp target gap, an optimal annealing temperature of 62° C. and an optimal primer length of 22-24 bp. Filter traps were generated as previously described14, except that PCR amplimers were not confirmed by dideoxy sequencing and all DNA cleanups were performed with AmpureXP beads (Agencourt). Target amplimers, including three autosomal, three X-chromosome, and three Y-chromosome amplimers that were added at one quarter the molarity of the TTN amplimers, as a control for capture and copy-number, are listed in Table 3.


Genomic DNA libraries were made from 0.5 to 3 μg of genomic DNA and captured largely as previously described14. However, most samples were sheared using a Covaris E200 in single tubes (10% duty cycle, 5 intensity, 200 cycles/burst for 13 m) with a sample volume of 120 μl and most subject libraries were barcoded with a 3 bp+‘T’ sequence (Table 4), pooled in groups of 10-21, and hybridized as a pool of 2 to 4 μg.


For dideoxy sequencing, ITN targets were defined as all coding exons in refseq transcripts NM133378, NM00319, and NM133379.


Next-Generation DNA Sequence Analysis:


For next-generation sequence data, short-read sequences and quality scores were generated with Illumina GENOME ANALYZER PIPELINE SOFTWARE™ v1.0 to v.1.80, mapped with NOVALIGN™ V2.07.05 (available on the world wide web at http://www.novocraft.com/), using full Needleman-Wunsch alignment, to the hg19 human reference genome. Sequence read base quality scores were recalibrated using the genome analysis toolkit (GATK) v1.0.441815. Sequence reads were realigned (using GATK) and duplicate masked with PICARD™ (available on the world wide web at http://picard.sourceforge.net/). Single-nucleotide variants (SNV) and small indels were detected using GATK UNIFIEDGENOTYPER™. Shorter single-end sequencing read lengths (32-46 bp) in 56 HCM subjects and 2 control subjects may have limited detection of insertions/deletions (especially those with sizes greater than 3 bp) in these subjects. Reported comparisons remain significant if these subjects or larger mutations are excluded (data not shown). The quality of identified nonsense, frameshift, and splicing variants was assessed using additional GATK tools and visual inspection using the INTEGRATIVE GENOMICS VIEWER™16. The quality of missense variants (excluding nonsense and frameshift variants) was assessed using automated GATK tools alone.


The effect of each variant on titin RNA and protein was predicted using transcript definitions from the UCSC hg19 refGene table, with slight adaptations (see below). To identify variants likely to affect splicing, for each variant within the splice-donor or splice-acceptor sites, the difference between the log 2(maximum-entropy) of the mutant allele and that of the wild-type allele was calculated using an existing maximum entropy model17. Twenty-seven variants were identified with a maximum-entropy difference less than −2. Twenty-three of these variants were seen either in a single subject or in subjects with the same diagnosis and were absent from the 1000 Genomes Project18, 19 Phase I data (629 samples; 20100804 genotypes) (1KG). However, variant c.40160-10A>G was identified in subjects with different diagnoses and present in 1KG, variant c.3100G>A was present in a subject with DCM and one with HCM, and variants c.10114+5G>A and c.30811+5G>A were present in 1KG. Excluding these four variants left 23 variants likely to substantially affect splicing (Table 9).


In all subjects studied by next-generation sequencing, the copy-number of ITN amplimers was assessed by visual inspection of heat maps and quantile-quantile plots of normalized median read depths (data not shown). TTN copy number heat maps were constructed using the log base-2 copy-ratio of a subject's target amplimer. Copy-ratios were iteratively median-normalized by amplimer and subject. The median read depths of each amplimer in each subject were median-normalized by both amplimer and sample. Newly identified variants were confirmed by the presence of sequence reads spanning likely DNA breakpoints and by breakpoint PCR (Table 10 and FIG. 7).


Forty-five TTN variants identified by next-generation sequencing in subjects with DCM from groups A and B, including 15 frameshifts, 17 nonsense, 12 splice, and one duplication, were assessed by independent dideoxy genomic sequencing and/or RNA sequencing and/or by restriction enzyme digest analyses (FIGS. 6A-6D and data not shown). All were confirmed (100% validation).


TTN Transcript Definitions Used For Variant Analyses:


The TTN consensus transcript sequence (Q8WZ42.nt) used to describe most variants was constructed to correspond to the protein sequence UniProtKB Q8WZ42 version 88. This transcript was defined as the combination of the UCSC hg19 alignments of RefSeq transcripts NM003319 (N2B) and NM133378 (N2A). The protein product predicted by the genomic sequence differs from Q8WZ42 at positions 17036-17043, because of a likely insertion of ‘C’ at position 51,102 of the cDNA sequence encoding Q8WZ42, and at several scattered single amino acids encoded by polymorphic nucleotides. The few exons (identified by black carets in FIG. 1 and defined in Table 17), with the least evidence for cardiac expression (Illumina Human Body Map 2.0 Project and unpublished) were excluded from analyses. Variants falling outside of this transcript are described using transcript definitions that include the corresponding alternatively spliced exon (novex-3, NM133379; novex-1, NM133432; novex-2, NM133437).


TTN Variants In dbSNP:


In dbSNP version 132, there are nine TTN nonsense SNPs (rs72646813, rs72646828, rs72646831, rs72646837, rs72646846, rs72648222, rs72648224, rs72648249, rs72648250) and two TTN frameshift variants (rs72647879, rs72648265) that were found by dideoxy sequencing of DCM subjects in group C (see the Methods, DNA Sequencing and Genotyping). For all other nonsense or frameshift TTN variants in dbSNP version 132, population frequencies are not provided and these have not been validated.


Previous studies have reported structural mutations within the M-band portion of titin in subjects with recessive, early-onset skeletal and cardiac myopathy or tibial muscular dystrophy (Table 16). Carmignac et al.,22 showed that truncated titin peptides lacking part of the M-band were incorporated into the sarcomere. No TTN truncating mutations were identified herein within the M-band portion in subjects with DCM, implying that M-band TTN truncating mutations may not cause isolated DCM. As such, in addition to assessing the uniformity of TTN truncating mutations in DCM subjects across all of titin, the spatial uniformity of mutations was assessed when excluding the portion of titin distal to the previously reported carboxy-terminal titin truncations that do not appear to cause dominant DCM. With this exclusion the associations remained significant (data not shown).


Results

Subject Characteristics:


Idiopathic DCM was diagnosed in 312 subjects from three independent cohorts: DCM-A, DCM-B, and DCM-C (Tables 11-13). HCM was diagnosed in 231 subjects; 249 subjects without known cardiomyopathy served as controls. Summary characteristics of each cohort are presented in Table 5.


TTN Genetic Variation

DNA Sequencing:


Using genomic DNA isolated from DCM-A, DCM-B, HCM, and control subjects, next-generation sequencing of 145 kb of TTN, including all annotated exons and splice sites was performed. For each subject, >97% of targeted bases were observed≧20 times (data not shown). From genomic DNA isolated from DCM-C subjects, TTN sequences were determined by traditional dideoxy sequencing. After excluding TTN variants with frequencies≧0.01 in the 1000 Genomes Project (available on the world wide web at www.1000genomes.org/data) or present in subjects (N=792) from all cohorts, 951 rare missense were identified, nonsense, frameshift, splicing or copy number TTN variants that are predicted to change the titin amino acid sequence (Tables 6, 7). Each subject in DCM, HCM or control cohorts had approximately one rare missense variant (range by cohort is 0.91-1.45 per subject.)


As the ITN transcript novex-3 (FIG. 1) is thought to not interact with the sarcomere M-band and is expressed in the heart approximately 20-fold less than full-length titin isoforms (data not shown), variants that exclusively altered novex-3 transcripts were not studied here. Analyses were prioritized for nonsense, frameshift, splicing, and copy-number variants, which unlike missense variants, are all predicted to have a profound effect on the structure of titin N2A or N2BA polypeptides (FIG. 1). These are denoted as TTN truncating variants.


TTN Variants in HCM and Control Subjects:


Among HCM subjects, three TTN truncating variants were found: two frameshift variants and one splicing variant (Supplementary Tables 8, 9). In each of these three subjects, concurrent analyses revealed a pathogenic mutation in well-established HCM gene MYH7 or MYBPC3 (data not shown). No family members were available for segregation analyses. In control subjects, two frameshift and five splicing variants were identified (FIG. 1 and Tables 8, 9). The frequency of TTN truncating variants did not differ significantly between subjects with hypertrophic cardiomyopathy and controls (1% and 3%, respectively; P=0.34).


Nonsense and Frameshift Variants in DCM Subjects:


Forty four nonsense or frameshift variants were identified that alter full-length titin in DCM subjects (Tables 1 and 8). Among cohorts studied by next-generation sequencing, subjects in cohort DCM-A (N=21; 23%) and DCM-B (N=14; 20%) were enriched for these variants as compared to HCM subjects (N=2; 0.9%; P=2×10−12) or control subjects (N=2; 0.08%; P=3×10−13). Twelve DCM-C subjects (8%) studied by traditional dideoxy sequencing had such variants. Strong co-segregation (LOD=9.3) of nonsense and frameshift variants was observed with clinical status among 60 members of 16 DCM families (FIG. 5, Tables 14, 15), indicating odds of ˜1 in 109 that the segregation of these TTN variants occurred by chance. Interestingly, subject MAO-92 who presented with DCM at age 17, had one rare variant on each TTN allele, a previously described pathogenic missense mutation9 and a nonsense variant inherited from her mother (FIG. 5). In all other families, there was co-inheritance of DCM and a single TTN frameshift or nonsense variant. The penetrance of TTN truncating mutations in available family members over the age of 40 (N=32) is >95%.


Splicing Variants in DCM Subjects:


Seventeen TTN variants were identified in DCM subjects that are predicted to alter RNA splicing, including 11 that altered absolutely conserved splice-site nucleotides (Tables 1 and 9). RNA sequencing of cardiac tissues from two subjects with splicing variants, MIV-14 (FIGS. 6A-6D) and MAM-12 (an adult offspring of a consanguineous marriage with a homozygous splicing variant; data not shown) confirmed aberrant TTN splicing. Among subjects studied by next-generation sequencing, significant enrichment for subjects with splicing variants in DCM-A (N=15; 16%) and DCM-B (N=3; 4%) was observed as compared to HCM (N=1; P=7×10−8) or to control (N=5; P=9×10−6) cohorts. Complete co-segregation of splicing variants and DCM (LOD=1.8 or 60 times more likely than a chance association) was found among 11 members of three families (FIG. 5, Table 15).


DCM Copy-number Variants:


Copy number was assessed across TTN in DCM-A and DCM-B subjects by comparing the distribution of sequence reads between individuals (data not shown). Traditional dideoxy sequencing of DCM-C subjects did not permit these analyses. A single copy-number variant (CNV) was identified in a DCM subject (MEQ-132). This tandem insertion of 28 kb (spanning introns 71 to 124) was predicted to incorporate a 13% internal duplication of a portion of titin and was confirmed in MEQ-132 and absent from two healthy relatives (FIG. 7). No TTN CNVs were observed among HCM or control subjects (N=480).


Consequences Of Ttn Truncating Variations:


Because TTN nonsense, frameshift, splicing and copy-number variants that are predicted to substantially alter titin structure, were significantly enriched among DCM subjects, compared to hypertrophic cardiomyopathy subjects (P=3×10−16) or controls (P=9×10−14), and were co-inherited with DCM in families, the data suggest that they cause DCM. Of note, six TTN mutations were each present in two subjects; analyses of one subject pair were consistent with a shared haplotype (data not shown).


To conservatively estimate the frequency of truncating TTN mutations in DCM, the total DCM-A population was increased by 40 additional subjects recruited concurrently with the subjects in cohort DCM-A whose TTN sequences were not analyzed because studies revealed a pathogenic mutation in another DCM gene. Accordingly, the frequencies of TTN truncating mutations in the DCM cohorts were 28% (DCM-A), 24% (DCM-B) and 9% (DCM-C). TTN mutation frequencies were not significantly different between subjects with and without a family history of DCM (Table 5; P=0.36).


Between the two different DNA-sequencing platforms used to analyze data for the subjects with dilated cardiomyopathy, more subjects in groups A and B than in group C were found to have TTN truncations (P<0.001). Clinical features among the dilated cardiomyopathy groups (ascertained during evaluation for cardiac transplantation) included more familial disease in group A than in group B (P<0.001) or group C(P=0.005) and more severe dilated cardiomyopathy among the subjects in group B, who had a significantly increased left ventricular end diastolic diameter as compared with group A (P=1×10−19) and group C(P=3×10−6), as well as an increased left ventricular ejection fraction (P=0.03 and P=0.001, respectively) (Table 2, and FIGS. 4A-4B).


TTN truncating mutations found in DCM subjects were non-randomly distributed within titin (FIG. 1). DCM mutations were overrepresented in the A-band region as compared to either the remainder of N2BA (P=0.0004) or N2B (P=0.01) and were notably absent from the Z-disk and M-band regions of titin (P=0.006, N2BA; P=0.001, N2B). The distribution of the 10 variants found in HCM and control subjects (FIG. 1) was distinct (P=0.001), in that it was less enriched for the A-band region of titin (40% versus 84%; P=0.006) and included Z-band variants (20% versus 0%; P=0.01).


Clinical Characteristics:


There were no significant differences (P>0.1) in ages at diagnosis, left ventricular end-diastolic dimensions, ejection fraction, or freedom from cardiac transplant, left ventricular assist device implantation, or death between subjects with and without TTN truncating mutations (Table 2, FIGS. 2A, 2B).


Subjects with TTN truncating mutations had DCM that usually was unaccompanied by conduction system or skeletal muscle disease (Tables 11, 12, 13), although an overt skeletal myopathy occurred in subject MAM-12 with a homozygous TTN splicing mutation. Cardiac histopathology from subjects with TTN truncating mutations was typical of idiopathic DCM. Of note, some sections revealed foci of myocytes with bizarre, stellate nuclear morphology, best appreciated in cross-section (FIGS. 3A-3B). Electron microscopy of one specimen obtained post-mortem, showed intact sarcomeric structures (FIG. 8).


Freedom from cardiac transplant, left ventricular assist device implantation, or death in subjects and family members with TTN mutations was influenced by gender (FIGS. 2C-2D). The mean age for these adverse events in mutation carriers within families (N=94; 19 families) was 68±5 years for women (N=33) and 56±3 years for men (N=61; FIGS. 2A-2D).


Discussion

TTN truncating variants were identified in 67 DCM subjects, three HCM subjects and seven control subjects (Tables 1, 2, 5, 6, 7, 8, and 9). TTN variants included nonsense and frameshifts that are predicted to cause protein truncation; variants of splice donor or acceptor sites that are predicted to cause exon skipping and/or to include intronic sequence or delete exonic sequence; and a large tandem insertion. Without out wishing to be bound by theory, it is possible that these mutant alleles produce shortened titin with abnormal properties that cause DCM.


There was a low frequency of TTN truncating variants in HCM subjects was not different than the frequency in controls. In addition, each HCM subject with a TTN variant also had a pathogenic mutation in an established HCM gene, suggesting that TTN truncations rarely, if ever, cause HCM.


The frequency of DCM subjects with TTN truncating mutations was significantly higher than that of HCM subjects (P=3×10−16) or control subjects (P=9×10−14). Amongst the 3 groups of subjects with dilated cardiomyopathy, the frequency of TTN truncating mutations in group A and group B, was comparable (P=0.7) and significantly different from group C(P=3×10−5) that was studied by a different DNA sequencing platform. Although distinguishing clinical features of dilated cardiomyopathy were identified in subjects in each group, the lower frequency of mutations identified in DCM-C subjects raises the possibility that the next-generation sequencing strategy provided better mutation detection than dideoxy sequencing.


TTN truncating mutations and DCM were co-inherited in families (combined LOD score=11.1; FIG. 5, Table 15). Segregation analyses of frameshift or nonsense mutations in 17 families (LOD=9.3) and splice site mutations in three families (LOD=1.8) confirmed the co-inheritance of each type of truncating mutation. Family studies also demonstrated that the penetrance of TTN truncating mutations was >95% for the studied subjects over the age of 40 years (FIG. 5, Table 14).


DCM subjects with and without TTN truncating mutations had similar clinical manifestations and comparable morbidity and mortality, but men with TTN mutations had adverse events at significantly earlier ages than women (P=4×10−5). Gender is reported to influence outcomes in heart failure caused by various etiologies35. However, that gender would substantially influence an autosomal monogenetic cause of heart failure is unexpected and warrants further study.


Mutations that significantly disrupt the structure of full-length titin can cause DCM by several mechanisms. RNA and protein surveillance pathways likely degrade some truncated titin peptides36. Decreased titin levels could limit sarcomere formation and might produce cardiac dysfunction and remodeling. Yet this is not the case for previously reported TTN mutations that delete only the M-band portion of titin14 (FIG. 1); immunohistochemical studies showed some of these truncated titin proteins integrate into the sarcomere and cause recessive, early-onset skeletal and cardiac myopathy and not dominant DCM. In addition, if more proximal TTN truncating mutations caused DCM via haploinsufficiency, the distribution of such mutations would be rather uniform across the susceptible portion of titin. In contrast, a biased mutation distribution was observed in DCM subjects that was distinct from that observed in non-DCM subjects (FIG. 1 and above herein). This unequal mutation distribution may indicate that truncated titin proteins found in DCM subjects, like previously studied carboxy-terminal titin truncations, integrate into the sarcomere and cause DCM by a dominant-negative mechanism.


If truncated titin proteins in DCM subjects incorporated into the sarcomere, they would likely be anchored at the Z-line and interact with the full complement of Z-disk factors (FIG. 1). However, these truncated titin proteins would not include the M-band residues that anchor titin to the middle of the sarcomere via myomesin, encode a kinase domain, and interact with many proteins37. The M-band portion of titin is implicated in sensing and modulating sarcomeric force12, 38-40. Without wishing to be bound by theory, it is suggested that loss of these interactions could lead to DCM by disturbing normal regulation of sarcomeric force.


The data presented herein indicate that TTN truncating mutations are the most common known genetic cause of DCM. Ongoing analyses of other classes of TTN variation (e.g., missense variants) may further expand the importance of TTN in DCM pathogenesis. Incorporation of next-generation sequence analyses of TTN into clinical genetic screens should substantially increase the detection of DCM mutations by approximately 50%, enabling earlier diagnosis and interventions to prevent disease progression. Further study of the functional consequences of TTN truncating mutations on myocardial physiology and myocyte signaling is warranted.


REFERENCES



  • 1. Morita H, Rehm H L, Menesses A, et al. Shared genetic causes of cardiac hypertrophy in children and adults. N Engl J Med 2008; 358(18):1899-908.

  • 2. Olivotto I, Girolami F, Ackerman M J, et al. Myofilament protein gene mutation screening and outcome of patients with hypertrophic cardiomyopathy. Mayo Clin Proc 2008; 83(6):630-8.

  • 3. Ahmad F, Seidman J G, Seidman C E. The genetic basis for cardiac remodeling. Annual review of genomics and human genetics 2005; 6:185-216.

  • 4. Dellefave L, Mcnally E M. The genetics of dilated cardiomyopathy. Current Opinion in Cardiology 2010; 25(3):198-204.

  • 5. Michels W, Moll P P, Miller F A, et al. The frequency of familial dilated cardiomyopathy in a series of patients with idiopathic dilated cardiomyopathy. N Engl J Med 1992; 326(2):77-82.

  • 6. Baig M, Goldman J, Caforio A, Coonar A, Keeling P, McKenna W. Familial dilated cardiomyopathy: cardiac abnormalities are common in asymptomatic relatives and may represent early disease. Journal of the American College of Cardiology 1998; 31(1):195.

  • 7. Mestroni L, Rocco C, Gregori D, et al. Familial dilated cardiomyopathy* 1:: Evidence for genetic and phenotypic heterogeneity. Journal of the American College of Cardiology 1999; 34(1):181-90.

  • 8. Zimmerman R S, Cox S, Lakdawala N K, et al. A novel custom resequencing array for dilated cardiomyopathy. Genet Med 2010; 12(5):268-78.

  • 9. Siu B L, Niimura H, Osborne J A, et al. Familial dilated cardiomyopathy locus maps to chromosome 2q31. Circulation 1999; 99(8):1022-6.

  • 10. Gerull B, Atherton J, Geupel A, et al. Identification of a novel frameshift mutation in the giant muscle filament titin in a large Australian family with dilated cardiomyopathy. J Mol Med 2006; 84(6):478-83.

  • 11. Gerull B, Gramlich M, Atherton J, et al. Mutations of TTN, encoding the giant muscle filament titin, cause familial dilated cardiomyopathy. Nat Genet 2002; 30(2):201-4.

  • 12. Lange S, Xiang F, Yakovenko A, et al. The kinase domain of titin controls muscle gene expression and protein turnover. Science 2005; 308(5728):1599-603.

  • 13. Hackman P, Vihola A, Haravuori H, et al. Tibial muscular dystrophy is a titinopathy caused by mutations in TTN, the gene encoding the giant skeletal-muscle protein titin. Am J Hum Genet 2002; 71(3):492-500.

  • 14. Carmignac V, Salih M A M, Quijano-Roy S, et al. C-terminal titin deletions cause a novel early-onset myopathy with fatal cardiomyopathy. Ann Neurol 2007; 61(4):340-51.

  • 15. Hackman P, Marchand S, Sarparanta J, et al. Truncating mutations in C-terminal titin may cause more severe tibial muscular dystrophy (TMD). Neuromuscular Disorders 2008; 18(12):922-8.

  • 16. Trinick J, Knight P, Whiting A. Purification and properties of native titin. J Mol Biol 1984; 180(2):331-56.

  • 17. Liversage A D, Holmes D, Knight P J, Tskhovrebova L, Trinick J. Titin and the sarcomere symmetry paradox. J Mol Biol 2001; 305(3):401-9.

  • 18. van der Ven P F, Bartsch J W, Gautel M, Jockusch H, Furst D O. A functional knock-out of titin results in defective myofibril assembly. J Cell Sci 2000; 113 (Pt 8):1405-14.

  • 19. Musa H, Meek S, Gautel M, Peddie D, Smith A J H, Peckham M. Targeted homozygous deletion of M-band titin in cardiomyocytes prevents sarcomere formation. J Cell Sci 2006; 119(Pt 20):432231.

  • 20. Granzier H L, Irving T C. Passive tension in cardiac muscle: contribution of collagen, titin, microtubules, and intermediate filaments. Biophysical Journal 1995; 68(3):1027-44.

  • 21. Horowits R, Kempner E S, Bisher M E, Podolsky R J. A physiological role for titin and nebulin in skeletal muscle. Nature 1986; 323(6084):160-4.

  • 22. Muhle-Goll C, Habeck M, Cazorla O, Nilges M, Labeit S, Granzier H. Structural and functional studies of titin's fn3 modules reveal conserved surface patterns and binding to myosin S1—a possible role in the Frank-Starling mechanism of the heart. J Mol Biol 2001; 313(2):431-47.

  • 23. Cazorla O, Wu Y, Irving T C, Granzier H. Titin-based modulation of calcium sensitivity of active tension in mouse skinned cardiac myocytes. Circulation Research 2001; 88(10):1028-35.

  • 24. Guo W, Bharmal S J, Esbona K, greater ML. Titin diversity—alternative splicing gone wild. J Biomed Biotechnol 2010; 2010:753675.

  • 25. Bang M L, Centner T, Formoff F, et al. The complete gene sequence of titin, expression of an unusual approximately 700-kDa titin isoform, and its interaction with obscurin identify a novel Z-line to I-band linking system. Circulation Research 2001; 89(11):1065-72.

  • 26. Herman D S, Hovingh G K, Iartchouk O, et al. Filter-based hybridization capture of subgenomes enables resequencing and copy-number detection. Nat Methods 2009:27.

  • 27. Mestroni L, Maisch B, McKenna W J, et al. Guidelines for the study of familial dilated cardiomyopathies. Collaborative Research Group of the European Human and Capital Mobility Project on Familial Dilated Cardiomyopathy. Eur Heart J 1999; 20(2):93-102.

  • 28. Maron B J, McKenna W J, Danielson G K, et al. American College of Cardiology/European Society of Cardiology clinical expert consensus document on hypertrophic cardiomyopathy. A report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus Documents and the European Society of Cardiology Committee for Practice Guidelines. Journal of the American College of Cardiology 2003; 42(9):1687-713.

  • 29. Bentley D R, Balasubramanian S, Swerdlow H P, et al. Accurate whole human genome sequencing using reversible terminator chemistry. Nature 2008; 456(7218):53-9.

  • 30. Mckenna A, Hanna M, Banks E, et al. The Genome Analysis Toolkit: A MapReduce framework for analyzing next-generation DNA sequencing data. Genome Research 2010; 20(9):1297-303.

  • 31. Team R D C. R: A Language and Environment for Statistical Computing. In: R Foundation for Statistical Computing; 2010.

  • 32. Gramlich M, Michely B, Krohne C, et al. Stress-induced dilated cardiomyopathy in a knock-in mouse model mimicking human titin-based disease. Journal of Molecular and Cellular Cardiology 2009; 47(3):352-8.

  • 33. Kontrogianni-Konstantopoulos A, Ackermann M A, Bowman A L, Yap S V, Bloch R J. Muscle Giants: Molecular Scaffolds in Sarcomerogenesis. Physiological Reviews 2009; 89(4):1217-67.

  • 34. Peng J, Raddatz K, Molkentin J D, et al. Cardiac hypertrophy and reduced contractility in hearts deficient in the titin kinase region. Circulation 2007; 115(6):743-51.

  • 35. Willis M S, Ike C, Li L, Wang D-Z, Glass D J, Patterson C. Muscle ring finger 1, but not muscle ring finger 2, regulates cardiac hypertrophy in vivo. Circulation Research 2007; 100(4):456-9.

  • 36. Grater F, Shen J, Jiang H, Gautel M, Grubmiiller H. Mechanically induced titin kinase activation studied by force-probe molecular dynamics simulations. Biophysical Journal 2005; 88(2):790-804.

  • 37. Noda S. Histopathology of endomyocardial biopsies from patients with idiopathic cardiomyopathy; quantitative evaluation based on multivariate statistical analysis. Jpn Circ J 1980; 44(2):95-116.

  • 38. Ferrans V J, Jones M, Maron B J, Roberts W C. The nuclear membranes in hypertrophied human cardiac muscle cells. Am J Pathol 1975; 78(3):427-60.

  • 39. Nikolova V, Leimena C, McMahon A C, et al. Defects in nuclear structure and function promote dilated cardiomyopathy in lamin A/C-deficient mice. J Clin Invest 2004; 113(3):357-69.

  • 40. Granzier H, Labeit S. The Giant Protein Titin A Major Player in Myocardial Mechanics, Signaling, and Disease. Circ Res 2004.

  • 41. Lakdawala NK, Dellefave L, Redwood C S, et al. Familial dilated cardiomyopathy caused by an alpha-tropomyosin mutation: the distinctive natural history of sarcomeric dilated cardiomyopathy. Journal of the American College of Cardiology 2010; 55(4):320-9.

  • 42. Schonberger J, Wang L, Shin J T, et al. Mutation in the transcriptional coactivator EYA4 causes dilated cardiomyopathy and sensorineural hearing loss. Nat Genet 2005; 37(4):418-22.

  • 43. Schmitt J P, Kamisago M, Asahi M, et al. Dilated cardiomyopathy and heart failure caused by a mutation in phospholamban. Science 2003; 299(5611):1410-3.

  • 44. Gerull B, Gramlich M, Atherton J, et al. Mutations of TTN, encoding the giant muscle filament titin, cause familial dilated cardiomyopathy. Nat Genet 2002; 30(2):201-4.

  • 45. Kamisago M, Sharma S D, DePalma S R, et al. Mutations in sarcomere protein genes as a cause of dilated cardiomyopathy. N Engl J Med 2000; 343(23):1688-96.

  • 46. Fatkin D, MacRae C, Sasaki T, et al. Missense mutations in the rod domain of the lamin A/C gene as causes of dilated cardiomyopathy and conduction-system disease. N Engl J Med 1999; 341(23):1715-24.

  • 47. Taylor M R G, Slavov D, Ku L, et al. Prevalence of desmin mutations in dilated cardiomyopathy. Circulation 2007; 115(10):1244-51.

  • 48. Taylor M R G, Slavov D, Gajewski A, et al. Thymopoietin (lamina-associated polypeptide 2) gene mutation associated with dilated cardiomyopathy. Hum Mutat 2005; 26(6):566-74.

  • 49. Carniel E, Taylor M R G, Sinagra G, et al. Alpha-myosin heavy chain: a sarcomeric gene associated with dilated and hypertrophic phenotypes of cardiomyopathy. Circulation 2005; 112(1):54-9.

  • 50. Taylor M R G, Fain P R, Sinagra G, et al. Natural history of dilated cardiomyopathy due to lamin A/C gene mutations. JAC 2003; 41(5):771-80.

  • 51. Lathrop G, Lalouel J. Easy calculations of lod scores and genetic risks on small computers. Am J Hum Genet 1984; 36(2):460-5.

  • 52. Gramlich M, Michely B, Krohne C, et al. Stress-induced dilated cardiomyopathy in a knock-in mouse model mimicking human titin-based disease. Journal of Molecular and Cellular Cardiology 2009; 47(3):352-8.

  • 53. Fujita P, Rhead B, Zweig A, et al. The UCSC Genome Browser database: update 2011. Nucleic Acids Res 2011; 39(Database issue):D876-82.

  • 54. Herman D S, Hovingh G K, Iartchouk O, et al. Filter-based hybridization capture of subgenomes enables resequencing and copy-number detection. Nat Methods 2009:27.

  • 55. Mckenna A, Hanna M, Banks E, et al. The Genome Analysis Toolkit: A MapReduce framework for analyzing next-generation DNA sequencing data. Genome Research 2010; 20(9):1297-303.

  • 56. Robinson J T, Thorvaldsdottir H, Winckler W, et al. Integrative genomics viewer. Nature Biotechnology 2011; 29(1):24-6.

  • 57. Yeo G, Burge C B. Maximum entropy modeling of short sequence motifs with applications to RNA splicing signals. J Comput Biol 2004; 11(2-3):377-94.

  • 58. Consortium TGP, Author C, Committee S, et al. A map of human genome variation from population-scale sequencing. Nature 2010; 467(7319):1061-73.

  • 59. Carmignac V, Salih M A M, Quijano-Roy S, et al. C-terminal titin deletions cause a novel early-onset myopathy with fatal cardiomyopathy. Ann Neurol 2007; 61(4):34051.

  • 60. Hackman P, Marchand S, Sarparanta J, et al. Truncating mutations in C-terminal titin may cause more severe tibial muscular dystrophy (TMD). Neuromuscular Disorders 2008; 18(12):922-8.

  • 61. Zimmerman R S, Cox S, Lakdawala N K, et al. A novel custom resequencing array for dilated cardiomyopathy. Genet Med 2010; 12(5):268-78.










TABLE 1







DCM, HCM and Control Subjects with TTN Truncating Variants*












Mutation
DCM
HCM
Control




class
(N = 312)
(N = 231)
(N = 249)
p-value
p-value{circumflex over ( )}





Nonsense
28{circumflex over ( )}
0
0
  1 × 10−10

5 × 10−11



Frameshift
19
2
2
0.0004
1 × 10−5


Splicing
19{circumflex over ( )}
1
5
0.001 
4 × 10−6


Copy-
 1
0
0
NA
NA


number


All
67
3
7
<2 × 10−16
<2 × 10−16


truncating










Comparison of the fraction of DCM, HCM, and control subjects with TTN truncation mutations. ̂ Three nonsense and three splicing mutations each occurred in two subjects. One DCM subject carried two different splicing mutations. P-values are calculated excluding the one copy-number mutation and including all subjects (†) or including only subjects studied by next-generation sequencing (̂) plus 40 DCM subjects who were excluded from TTN sequencing for a total of 352 DCM subjects (detailed in text, see CONSEQUENCES OF TTN TRUNCATING VARIATIONS).









TABLE 2







Characteristics of DCM cohorts











DCM-A
DCM-B
DCM-C



TTN truncating
TTN truncating
TTN truncating



mutation
mutation
mutation














Yes
No
Yes
No
Yes
No



N = 37
N = 55
N = 17
N = 54
N = 13
N = 136

















Female, no.
11 (30)
17 (31)
2 (12)
14 (26)
4 (31)
56 (41)


(%)


Familial DCM
92%
79%
21%
24%
85%
66%


(%)


Age (yrs)
45.9 ± 13.6
46.7 ± 14.3
48.4 ± 11.0
51.7 ± 12.8
49.2 ± 12.8
49.7 ± 14.0


Age at
37.1 ± 13.2
37.3 ± 14.6
38.5 ± 9.8 
41.3 ± 13.4
37.6 ± 14.7
39.6 ± 12.3


diagnosis (yrs)


LVEF (%)
26.4 ± 10.8
30.7 ± 11.6
24.6 ± 11.0
24.8 ± 11.7
29.3 ± 8.5 
31.4 ± 11.8


FS (%)
19.6 ± 9.90
19.6 ± 10.2
NA
NA
13.5 ± 3.4 
16.6 ± 6.9 


LVEDD (mm)
63.7 ± 10.9
61.4 ± 9.7 
70.8 ± 8.1 
74.2 ± 10.2
65.5 ± 9.2 
65.6 ± 10.4


LVEDD/BSA
33.0 ± 5.1 
NA
37.6 ± 6.0 
NA
34.0 ± 7.1 
NA


(mm/cm2)


NYHA III-IV
62%
41%
94%
91%
46%
43%


(%)


ICD, no. (%)
22 (59)
30 (55)
1 (6) 
2 (4)
 3 (100)
NA


Tx/VAD/Dead
14 (38)
17 (31)
17 (100)
47 (87)
1 (8) 
31(23)


no. (%)





*Data are provided as means ± SD, standard deviation.


No., numbers of subjects,


NA not available.







Measurements are from echocardiograms, including LVEF, left ventricular ejection fraction; FS, fractional shortening; LVEDD, left ventricular end-diastolic diameter and LVEDD divided by BSA, body surface area.


NYHA III-IV: Percentage of subjects with New York Heart Association functional class III or IV.


ICD: Number of subjects who with an implantable cardioverter defibrillator


Tx, VAD, death: Numbers of subjects with a cardiac transplant (Tx), ventricular assist device (VAD) or had a cardiac death.









TABLE 3 







TTN and Control amplimer PCR primers*
















SEQ

SEQ







ID

ID





Amplimer
Forward primer
NO
Reverse primer
NO
Chr
Start
End

















TTN_ 1#1
TTTGAACTTGAGTTGCCTTATC
004
GATGTTTGCATTTACCCTCCTG
0342
chr2
179390615
179391238



TATC











TTN_3#7
ACCTCAGGCGTTCCACTTGTA
005
ACCAGAATTTACCCTGCCTCTC
0343
chr2
179397970
179398571



G











TTN_7#4
TGTATTTTCCAGCATCGTACCG
006
GAGCAATAATGCCTTTGTGAGT
0344
chr2
17940862
1794093





C


1
31





TTN_10#4
GGTTCACCAACTCCATTTTCTA
007
CTTGGCTGGAGTAAGCCTGTC
0345
chr2
17941786
1794184



TTG




5
75





TTN_13#5
CAAGCATGGTAGATTTTGTGG
008
GAGTGCCAGTAATTGCAAGGG
0346
chr2
17942634
1794270



C




2
07





TTN_13#16
GGAAATGGCTGTGGCAAATG
009
CCAAAGATCTGGTAATTGAGC
0347
chr2
17943285
1794334





C


0
87





TTN_13#27
GCCAGCTAATTGTCATTGAATC
010
GTCAAG GAG CAGACAATG CTT
0348
chr2
17943931
1794399



C

C


7
89





TTN_18#1
TCCGAGATTTAAATGTGCCTCC
011
TGGAGAAGTGTGATGTAAGCC
0349
chr2
17944893
1794495





G


6
78





TTN_ 1#2
TGGCCGTTACACTTTGCTCTG
012
GATCCATTTAAGAGGGCCTGT
0350
chr2
17939112
1793917





G


8
49





TTN_3#8
TTACTCCAAACCGGACATTTTC
013
AG CACCCATGGTTGAAG CAG
0351
chr2
17939850
1793991








4
36





TTN_8#1
GATTGTGTGTTGGAAAATTATC
014
GAATGCCCAACCCTATCCTATG
0352
chr2
17941004
1794106



TGTC




1
82





TTN_10#5
TCTTCCTCTCCTTGTCTTATCTC
015
TCTTGGAAAGAATGCGAACAT
0353
chr2
17941839
1794190



G

C


6
27





TTN_13#6
CATTGAAAGTATGGAAAGGAA
016
TCCAAAACAGCTTTTGTTAACG
0354
chr2
17942693
1794275



GCTC

TC


9
99





TTN_13#17
ATCTTGGCCAACCTGTACACTG
017
CTGTGAGAGTTCTGGACACGC
0355
chr2
17943341
1794340





C


7
75





TTN_13#28
CTTTGGCAATGACCAGTTTCTG
018
AAAGTGGCTTTGTGAACGTCA
0356
chr2
17943992
1794405





G


0
88


TTN_18#2
CTTTCCAACCCTGCAGGAAG
019
TGCACCAGTATAGCTCTCCACC
0357
chr2
17944949
1794501








9
49


TTN_1#3
TTCAGAAAGATTAGTCCGTGTG
020
TCCAGCAGCTTTATGGGAATAT
0358
chr2
17939166
1793922



AAAC

C


4
51


TTN_3#10
CAAAAGCACTTGTGTTAATGCG
021
CGAGATTCTGTCAACTTAACAT
0359
chr2
17939961
1794002





GG


4
35


TTN_8#2
TACTGCCCTCACTCGGAATATG
022
GAACTGGATGCCCGATTACAC
0360
chr2
17941060
1794112








8
15


TTN_11#1
AAAGCATATGCACAGGTTAGC
023
GGTGCTGAATACAGACCACTG
0361
chr2
17941910
1794195



G

TC


4
88


TTN_13#7
TATGGTCAAATTCACAGGGGC
024
GTGCGTGTGATGCCTTGTATC
0362
chr2
17942753
1794281








4
71


TTN_13#18
TGTGACTTTCAGGTTAACAGGT
025
CTTCTGCTTACCAAAAGGCTTG
0363
chr2
17943402
1794346



GG




6
81


TTN_13#29
ATGTCTGTAGGCCGCAGGTTG
026
TTGTTTCTTTCTCAGATCCTCC
0364
chr2
17944051
1794411





AG


9
58


TTN_19
CATGTGTTTGAAAGCCACTGTT
027
TTCTATGTACATTGGAGCAAAT
0365
chr2
17945109
1794516



C

CC


3
40


TTN_1#4
CCTCTACCAGTAATTTTATTGC
028
TG CCGTGTAACATTTATTTGCA 
0366
chr2
17939214
1793927



TCACC

G


7
88


TTN_3#11
TTTGCTGCCACCATCAGAGG
029
GATGCCACAGTTTACCAAGTCA
0367
chr2
17940018
1794007





G


2
84


TTN_8#3
TCAGAACCTGCTCTGATGGTAA
030
CGAAGCTTCAAGACCTATAATG
0368
chr2
17941116
1794117



C

GC


3
67


TTN_11#2
GTGGGCCTGGTTTGTCTATCA
031
TG CATAGAATTATAATGATGGT 
0369
chr2
17941946
1794200



G

GTGTG


5
11


TTN_13#8
AGTGTCCGTCACTTTTGGATTG
032
TCAGCAATGTTGGTGGTACAA
0370
chr2
17942811
1794288





AG


3
07


TTN_13#19
TGGTTATGTCTATGACTTTGGG
033
TCAAGGTTCTTGACAGACCAG
0371
chr2
17943460
1794352



G

G


4
63


TTN_13#30
TGGTAGTATCAGTGACATGCG
034
CATGTGGCAACCAAACTTATCC
0372
chr2
17944109
1794417



G




9
67


TTN_20#1
TTTTAGTATGGATTTGCTCCAA
035
GGATGCCCTATTCGTCTCTTTG
0373
chr2
17945160
1794524



TG




8
71


TTN_2#1
GTTTTCTGTGCTTGAAAGAGA
036
CAGCGATCAGACCCTAACCAT
0374
chr2
17939292
1793935



GG

C


0
07


TTN_4#2
GCTGGACTTTGTATTTCCCAGC
037
TGTGCTCATTGAATCCCTATTT
0375
chr2
17940212
1794028





ACC


0
34


TTN_8#5
GCGCTTGACACTGGAATTGAC
038
GTTGAAGCTGGTCACACCAAA
0376
chr2
17941225
1794128





C


8
72


TTN_12#1
GGAAAAGGTATGCGGAAATAC
039
GCTGGAGTTGGAAAACCTAGC
0377
chr2
17942151
1794220



TG

C


0
37


TTN_13#9
CTCCAGTAACTTTCAGAGGCC
040
CAGAATGCTTTGTTGCTCGTG
0378
chr2
17942871
1794293



C




5
74


TTN_13#20
TATGATGTCACTCCCACCATCC
041
GGCAAGCCGAGTAAAGTATCA
0379
chr2
17943514
1794358





G


5
85


TTN_13#31
CAGGTTCTCCTTTGCCATAGTG
042
TTGCAAGGATGAATACGGTAG
0380
chr2
17944168
1794423





G


6
45


TTN_20#2
GGTAGAATTGGGGATGACAAG
043
AG CAACTCTTAATCTCAAGCAC
0381
chr2
17945232
1794532



G

AG


5
34


TTN_2#2
TTGCTTATGCAGGTGAGGATT
044
CAAAAGTGGAACTGAGAAAGT
0382
chr2
17939343
1793941



C

ATTAGG


9
00


TTN_5#1
CCAGGGTTCTACTTAGTATAGA
045
CACAGAATGATGTTGGCCTGA
0383
chr2
17940318
1794037



GGGG

G


2
41


TTN_8#6
CATTCTTGGCCTTCACACGG
046

0384
chr2
17941278
1794134





ACCCTCACTGTGGAAAACTGC


2
15


TTN_12#2
CATCAATTGCCAAGACTGGTTC
047
GTTGCAAGAACCATGATGAAA
0385
chr2
17942198
1794225





G


6
07


TTN_13#10
CATTTCAGTGTGACATTGTTTC
048
TCGTTTTAGAAAAGCCTGGACC
0386
chr2
17942928
1794299



TTG




7
56


TTN_13#21
ACCGTCATAGGTGGGTTTCTTC
049
TATCGTTCTTGACAAACCAGGG
0387
chr2
17943575
1794364








7
50


TTN_14#1
ACATGTTCAACTGTTCTCAGGG
050
TGGCACCAAGGAAGGAACTAT
0388
chr2
17944326
1794438





C


0
70


TTN_21#1
TTTATCGAATACTTCTGTGCTT
051

0389
chr2
17945319
1794538



GAG

AG CCTGGTCCTGTGAGAAATC


7
61


TTN_3#2

052

0390
chr2
17939520
1793958



TAAAGGCTTGCCCCATAAATTG

CACCAAGTGACCACCACAAAG


4
19


TTN_5#2
GGATCTTTGCAAACAACTGGTT
053
 GGTGGTAAAAGGCCTCAAAGA
0391
chr2
17940367
1794042



C

G


8
27


TTN_8#7
ACCACTGTTGTTTTCCACAGTA
054
GAAAATGCTGCAGGAGTTG GA
0392
chr2
17941334
1794139



AG

C


4
61


TTN_12#4
ACTTTGGGAAGTGGTTTTCCA
055
TGGTTCAGAAGCAAAATGGG
0393
chr2
17942285
1794235



G




4
22


TTN_13#11
GAGGTTCCCAAGATATGACTA
056
TGAAAGCTGCTGAACCTCCTT
0394
 chr2
17942987
1794305



CAAAG




0
35


TTN_13#22
CTTCCACCATCATACTTGGGTG
057
AAGCATCAGAACGACCTCTTCC
0395
chr2
17943634
1794370








3
24


TTN_14#2 
AAATTTGATTGGTCCAGTGGG
058
GAATATACCTTCAGAGTGAGT 
0396
chr2
17944379
1794443





GCTGAG


8
72


TTN_21#2 
TCTGGTGGATCCCAGCAAAC
059
TAAAGTGGCGGAGGCCTGAC
0397
chr2
17945378
1794543








6
92


TTN_3#3
ATCGGAAGCCTGGACTGAAGA
060
AGTCCTCACCCTGGAAATTCTG
0398
chr2
17939575
1793963



G




7
57


TTN_5#4
GTCTGATGACGCCACCTTGC
061
GGTGGCACAGCTAATTTTCAAC
0399
chr2
17940462
1794051








2
62


TTN_8#8
GACCACTGTGGGAGGACCTG
062
TTGTGCTCCCATAACCCACTAC
0400
chr2
17941386
1794145








9
05


TTN_13#1
GAATAGTTTGGGGTGTGAAGG
063
CCCAGTGATAGCTCTGACCCT
0401
chr2
17942390
1794246



G

C


8
53


TTN_13#12
ATAAGTGATGCACTGGTCTGG
064
GTGAGGCTACATCAGTTCCTG
0402
chr2
17943046
1794311



G

G


0
44


TTN_13#23
CTAGAATTCGGCTGCCTCCATC
065
TGGAACTCCAAAGGCTGAAGA 
0403
chr2
17943691
1794376





C


1
17


TTN_14#3
AACATCATCCCTTGCCACAAC
066
CCCATTATTGATGGCGGAAG
0404
chr2
17944428
1794448








9
82


TTN_21#3
ATGACAGATTTGGACTGCCAC
067
GGCAGACCTGAACCAGACATA
0405
chr2
17945434
1794549



C

AC


5
48


TTN_3#4
CGGTAGGTTCCACTGTCATCA
068
AAATCAAGGCGACAAAGAGAA
0406
chr2
17939630
1793969



G

G


4
38


TTN_7#1
GGTTTAGAAACCTGAGAAAAG
069
ACAATGCCTCAGAAGACCATC
0407
chr2
17940682
1794076



GAGG

C


7
53


TTN_9

070
TCCATGGATGATTAAGACCTG
0408
chr2
17941560
1794160



TACCATTTTACAGGCCAGGGG

G


8
52


TTN_13#2
CAATCAAGGTCTTCCTCATTTC
071
GGCACTAGATCCATTTACAGTT
0409
chr2
17942456
1794252



AC

CC


3
30


TTN_13#13
CAAGGTCAAGTTCAGGTGCTT
072

0410
chr2
17943107
1794317



C

AATCCATTTGTGCTTCCTGGAC


6
15


TTN_13#24
GTAACAACTTTGCGCAGGTCA
073

0411
chr2
17943754
1794382



G

TTGCAGTGAATCCTTATGGACC


3
15


TTN_14#4
ACCAGGATTTCCTCTCTGCATC
074
ATTTTAGGAGTGACAAGGCCA
0412
chr2
17944481
1794454





G


0
19


TTN_21#4
CTCGGACCAATACTTTGCCTTC
075
GGAACGTCGAGAAGTAACTGG
0413
chr2
17945489
1794555





C


4
28


TTN_3#5
TAGACGCAGATGAGGATGATT
076
AAATGGCGTATTCCTAAACTGT
0414
chr2
17939683
1793974



C

CC


9
90


TTN_7#2
CAACTGTGACACGCTCTGATTC
077
TATGGCATTGGAGAACCTTGT
0415
chr2
17940751
1794081





G


5
47


TTN_10#2
TGCTGTAACAAGTAATTTCTCC
078
AAAGGTGAACAGACGTGGTCC
0416
chr2
17941684
1794174



TCC




3
13


TTN_13#3
GAAGTAATTTCCAAAGACGTG
079
CCACTGGGCCTATTATAGTTAA
0417
chr2
17942517
1794258



GG

AGATG


7
34


TTN_13#14
ACAGACGGTCATGGAGTCTTT
080
AG GATGAGGTTGAACTCCCAA
0418
chr2
17943164
1794323



G

G


1
05


TTN_13#25
CCAGCAGACAACCATCGAATC
081
GCTATCACTTGCAGGGATGAT
0419
chr2
17943813
1794388





G


0
10


TTN_15#1
TCTTGGAGTTACCAGCTCTACA
082
CTCACCCATCACTGGCTATTTG
0420
chr2
17944610
1794468



CC




6
44


TTN_21#5
TGAACTTCAGGTCAGCGATAG
083
TGAGCACTACACAGTTGAAAC
0421
chr2
17945546
1794560



G

AGAC


1
86


TTN_3#6
GGTCGGTAGTAAAAGTCATAA
084
GAGTCACAGCCACTAACACAG
0422
chr2
17939739
1793980



TCAGG

C


3
38


TTN_7#3
TCGACCACTTCTAGCTTTGCAG
085
AAGACCACCTCCTGTCATAACG
0423
chr2
17940806
1794087








2
51


TTN_10#3
GGCTAACCTCAATTTCACATGT
086
CACCATTCCTCAAGTTACTCGC
0424
chr2
17941735
1794179



CTTAC




3
36


TTN_13#4
AACGTCTCGGAACTTGACATC
087
TCAGCTGTTGTTGCAGAGTATC
0425
chr2
17942576
1794264



C

C


1
29


TTN_13#15
TCAAGTCTGAATGTTTCTCCAG
088
CTACAAAGAACCAGGCCCTCC
0426
chr2
17943221
1794328



C




8
98


TTN_13#26
GCTTCACCTGCTTTTAATATAA
089
TGCCAACTATCCATTTAAGGTT
0427
chr2
17943872
1794394



CCG

CC


2
02


TTN_16
TGAATGTCTTCTCCCACATTAT
090
GAAGCAGCATATGAATTTCCCA
0428
chr2
17944757
1794480



TC

C


6
44


TTN_21#6 
GCCCAGGAACATCAAGAACAG
091
TTGGATCCTCTCCGTAAGTTGC
0429
chr2
17945593
1794567








2
17


TTN_21#7
CTAGGGAGACTTCAGTCTTGT
092
TGAAGATGGTGGAATTTATTCT
0430
chr2
17945655
1794571



CAAC

TTAAC


7
85


TTN_29#4
TGGTTCATTCCATTTCACTAGC
093
CCACAAATCATGTCAGAG GAG
0431
chr2
17947312
1794737



ATAC

C


8
32


TTN_34#1
TTGTATGGCATCCCAACCTTC
094
CACTGCAAACTCATTAACTTTT
0432
chr2
17948113
1794817





CTTC


0
73


TTN_44#1
GCTGCTTTCATGCAATATAACA
095
AGGAACCCAGGAAATCACAGG
0433
chr2
17949676
1794974



CTTAG




7
07


TTN_54
GAGCAAGGAGTCAGGTAAAGG
096
CAAAGCTTGGTTTTGATTCTTG
0434
chr2
17950921
1795094



G

G


5
35


TTN_85#1
GGAGATGAACAAAAGGATGGG
097
GTGATCACAGAATATTTGCCTT
0435
chr2
17954227
1795427





TC


2
40


TTN_109
CCTTCCTTCACCCTCCACTG
098
CCTGAAAGCATTTTAGTTACTT
0436
chr2
17956348
1795637





ACACC


5
09


TTN_121#2
CTACAATTGTTGCAGGCTCTG
099
TTATCACTAGTTTGATTCCCGG
0437
chr2
17957888
1795793



G

C


5
94


TTN_21#9
TCATCTCCAACTTTCTGGTACT
0100
TGAAAGCCAAAGATCGTTTCA
0438
chr2
17945767
1794583



CAAC

G


2
15


TTN_30#1 
TGCTGAAGCTATGTCCCATTTC
0101
GAGTTGCCACTTTCCCATTTTG
0439
chr2
17947383
1794743








2
93


TTN_34#2
GGCCTTGGATAGCCTGTACTT
0102
TTTTAGAAACCCAGACTGTG CC
0440
chr2
17948164
1794823



G

TAC


1
80


TTN_44#2
AAAAGCAGCTGACTTGATCAC
0103
GAGGGGTTGCTTGGTTGTGTA
0441
chr2
17949732
1794978



C

G


0
84


TTN_56
CCACAGTTGACATGAGAGAAA
0104
TGCATCAATGTGAAATTTGTAT
0442
chr2
17951102
1795113



CAG

GAG


5
67


TTN_88#1
CTCACCAAGTTATGCTGCATG
0105
AGTGGTGCCAGTGATACCAGT
0443
chr2
17954400
1795446



G

C


1
46


TTN_110-
AG GG CCAATGCGTTGTATTAA
0106
TTCTGCCATTAGATATGCCTGA
0444
chr2
17956576



111
G

C


0
30


TTN_122
GATAAGACTGGGCTGGGGTG
0107
TCTCTCTGATGGAGTGGAAGA
0445
chr2
17957962
1795800





TTG


6
77


TTN_23
TCTCATGATGTGAAATGGTCAG
0108
CAAAGCTATGCTGTATGCTGT
0446
chr2
17946175
1794622



C

GTAAC


0
05


TTN_30#2
GGGGCATCTATAGTGATCATA
0109
TTTTGTTTTAAGAAGGTGGTTT
0447
chr2
17947429
1794747



ACC

TCC


1
68


TTN_35#2
TCAACTCCTCCTTTCTGTAGAC
0110
GCATAATAACATGATGTAGCTT
0448
chr2
17948300
1794836



CAG

GGC


5
50


TTN_44#3
TGTTTGGAGTGAGGGTTAGAA
0111
GACAAAACTGCAGTGGAGAAG
0449
chr2
17949777
1794983



GG

G


3
71


TTN_60
ATGCAACAACAATGAGGACAA
0112

0450
chr2
17951419
1795147



C

ACTTTGGACCCACAGAATTTGG


2
45


TTN_88#2
AGTAAGCAATCATTGGTGCTG
0113

0451
chr2
17954451
1795452



C

TGAAGCTCAGAATCCTTTCCAC


5
32


TTN_112
CTAGCATCAGCTGAGTGAGAC
0114
CCCAGACGATGAAGGTGATTT
0452
chr2
17956654
1795671



C

C


3
94


TTN_123
AAGAATCAATCTTCCACTCCAT
0115
CATTTAACTCTTCTTCCTTGAA
0453
chr2
17958004
1795806



CAG

CTGC


8
45


TTN_24
TTGTTACACAGCATACAGCATA
0116
GGAAGCCATTAAAGCAAGAAG
0454
chr2
17946217
1794628



GC

C


8
53


TTN_30#3
ACATCTACAGGTGGATCAGGG
0117
AGCCCCACTTAGAGACAAGTG
0455
chr2
17947468
1794751



G

C


9
95


TTN_36#1
GAAATGAAAGACCCTCACAAG
0118
TTGCAAGCTAACTGATAATCCA
0456
chr2
17948424
1794848



G

ATG


6
74


TTN_44#4
TCACCATCTTTGAACCATTTCA
0119
AATG GGTCCCTTTTACTCAG GC
0457
chr2
17949828
1794988



C




6
99


TTN_61
TTCTGTGGGTCCAAAGTTTTAT
0120
CAAAGAAGTCAGTGCAAATGA
0458
chr2
17951472
1795151



G

GATAG


9
64


TTN_89#1
GGCTAACAGGCTATGGATGTA
0121
AAGTGCTGAAGAAAGCTGTCC
0459
chr2
17954559
1795461



TTTAG

C


9
80


TTN_113
TGAGCGCATACAGGGTAGAAA
0122
GCTACATTCGACCAGCATGAAT
0460
chr2
17956710
1795676



TAC

AC


1
08


TTN_124#1
CTCTCTCTCAAGCACACCCACC
0123
GTGTCTTGGTACAAGGATGGG
0461
chr2
17958173
1795824





G


8
30


TTN_25#1
ACAGAAGTTAATGGGATTGAG
0124
AGAGCCAGAATATGATG GAG G
0462
chr2
17946315
1794637



AATAAC

C


1
19


TTN_31#1
GCACTGCAAAGTTAACTAATTT
0125
GATGCTGGGAGAAAACACATT
0463
chr2
17947565
1794763



CCTC

G


2
69


TTN_36#2
ACTCATCATCCAGCCTGCAATC
0126
GAAGACCTTAGGATTGTTGAG
0464
chr2
17948477
1794853





CC


2
50


TTN_47#1
TGTAATACTGGGAAACGAGGT
0127
AAGACCTCCACGGCTAAACTT
0465
chr2
17950060
1795011



CC

G


7
55


TTN_72
AAAG GTG GTCCTTTCTATCG CC
0128
GTGGAACAAAGGGAGGATGGA
0466
chr2
17952912
1795297





G


4
75


TTN_91
G GAACTAGAAG GCAAAGAG CC
0129
TGTCTTTGTCCTTGAATTGTTT
0467
chr2

1795481



AG

G


3
11


TTN_114#1
CACAACATTTGCCATTGACC
0130
CAACTTAAAGACCAGGGCAAT
0468
chr2
17956881
1795693





TATC


5
09


TTN_124#2
ATACTGCCCTATGTGGCTCTG
0131
GCCTGATAGAAAATGAGGCTG
0469
chr2
17958231
1795831



G

G


1
05


TTN_25#3
CCATCATCTTTAGGTGGAAACC
0132
TCTGCCTACAGTGGATCCAAA
0470
chr2
17946408
1794646





C


2
80


TTN_31#2
TCCATTTCTCAGTGCCTACTGG
0133
TATTTTCAGCCACTCCTGGACC
0471
chr2
17947625
1794768








8
98


TTN_36#4
TTTTCTGAAAGCAACCGACAAG
0134
CGGAAGAGAATCCTGGTCATT
0472
chr2
17948571
1794863





C


7
04


TTN_47#2
TGCAAGTTGCTACTAAGGTTTG
0135
TGCTTTACGGCTGGTTTTATCC
0473
chr2
17950105
1795016



TTAC




6
22


TTN_74
TTGGTTGAGCTTCTACTTGGG
0136
CCAAGAAAGTCAAGTCCCACA
0474
chr2
17953140
1795318



G

C


7
02


TTN_94
CCACCAACATATAAACAGTATG
0137
AAAAGTGAATGCTAAGCCCCA
0475
chr2
17954929
1795497



ACCC

C


5
96


TTN_114#2
TAGCGCTAGCGATGTGTGGAC
0138
ACCTAACCAGCATCCAGAATG
0476
chr2
17956925
1795697





G


2
91


TTN_124#3
TCAGTCATGCCATGTAAAAGA
0139
GAATGAGCCTCACATTCACCA
0477
chr2
17958297
1795836



GG

G


8
52


TTN_26#2
CTGGATTGACTTTGGTCCAGG
0140
GGAATTTGATGGAAAGGCAAA
0478
chr2
17946613
1794667





G


0
62


TTN_31#3
TCGTTTTGTCACATCAACCAC
0141
TTCTAATATCTG CCTGTGAG CT 
0479
chr2
17947684
1794774





ACG


0
64


TTN_37
AAGTAAAGTGGTGACCAGAGA
0142
GGCAGCTTCAAGTGATTTCAAA
0480
chr2
17948728
1794876



AGTTG

G


1
43


TTN_48
TTCCCCTTGAATATGAACTTTG
0143
TTTATAATG GAG CATGACTCAC 
0481
chr2
17950202
1795022



G

CC


3
41


TTN_77
TGTGGCATTGAGAAGAGAAAG
0144
TGTTTATATCTCTCATGCTCTG
0482
chr2
17953485
1795351



G

CTTG


6
01


TTN_96
TGGTAGATGTTCACTGAATTTG
0145
CCATGAAGCTATGTCCTAAGCA
0483
chr2
17955271
1795530



TGTC

G


4
69


TTN_117
AG CCTACAAATTG CAGATGAG
0146
TTGTCTTTAGAGAGGAATAAAC
0484
chr2
17957212
1795726



C

CAGG


0
98


TTN_124#4
GCTGCCTTTAAACCACTTGACC
0147
GCACTGTATCCGTCCATGTTTC
0485
chr2
17958358
1795843








2
04


TTN_26#3
TCCATCCTATTAGAAAAG GAGA
0148
AAGACTCCAGCACTTCATCAGC
0486
chr2
17946663
1794673



CAG




6
80


TTN_31#4
TCATTAAGAAGTAATGTAGCCA
0149
AAAATAAACATAGGCTTCTTCT
0487
chr2
17947737
1794780



GGAGG

GCTTC


7
69


TTN_40
G CAG GAG CTAGTTATTTACCAA
0150
GTTTGATTAACCAGAAGGCAG
0488
chr2
17949336
1794937



AGC

G


4
45


TTN_49-50
GGGCTTGAATTTTAATCAAGTG
0151
CGTGGGCTTAATTTTAGTTTTG
0489
chr2
17950434
1795049



TG

AC


4
19


TTN_78
TTGGTAGAACTTCCCTTGGACC
0152
ATGTGTAATATGAAGGAATGT
0490
chr2
17953564
1795361





GTGTG


1
21


TTN_97
TGTGGAAGAAGAAGAGACTTT
0153
GGTGTACTGTGCAACTCTTCCC
0491
chr2
17955333
1795535



GAGG




5
95


TTN_120#1
GGATTACTTAAGCAGAATTTTA
0154
CCCATTTCGGTTACCTGGAAG
0492
chr2
17957654
1795772



AGCC




8
31


TTN_124#5
ATGTGGGGAAGGGTAGTTTTG
0155
TGATAACATTGCAACCCTCCAG
0493
chr2
17958418
1795848



C




1
28


TTN_29#1
AGACTGTTGGGAGTTTGAAGC
0156
GAGCTGGATAAAGACCGTGTT
0494
chr2
17947165
1794722



C

G


4
81


TTN_32#2
TCTGCAGCAACTCTGAAGATG
0157
TTGAAAATGTCCCTAAGAAATC
0495
chr2
17947884
1794793





CAC


1
21


TTN_41
GCGGAAAGAGAAAGGCAAAG
0158
GAACCCTTAAAGACCACTTCCC
0496
chr2
17949390
1794946








3
75


TTN_51
GAGTGAGATGGTAAAGAAAAT
0159
G GGAGTG GAAGATAAGTG GAT 
0497
chr2
17950512
1795055



TAAG CC

GG


5
61


TTN_80
GAGGGATCCATTGCTATGTGT
0160
CACCTCTAGGGTTCCTACTCCA
0498
chr2
17953807
1795385



ATAAG

C


8
96


TTN_99
GGGTGGACAGACACTTTTGTT
0161
GATTTGTGCATGTGCCTATGTT
0499
chr2
17955440
1795546



C

C


1
99


TTN_120#2
TGTCCTGCATCCTCTACTGTGC
0162
TCCCATGGATGTTTTAACTGGG
0500
chr2
17957710
1795776








7
74


TTN_124#6
AATTAGATGCTTCTGGACTCCC
0163
TTTCTTGTGTCAGCATGTATTG
0501
chr2
17958478
1795855



C

TC


2
13


TTN_29#2
GCCATGGTCTTTTCGCAGTG
0164
GGGGAAATTGTTGGCTATTTT
0502
chr2
17947219
1794727





G


5
19


TTN_32#3
AAGACGTTCACTTCCACCACAG
0165
CTGCTGAATGTACTGCCTGCTC
0503
chr2
17947919
1794798








5
95


TTN_42
TTTGAACCACTTCTGTATTGGA
0166
GTTTCATCACCATTTATTTGTT
0504
chr2
17949480
1794952



ATG

GC


1
05


TTN_52
AGAAAGCAGACAATGGAAAAC
0167
TCGGTGTAAATGCTTACTTTCC
0505
chr2
17950581
1795061



AG

AG


6
88


TTN_81
GCTACTGAGAAAGATTTGGAA
0168
TTTACACAAGCGTTTCGAAGG
0506
chr2
17953897
1795392



CACC




8
42


TTN_106
TTACGCACAACTTTGAACTCTG
0169
CTGCTTGATCCTGCTTCAAATC
0507
chr2
17955980
1795603



G




7
10


TTN_120#3
TCACTGCTACCTTTGAACCAGC
0170
AAAAGAGCCAAATGGATCTAG
0508
chr2
17957757
1795782





GG


9
23


TTN_126#1
TGTTGCCTCCAACACTAATACA
0171
ATTAAATGGCTCTGCACCCATC
0509
chr2
17958643
1795871





C


4
91


TTN_29#3
CATTTGTGCCAACCAACTGC
0172
ATGCACCAGATAAGCCCATTG
0510
chr2
17947267
1794731








1
95


TTN_33
TTAAATTTTCCCCAACAAAGCC
0173
CTGCAGTTTGTATCCCTGGTC
0511
chr2
17947995
1794806








9
08


TTN_43
TCCTGCATCCACTCTGACTTTC
0174
TTCCAAGAGATTGTCATTTCCC
0512
chr2
17949544
1794960








7
92


TTN_53
GCGAACCAATTCAAAGAAAAC
0175
TGCAGCGAAAATTGTTACTTAA
0513
chr2
17950685
1795071



C

TG


9
64


TTN_83
TGAGTGTCCTGTGTGGATAGA
0176
ACAGACCTGTCTTGAGCGACT
0514
chr2
17954032
1795408



ACC

G


4
38


TTN_108
GAGGAGACTCCACAACTTTCA
0177
GGAAGGGCTGGTGTATGAGTA
0515
chr2
17956173
1795620



ATAAG

G


4
24


TTN_121#1
TTGCTGATTCAGAAGATGTCG
0178
CAGATACAAGGCACACACTCA
0516
chr2
17957849
1795789



G

CC


9
94


TTN_126#2
GTGGGTTGGAAGCTGAGCAAG
0179
TTCCTTAGAACCGCCTTATTTT
0517
chr2
17958702
1795876





G


5
72


TTN_126#3
CTCCAGAGGTTCCAGTTCCG
0180
AGTGGGGAATACACCTGTGTG
0518
chr2
17958762
1795882





G


7
17


TTN_129#9
TGGGTGGTTCTGAAGAAGGG
0181
TTGCCCAGCTCAAATTTTATTC
0519
chr2
17959743
1795980








7
76


TTN_134#4
GAAATGCTCATTTGGTGTACC
0182
AATCACTTCAATG GAG GTG GA
0520
chr2
17961168
1796123



G

AG


3
25


TTN_136#2
TCTGCAATTTGTGAAAGG GAT
0183
AAGGAACCAGATACTGCATGG
0521
chr2
17962127
1796217



G

C


5
70


TTN_147#5
CAACTTCAACATTTACTTTG CA
0184
GATTCTGGGGAATGGACTGTG
0522
chr2
17964164
1796422



TCTTG




3
22


TTN_170
CTACCCCATGGCTCTGTG
0185
CTGCAAAGCAGCTCCAGAGT
0523
chr2
17966889
1796694








9
47


TTN_17b
TTTTTATTAGGAAAAATGACCA
0186
CAGGGATGATAATGTGAAATA
0524
chr2
17944826
1794487



TCA

TG GA


4
12


TTN_3#9b
GCTGGAGAGCCTCCGATG
0187
CACAAGTGCTTTTGAACTTAAT
0525
chr2
17939913
1793996





GAAAG


4
27


TTN_126#4
AAACCTTTCACAAAGAGACGG
0188
TCATCACCTATTTCAGTTGCCT
0526
chr2
17958814
1795887



G

G


1
78


TTN_130#1
TTCTTCCATGGGGTAAGAAAG
0189
TCATGTGAAGCAGTGAATGAC
0527
chr2
17959898
1795994



C

G


7
94


TTN_134#5
GTGGACGACCTAGTGATTCCT
0190
TTGTCCTCTAGAAAATGGAGG
0528
chr2
17961224
1796128



G

C


0
71


TTN_137
CAGTATGCAATAAAACAAACAG
0191
AAGGACAAGGCAGTAGGAGTG
0529
chr2
17962217
1796227



CAG

G


1
75


TTN_148#1
GCCATTTTAGCCCTCGATTTTC
0192
TCATAGCTCCTTGCCACAGTAA
0530
chr2
17964349
1796439





AG


5
37


TTN_13#32b
ATGGACCTACCGTATTCATCCT
0193
TGTATAAATAATTTGGGCAACA
0531
chr2
17944231
1794429



T

CA


9
93


TTN_90b
TTGGGTATCCAAATTTTTATCT
0194
GCAAAAGAATAAAATCAAAGC
0532
chr2
17954732
1795477



TG

ACATT


3
22


TTN_163
AGAAAATCTATTCAAATGCGAG
0195
TCAAGGTAGGAAGGCAAAGAG
0533
chr2
17965862
1796588



GTG

TG


4
61


TTN_126#5
GTGGTTTGGTACTTTGCTCCAC
0196
CCATCTCACTGGGAGAAACAA
0534
chr2
17958870
1795893





TC


8
60


TTN_130#2
TGAAAGCATTGTGTAAGTAATG
0197

0535
chr2
17959934
1795998



GG

TGTGTGTTGGGTCTCTTTGTTG


5
13


TTN_134#6
AAAGGCTTTTCCTCACTTGCTG
0198
CCTGCAATTTCTAAAAGAGCTG
0536
chr2
17961280
1796134





AAC


0
49


TTN_139
TTGTGATG GAG GAGAAGCTGA
0199

0537
chr2
17962881
1796290



C

CTACATGCTCCCTTCTGTGAGG


7
99


TTN_38b
TCTTTAAACACATAAAGGATCA
0200
AAAGTTGAGAAGGTACCAAAA
0538
chr2
17948903
1794895



GGT

CTCT


0
29


TTN_175
TAGAATTAGTAAACTAGTCTTG
0201
CATTGAATATTGTGCTTAAAGA
0539
chr2
17967442
1796746



TGGAAACC

GAGAA


6
25


TTN_153
GAAACTGATCTTTGCAAACGTG
0202
CATGAGCTAAGAGTTTATGAG
0540
chr2
17964836
1796485



TAT

ATTTCC


6
83


TTN_154
CACCTGGCCCTGCTCAAT
0203
G GG CTAG CCATCG GAG GAT
0541
chr2
17964874
1796491








0
36


TTN_127#2
GATTGTCCAGATCATGCGAGA
0204
GTGCTGGACTCTACTTACCCAT
0542
chr2
17959041
1795908



G

TAG


2
96


TTN_131
AAAGTGTACTGACTGAATTGTT
0205
CTTAACTCATCTTTTCTCTGTCT
0543
chr2
17960014
1796008



TG CC

TGG


6
93


TTN_134#7
TGTGTTTTATTTGAGTGTGAAA
0206
CCAAGCAGAGGGTACTGTTTA
0544
chr2
17961337
1796140



CTGC

TCC


1
08


TTN_140
CACTGGGAAAGGACAAAAGCC
0207
GAGTTTTGCATGCTCTCTTGTG
0545
chr2
17962918
1796296








6
04


TTN_164
TGAGTTAATGTGCACTGAAGG
0208
GGTGGCTCAGTTTTCCAGT
0546
chr2
17965906
1796593



A




5
64


TTN_87
AAAGAGTTGCATCCCAAAGAG
0209
ATTGCCAGCCTTAAAATCTTAG
0547
chr2
17954337
1795436





C


6
25


TTN_100b
GGCAGCATAGTACATATGAAG
0210
TGTAATCTTAGATGTCTCAGAA
0548
chr2
17955665
1795568



ATCG

GGTGA


8
94


TTN_10#lc
AG CATTCCTTGATATATTTGTT
0211
ACCTGAAGATAATG GAG GAG G
0549
chr2
17941628
1794168



TCTAA

AGA


4
83


TTN_128#1
TAATACCCAGGGGAGAAGGTG
0212
TTTCTGTCTGACTGTGTCTTTG
0550
chr2
17959175
1795922



G

G


7
87


TTN_132
TTTCCCACACATGTACAGAAAG
0213
CACCAATTTCAACTGTTTTACT
0551
chr2
17960272
1796031



C

CC


8
98


TTN_134#8
CATTTTCCTTTTCTGATCTACC
0214
AGAGAAGGGGATTCCATCATT
0552
chr2
17961389
1796146



AAG

C


9
16


TTN_144#2
TGGACTTTCCTTTCTGAGACCA
0215
CAGGACTTCTTTTCCTTGATGA
0553
chr2
17963488
1796354



G

TG


2
66


TTN_128#2b
ACCAAAGACACAGTCAGACAG
0216
AATGTCCATAAAACTGTCACAA
0554
chr2
17959226
1795926



AA

TAAT


4
63


TTN_79#1b
TCTTCCAAACTGAACACAAAAT
0217
TAGATGCTCTTTCAGGGAGAA
0555
chr2
17953661
1795370



TTAC

CT


2
09


TTN_141b
TTACAAGAATTTAGTGACTTAA
0218
GGGAGGAAACCAGAGCTTCAA
0556
chr2
17963102
1796314



ACAGGA




9
04


TTN_102c
GTATGCACACTGTGACTAAATC
0219
TCATTTCATGTTTGTCTCTGTTT
0557
chr2
17955822
1795585



TATTATT

TC


5
24


TTN_129#1
ACAAAGGCAAGAGTGATACAT
0220
ATGACCTTGGAATGTGTTGTG
0558
chr2
17959276
1795934



TTAAG

G


6
49


TTN_133#1
AAGTGAAAATTTAAGTGATG CA
0221
AACAAGTTTGCAAGAAGAAAT
0559
chr2
17960378
1796044



AGC

GG


4
60


TTN_134#9
CAACTGCCCCTGAATTGTTTTC
0222
ACATGCCAATCCCACGTAATC
0560
chr2
17961440
1796151








6
85


TTN_146#2
CTTCGTCTGAAGCATGAGTTC
0223
GAGGGTGACATTGTTCAGCTT
0561
chr2
17963824
1796387



G

G


6
90


TTN_166
AGAGACATGTGCTTTTAAGTTT
0224
GATTTTTGTTGTAGAGTAAGGC
0562
chr2
17966138
1796616



CAC

AAAC


4
59


TTN_92b
GATTAGTTTTAGTGTCTGGATG
0225
TGTCAAGTGTTTATGTTTGG GC
0563
chr2
17954866
1795489



CTT

TAC


4
04


TTN_39b
TGGCATTGTTCATGAGCCTCT
0226
AAATGCACCCATATACTAGATT
0564
chr2
17948994
1794901





GC


2
80


TTN_104c
ATGTTTTGATCTATAGGGACAA
0227
ATTAGTAGTTTGCTAACTTCAC
0565
chr2
17955888
1795590



ACAATGC

TATGC


9
85


TTN_129#2
TGGGAACTATTTGTTTCCCGTC
0228
TTGCCCTGAAAGGTCAGTTATA
0566
chr2
17959337
1795940





C


1
26


TTN_133#2
AACTTCTGGTTCAGTAATGGGT
0229

0567
chr2
17960438
1796049



TC

AGAATTGAGGAAGGCAAGTCC


8
99


TTN_134#10
TTCCATATTG GTTAAATAG CAC
0230
AG CCAAGAAG CACTGGTAAAG
0568
chr2
17961505
1796157



ACAAG

G


7
02


TTN_146#3
CACAATGTGAACCCTGTCACT
0231
GGGGAGATAAAAGACATACAA
0569
chr2
17963868
1796392



G

GACATC


3
96


TTN_101b
GATGCTATTTTGTTAATACACA
0232
ACAACAACATATGCCTTGTCAA
0570
chr2
17955713
1795573



GATCTTA

A


8
67


TTN_12#3b
AACTGTTGCAGACACTTCAGTC
0233
TGACCTTATCAAGAGATGGTGT
0571
chr2
17942250
1794228



C

C


5
54


TTN_73b
GAAACCATAGTTTTAAGAGCA
0234
AAATATCACTTTGGTTCAGCTC
0572
chr2
17953003
1795302



GAGGAC

ATTT


7
70


TTN_115c
TTCTGGTAATTCAAAGGGAATA
0235
TTGAATGATGATAGACAATTAA
0573
chr2
17956982
1795701



TGTG

AAATAAG


2
56


TTN_129#3
AATGAATGATGGTGGTTCTGT
0236
AAGCCCAGTCCAGTCTTAGTG
0574
chr2
17959388
1795946



G

C


0
52


TTN_133#3
CAGTACCTGCTTTTCTTCAAGT
0237
GGGAATTTCTTTGCATCAATGG
0575
chr2
17960494
1796055



GC




0
74


TTN_134#11
TGTCCTCTTGCTTGGGTATTTT
0238
GAGG CTTTAGTTGAAAG GG GA
0576
chr2
17961561
1796162



C

G


4
66



TTTTACCACATGCTAAGGGTGA
0239
TGGTCATAAGAGATGTGACTG
0577
chr2
17963958
1796401


TTN_147#1
C

CTG


2
84



CAGAGATGTACCTCGGACTCT
0240
CAGACTTACCATACCAATCAAA
0578
chr2
17940422
1794046


TTN_5#3b
G

GG


7
26


TTN_129#5b
TTTGACTGTCAGATGCCCACT
0241
GCAGCTGCAAGGCTAGAAT
0579
chr2
17959523
1795956








3
81


TTN_169
TCCCAATTTGCTGGAGATGT
0242
GCAGTGAACATGATGGGACAG
0580
chr2
17966680
1796671








3
47


TTN_119#1c
CACCTTCAGGCTATACTACAAA
0243
AATCAAATATGAACTTAGGGA
0581
chr2
17957529
1795757



ATGA

GAAAT


8
44


TTN_129#6
CAGCTGCTCCTCCCAACATC
0244
GTTGGTTCCAGTGAGGGACTG
0582
chr2
17959567
1795964








5
13


TTN_133#4
AGGTTGGGAGATGGTTCCTTG
0245
AATTCGCTGTG CTCAAGG GC
0583
chr2
17960551
1796061








1
22


TTN_135#1
AAGTCCATGCCAAACAAACTAT
0246
CAGGATACCACGTGGACTGAA
0584
chr2
17961804
1796185



TG

G


1
40


TTN_147#2
GACTG GAG GTTTCTCCAGCTA
0247
TGTTGACACCACTGAAACCAAA
0585
chr2
17964010
1796406



TG

G


4
90


TTN_93b
GCCCCCATAACCAGTGTATT
0248
TCTAAAATCACAGTACTTTCTG
0586
chr2
17954898
1795492





GCTAGT


2
23


TTN_103b
GACAACAAGAGGGATAAAAAT
0249
TCTAAATACTTGAGTATAAAAT
0587
chr2
17955857
1795588



CTGC

CCATGTG


8
24


TTN 162b
GGTAAAGGTGATTATCTGTGTT
0250
CGAGGCTGGTCTTGAACTAAT
0588
chr2
17965806
1796583



GACC

TT


1
40


TTN_125c
CTCATAACTTTGCTAAGAGCCC
0251
ATGGGATCTTCAGCTACAAAAA
0589
chr2
17958558
1795860



AAA

CAA


1
11


TTN_129#7
GGGAGCTGGTACTCTCGATCT
0252
GAGAAATTAAGGAGAGCAGCA
0590
chr2
17959626
1795969



TC

AAC


9
51


TTN_134#2
TTGATAAACCTGGGAGGCCC
0253
TCAAGAAATTGTCCTGGAAGTT
0591
chr2
17961057
1796112





G


4
06


TTN_135#2
TTCCACAAGGAATATGCACAG
0254
GAATGTCAAACCCCTGGAAGT
0592
chr2
17961843
1796189



C

C


1
38


TTN_147#3
TTTCATGGGTAATTCTTTCAGC
0255
TGAGAAAAGTCTTGTGGAAGA
0593
chr2
17964062
1796412



C

ATCC


6
03


TTN_84b
TTTTCCTAAAACCCAGTTTCAT
0256

0594
chr2
17954184
1795421



CA

GTTTGTGGTCTTATCCTCACG


7
19


TTN_160
TGATGAAAATGTAGGTGATTTG
0257
CATCCTGTATTTTCCTGAGTGT
0595
chr2
17965536
1796556



C

-ITC


4
63


TTN_21#10b
CATACTGAAACGATCTTTGG CT
0258
CCTGGTTCAAAGATGAAGCTG
0596
chr2
17945829
1794588



TT

AT


0
64


TTN_127#1c
GCCAGTAACTCTTAGTAATTCA
0259
CATGATCTGGACAATCTTTTCA
0597
chr2
17959002
1795904



TTCCA

AC


8
27


TTN_129#8
ACCACTGTCTTCGATGCCAAC
0260
TCATCTTCCCTTAGATCAACCC
0598
chr2
17959686
1795975





TC


0
07


TTN_134#3
CGGGAACTGTCACTATTTTCAC
0261
TCCTACTGGAGGACCAAACCC
0599
chr2
17961112
1796117



C




4
70


TTN_136#1
TTGAATTTGCATGGCAGAAAA
0262
CTCAAAGTTCCATCCACACAGC
0600
chr2
17962087
1796213



G




2
53


TTN_147#4
TTCTCTGTAAGCCTTTCCTCCC
0263
TCTGCCTGGTATACTGCGACT
0601
chr2
17964114
1796417





G


5
18


TTN_118b
GCTCATGGATATATAACAGGC
0264
AGTTGGTTAGCCTTGATTTTTA
0602
chr2
17957422
1795746



AGTG

AC


8
97


TTN_143b
CCCACATTTTATTTATCCATTCA
0265
CCTCAACAGACGCACTAAAAA
0603
chr2
17963330
1796337



TT

CA


4
69


TTN_155
AATCCTCCATTGGCCTACCC
0266
CATCTGCCTGTCCTGATGC
0604
chr2
17965025
1796509








2
51


TTN_129#10c
AAAATTTGAGCTGGGCAACAT
0267
AAAACTCTTAAATATAGATG GT
0605
chr2
17959805
1795986





GTTGAAAG


9
94


TTN_134#12c
CCTTTCAACTAAAGCCTCCACA
0268
TAAACAGAGCATGGTGTTGC
0606
chr2
17961624
1796169








9
39


TTN_158c
ATTTCACATGATATGTGGTATT
0269
GGAAGGGGGAAGGGTCATACT
0607
chr2
17965401
1796542



AATGT

A


9
99


TTN_27c
AAGGCAGAATTATCCATTTAGT
0270
TACAGTCATCCCTCCCAAAATA
0608
chr2
17946853
1794691



GA

AGTT


4
29


TTN_79#2c
GACAGTTCTCCCTGAAAGAGC
0271
GTAATGTTGGCGTTGTCTCTG
0609
chr2
17953698
1795375



AT

G


4
16


TTN_21#11b
CATCTTTGAACCAGGAAACCTT
0272
GGAATATTCACTACATCCTACT
0610
chr2
17945885
1794594



AG

ACATTCTT


0
49


TTN_3#1b
TCAAATGTGTGTTTCTGCTTTG
0273
ATTTATGGGGCAAGCCTTTAAG
0611
chr2
17939452
1793952



G

A


4
23


TTN_134#1c
GAGTCCATTCCACTGAAACACT
0274
GGTTTATCAAAGGTATTTCTGA
0612
chr2
17960998
1796105



TT

CTG


4
83


TTN_159c
AGGGATTTTAAAAGGCAAATA
0275
GCCTCCTTTCTTTTTGACTTAC
0613
chr2
17965459
1796549



CA

G


3
28


TTN_28#1 c
ATGGCATCAAACCAGAGTCAT
0276
ACAGGCCTTCCAATGCCTAAG
0614
chr2
17946937
1794699



GTA

A


3
33


TTN_36#5c
TCCGTCCTTCAGTCAGTATTTC
0277
TTTCTTAAGGACACCTGTGTGA
0615
chr2
17948630
1794868



AT




1
00


TTN_4#1 b
TTAAAAGAATTTTATGCAAAGA
0278
AAAGTCCAGCTCAGCAATGTTT
0616
chr2
17940159
1794021



TGG

T


9
29


TTN_105c
TTTTGGTCGTTTCAGATTTGTG
0279
AAGTTGTGCGTAAAGGTCAAA
0617
chr2
17955922
1795598



AG

G


0
19


TTN_138c
TTGTTACAGACATTGTTAAGAT
0280
GTGTTTGCAGGTAAATTGTAAA
0618
chr2
17962364
1796239



TCGAT

AATCC


5
70


TTN_161c
TGCAGCTGGCTGTAATGTGAT
0281
GAAAAGGGATTTTACATCTTCA
0619
chr2
17965673
1796570





AA


7
33


TTN_35#1 c
TTTATTTTAATTGATAGGCCTA
0282
GGTCTACAGAAAGGAGGAGTT
0620
chr2
17948243
1794830



ATATCTG

GA


6
27


TTN_107c
TG CAAATCAGGTTCATAG CA
0283
TAGTGAAGCAGTTGGATGGAT
0621
chr2
17956050
1795610








5
54


TTN_134#13b
TGCATGCTACAGATCTCACAAA
0284
AAAATAGACAATAACCTAGCTG
0622
chr2
17961685
1796174



TC

TCATTC


8
97


TTN_116c
GTTGACTGTGGATGCGGAAC
0285
AGGCTTATTTTACAAAGGGGG
0623
chr2
17957111
1795717





ATA


0
59


TTN_145c
TTTTAAAACGATAACGATCAAG
0286
TACTTCTGCAAAGATTTTCCCA
0624
chr2
17963587
1796362



ATT

TT


4
70


TTN_165c
GAGTTTCATGGCAGAAATCCA
0287
TTAAAGCACTTCCAGCTTTTCA
0625
chr2
17965957
1796600



G

TC


5
69


TTN_36#3c
TAAGGTCTTCCTCTGTTGTAAA
0288
CTTGTCGGTTGCTTTCAGA
0626
chr2
17948534
1794857



GG




1
38


TTN_146#1c
AGGAATTTTGGGGGAAATGAA
0289
AGACGAAGGACCTTACAAGCT
0627
chr2
17963775
1796382



TA

G


4
53


TTN_98b
AGAATTTAACACACTCGAAGAT
0290
TGTCCACCCGTCTGCTTTC
0628
chr2
17955371
1795544



TTTT




0
09


TTN_167#1c
ATGATACATGATCACCTTCTAA
0291
CCCTCCTCCCGTGTAAGTTTC
0629
chr2
17966313
1796638



AATACTTC




5
32


TTN_148#c
ACTGTGGCAAGGAGCTATGAT
0292
TCTCCCAGCTAAAAACTTCAAC
0630
chr2
17964391
1796442








8
93


TTN_167#3c
CTTTCTCGTTTCAAAACCTAGT
0293
TCTTTTAAAATGGGTCATTGTG
0631
chr2
17966415
1796646



TCC

C


0
94


TTN_45c
TTAGAACTTGGCGTCCTATCTT
0294
AAATTTGTATCTGAAACACTTC
0632
chr2
17949901
1794996





TCCT


3
51


TTN_129#4c
GACTGGACTGGGCTTCTTAAT
0295
GGTTGGCAAATTTTATTTCCAC
0633
chr2
17959463
1795952





TTA


8
32


TTN_89#2b
GACAGCTTTCTTCAGCACTTCA
0296
CCTACTTCAATAGGCTATGGCT
0634
chr2
17954616
1795465



AA

ATACT


1
92


TTN_46b
TAGATCCTGAATATTGGATGTG
0297
TCCTGAAATTTACTAAGGAAGC
0635
chr2
17949982
1795005



GT

TATGTA


7
20


TTN_149c
CCGAGCTCATCACTTGAA
0298
TTATCAACTTTCCCCAGTGTAG
0636
chr2
17964463
1796450





AG


5
31


TTN_168c
TGGCCCCATTTAGACACAAAC
0299
TTTAAAAATACCTTGTAGGGAG
0637
chr2
17966505
1796654





CAC


7
74


TTN_55c
TCATTTCAGATGGCTGGATAGA
0300
TGTTTTATGTTGGATACTGGTA
0638
chr2
17951058
1795109





TTACG


1
28


TTN_134#14c
AG CTAG GTTATTGTCTATTTTC
0301
AAAAATGTTTTAAATCTTTCTG
0639
chr2
17961747
1796181



AATGTAAT

TCAAC


7
13


TTN_32#1b
TCCCAAGGAATACTAAAGAGT
0302
AGTTGCTGCAGAAAACATGTAT
0640
chr2
17947840
1794788



AAACA

GG


4
52


TTN_95b
AGACAGTTATGCAAATGTGAA
0303
AGGGGTTTGGCAAGGGTTA
0641
chr2
17954990
1795504



GGTATTATT




2
46


TTN_15#2c
TGAGCCCCCATCATCTGC
0304
GGTCTCCAGACCTCTACTCTAT
0642
chr2
17944684
1794473





ACTCAT


1
88


TTN_171c
AACCAGCTTAAATTGATCTTAC
0305
TTCACTTAATATTCTAAAAGTT
0643
chr2
17967130
1796716



ATTCC

GAGGTC


2
96


TTN_59c
TTTCTGTGCAATATGGTTTTAA
0306
TG CTG CTCCAATAAATACAG GT
0644
chr2
17951385
1795141



CATAAAT

TT


8
47


TTN_142e
GGTTAGAAAATGTAAAAGGGA
0307
TGAATTATATCTCAATAAAG CT
0645
chr2
17963239
1796329



AACATT

GTTAAAAA


3
41


TTN_21#8b
TCATGGGCAGCATTACGAA
0308
ATGAAGAGTGGAGAAGAGCCA
0646
chr2
17945718
1794576





ATC


3
75


TTN_62-63
GGAAGGAAGAAGAACAAAGCT
0309
TCCAATTCCTCTGCTGTATATT
0647
chr2
17951540
1795181



TAAAT

TTG


2
21


TTN_150c
CAGCAAACGGACAGCACT
0310
TCACAGATAAGTCCAATTATTT
0648
chr2
17964577
1796460





TACCC


3
68


TTN_172c
TCCAGAGCCAGAGATCAATAA
0311
CAACACAGTTATTCTTTAAATA
0649
chr2
17967187
1796723





CAGTTCTA


6
75


TTN_82c
AATATGTTGATTTCCTGGGGTA
0312
TCATCTTGTGTATGTGCCTTG
0650
chr2
17953965
1795399



AA




9
58


TTN_152c
G CACAGAAACCATATTGTG GA
0313
GCATAAGTTCAAACTCTACATT
0651
chr2
17964745
1796478



AAAG

TGTT


1
81


TTN_6b
CAAAAAGGTGAATTTTCCCACA
0314
CCAACCAAAACCTAAGGAAAA
0652
chr2
17940594
1794064



TA

CAC


1
02


TTN_67-69
CCAGAGCACAAGAGATAGATC
0315

0653
chr2
17952217
1795240



A

TGATGTGCTGCTGGAAAA


5
56


TTN_151c
CTACTCTAGGCTTCATGCACGT
0316
TCAGTGCTAAGCAGGGGTCAC
0654
chr2
17964687
1796473



AT

T


3
91


TTN_173c
CACTTGTGGGCAGTCATACAA
0317
TGGATTCTGGAGAATGTAGGT
0655
chr2
17967245
1796726



AA

AG


1
81


TTN_85#2c
ATGAAGAAGCTTATGAAAGGC
0318
TTTCATATCTATTGCTCTAAGA
0656
chr2
17954270
1795430



AAAT

CATTTT


3
02


TTN_26#lc
TGTGAACTATTATTGAACACCT
0319

0657
chr2
17946550
1794661



AGGAAG

AAGCCTGGACCAAAGTCAA


9
53


TTN_8#9b

0320
TGGCCTTGTGATATGGCACTA
0658
chr2
17941449
1794150



GAGCACAACCGTCATTGAGTG

C


8
92


TTN_71
CCAGAGCAGAAGAGATACATC
0321

0659
chr2
17952830
1795291



A

TGGGTGGGGCGATAGAAA


4
52


TTN_156c
TGATTCCCTAAGGGTAAGATTG
0322
TTGTTTCATTCTTTGCTTTAACT
0660
chr2
17965142
1796516



T

CTCC


2
24


TTN_174c
ATG CTTAAATCATCCATAAG GT
0323
ACAAGACAATTTTCCTCCCTCT
0661
chr2
17967288
1796731



TCC

AAG


8
25


TTN_86c
TGTAATGGGGAAATTTGTATGT
0324
CTTAATCTTTTGAAACTATGCT
0662
chr2
17954306
1795433



GAG

TGG


1
01


TTN_22c
GAATGAAATGTACGGCATTTAT
0325
TCAAAACAACATTTTTGTCTCC
0663
chr2
17946013
1794606



ACACA

TTT


4
33


TTN_8#4b
GCTTCGCTGGCCTTGCTA
0326
GCGCACTCAAATTAAAGTCACT
0664
chr2
17941176
1794122





CAT


2
61


TTN_147#6c
TCCCCAGAATCACTGGGTGT
0327
TGTTTTGCTGAGTTTTCTTTAT
0665
chr2
17964221
1796427





GCC


2
59


TTN_157c
G GAG GATG GACTGAATCTAAC
0328
AAAATCTGCTGAACCAGCCAC
0666
chr2
17965275
1796529



CA

A


3
97


TTN_25#2c
CTGGCTCTTCCCAGTTGACAG
0329
TGATGGTGGGTCTAAGATTAC
0667
chr2
17946371
1794640





AAAC


2
87


TTN_76c
AAAAGACAAACATAGTGAATTT
0330
TCATCTTGTTAGATGCCCCTTC
0668
chr2
17953403
1795344



AAGGA




0
91


TTN_119#2c
ATTTCTCCCTAAGTTCATATTT
0331
ATTGCATATATACGTGCGTTTG
0669
chr2
17957571
1795761



GATT




9
67


TTN_3#12b
TAGCTCTGACTTGGTAAACTGT
0332
CCAATTTCATCTTTCTGGAATA
0670
chr2
17940075
1794014



GG

GC


7
35


MYBPC3_8
GGGCTGGGGATGATTTG
0333
GGGACACTAGCCAGATTGG
0671
chr1
47369117
4736933








1
1


MYL2_2
CACCCAGAGTAGGGGCCTGAC
0334
TTCAGGCCGAATTTGGGATTG
0672
chr12
11135677
1113570



CTA

TTT


0
73


MYH7_12
GGGAGTCTCAGAACCCACAG
0335
TGAGCAGACATGGCCCTCC
0673
chr14
23898925
2389924









0


GLA_2
GTGAAATCCCAAGGTGCCTA
0336
AGAAGTGCTTACAGTCCTCTGA
0674
chrX
10065874
1006590





A


7
58


LAM P2_4
GGGTAGGGCAGGCAGAGA
0337
CAGATAATGACTTCAATGAAAG
0675
chrX
11958274
1195830





CTA


1
87


G6PDe11
TAGCTCCACCCTCACCCCG
0338
GTGGCCTTTGCCCTCCCT
0676
chrX
15376033
1537605








8
57


G6PDe10
GGTCCAGCTCCGACTCCT
0339
GGTGCCCTTCATCCTGCG
0677
chrX
15376080
1537610


SRYc





4
16



TGGCTGTAGCGGTCCCGT
0340
TCGTCGGAAGGCGAAGAT
0678
chrY
2655045
2655252


SRYb
ATCCTGGACGTTGCCTTTACTG
0341
AAATAAGTTTCGAACTCTGGCA
0679
chrY
2655470
2655702





CCT





*Positions correspond to hg19. MYBPC3, MYL2, MYH7, GLA, LAMP, G6PD, and SRY primers were used as controls for CNV analyses.













TABLE 4 







Genomic library adaptor oligonucleotides











SEQ




ID


Name
Sequence
NO





Solexa_PE_Fh_AACT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTAAC*T
0680





Solexa_5′AACT
/5Phos/GTTAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0681





Solexa_PE_Fh_AGGT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTAGG*T
0682





Solexa_5′AGGT
/5Phos/CCTAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0683





Solexa_PE_Fh_ATGT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTATG*T
0684





Solexa_5′ATGT
/5Phos/CATAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0685





Solexa_PE_Fh_CACT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCAC*T
0686





Solexa_5′CACT
/5Phos/GTGAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0687





Solexa_PE_Fh_CGAT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGA*T
0688





Solexa_5′CGAT
/5Phos/TCGAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0689





Solexa_PE_Fh_GGAT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTGGA*T
0690





Solexa_5′GGAT
/5Phos/TCCAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0691





Solexa_PE_Fh_GTCT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTGTC*T
0692





Solexa_5′GTCT
/5Phos/GACAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0693





Solexa_PE_Fh_TCGT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTTCG*T
0694





Solexa_5′TCGT
/5Phos/CGAAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0695





Solexa_PE_Fh_TGAT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTTGA*T
0696





Solexa_5′TGAT
/5Phos/TCAAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0697





Solexa_PE_Fh_TTCT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTTTC*T
0698





Solexa_5′TTCT
/5Phos/GAAAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0699





Solexa_PE_SACGT
/5Phos/CGTAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0700





Solexa_Fh_ACGT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTACG*T
0701





Solexa_PE_SAGCT
/5Phos/GCTAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0702





Solexa_Fh_AGCT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTAGC*T
0703





Solexa_PE_5′TGGT
/5Phos/CCAAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0704





Solexa_Fh_TGGT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTTGG*T
0705





Solexa_PE_5′TCCT
/5Phos/GGAAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0706





Solexa_Fh_TCCT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTTCC*T
0707





Solexa_PE_5′GACT
/5Phos/GTCAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0708





Solexa_Fh_GACT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTGAC*T
0709





Solexa_PE_5′CGTT
/5Phos/ACGAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0710





Solexa_Fh_CGTT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCGT*T
0711





Solexa_PE_5′GTGT
/5Phos/CACAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0712





Solexa_Fh_GTGT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTGTG*T
0713





Solexa_PE_5′CTCT
/5Phos/GAGAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0714





Solexa_Fh_CTCT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCTC*T
0715





Solexa_PE_5′CAGT
/5Phos/CTGAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0716





Solexa_Fh_CAGT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTCAG*T
0717





Solexa_PE_5′GCAT
/5Phos/TGCAGATCGGAAGAGCGGTTCAGCAGGAATGCCGAG
0718





Solexa_Fh_GCAT
ACACTCTTTCCCTACACGACGCTCTTCCGATCTGCA*T
0719





*phosphorothioate linkage;


/5Phos/ = 5′ phosphate













TABLE 5







Summary of cohort characteristics*













DCM-A
DCM-B
DCM-C
HCM
Control
















Subjects, no.
92 
71
149
231
249


Women, no. (%)
28 (30)
16 (23)
60 (40)
NA
NA


Age, yr*
37.2 ± 14.0
40.6 ± 12.7
39.4 ± 12.5
NA
NA


LVEF (%)*
29.4 ± 12.2
24.8 ± 11.5
31.2 ± 11.5
NA
NA


Sequencing Methodology{circumflex over ( )}
Next-gen
Next-gen
Dideoxy
Next-gen
Next-gen


Mutation Positive, no. (%)
37 (40)
17 (24)
13 (9) 
 3
 7


Family History, no. (%)



NA
NA


Yes
78 (89)
 9 (21)
102 (68) 


No
10 (11)
30 (70)
47 (32)


NA
4
29
 0


Mutation Positive


Family History, no. (%)



NA
NA


Yes
33 (88)
 3 (21)
11 (85)


No
 3 (12)
11 (79)
 2 (15)


NA
1
 3





No., number;


*Values are means ± SD.


{circumflex over ( )} Sequencing methods were Next-gen, filter-based hybridization capture of TTN and next-generation sequencing or Dideoxy, conventional Sanger dideoxy sequencing.


Age and left ventricular ejection fraction (LVEF) are taken at the time of initial diagnosis of DCM.


Percent of subjects with family history excludes subjects in whom family history was not available.


Across all DCM cohorts, TTN truncating mutations were found in 25% of subjects with and 18% of subjects without a family history of DCM (P = 0.3).













TABLE 6







Sequencing data for TTN truncating mutations










# Reads



















Chr

Var
Nucleotide


Allele
Major
Minor





position
Ref base
base
change
Quality
Filter
balance
allele
allele
Total
Subject
Cohort





















179659280
TA
T
c.1246 − 3delT
23585.55
PASS
0.14
1146
6989
8135
pv-75
control


179647331
T
A
c.3101 − 2A > T
7728.48
FDRtranche10.00
0.58
348
250
598
N-50
control







to 50.00+


179604949
CT
C
c.12059delA
10405.56
PASS
0.43
1155
1551
2706
pv-1
control


179604264
G
A
c.12745C > T
1962.07
PASS
0.57
136
103
239
HFA-42
A


179591957
TC
T
c.19183delG
2609.57
PASS
0.54
120
104
224
HFA-9
A


179583071
ACAGAT
A
c.23798_23810delG
1797.98
PASS; StrandBias
0.22
124
451
575
PA-1
HCM



ATCTTGAC

TCAAGATATCTG



(SEQ ID

(SEQ ID NO: 723)



NO: 724)


179558736
C
T
c.30476 − 1G > A
2380.12
FDRtranche10.00
0.58
155
113
268
D13KD-1
A







to 50.00+; PASS


179535816
C
A
c.34186 + 1G > T
833.79
PASS
0.49
38
39
77
SS-333
HCM


179516991
C
A
c.34690G > T
1122.33
PASS
0.10
109
955
1064
12s-C11
control


179506963
C
T
c.35635 + 1G > A
7327.23
PASS
0.54
65
55
120
HFA-26
A


179506963
C
T
c.35635 + 1G > A
7327.23
PASS
0.55
263
217
480
HFA-41
A


179506964
C
G
c.35635G > C
6450.31
PASS
0.43
55
75
131
UK-B12
B


179498055
T
C
c.38024 − 2A > G
1440.31
FDRtranche10.00
0.58
288
207
495
MIY-11
A







to 50.00+


179497076
T
TA
c.38621_38622insA
2339.05
PASS
0.20
145
595
740
MDT-11
A


179485012
G
T
c.41313C > A
12126.6
PASS
0.52
421
383
804
UK-H11
B


179478864
CT
C
c.44336delA
4754.46
PASS
0.56
240
187
427
HFA-68
A


179477885
TA
T
c.44725 + 2delT
5928.55
PASS
0.48
90
98
188
MDD-22
A


179477885
TA
T
c.44725 + 2delT
5928.55
PASS
0.48
90
98
188
MIV-14
A


179477004
TA
T
c.45322delT
4088.96
PASS
0.52
178
166
344
UK-G1
B


179472127
C
A
c.48364 + 1G > T
3132.58
FDRtranche10.00
0.57
215
159
374
HFA-71
A







to 50.00+; PASS


179471841
C
A
c.48565G > T
17194.16
PASS
0.48
479
516
995
UK-A3
B


179467006
T
C
c.50197 + 3A > G
4964.76
PASS
0.55
226
183
409
12s-G11
control


179458075
AG
A
c.53935delC
4133.57
PASS
0.51
179
172
351
HFA-63
A


179457392
A
T
c.54422 − 5T > A
6168.22
PASS
0.54
215
186
401
UK-B1
B


179457005
C
T
c.54704 − 1G > A
1273.14
PASS
0.49
56
59
115
UK-B12
B


179456704
C
T
c.55003 + 1G > A
7835.09
PASS
0.53
310
276
586
MGR-11
A


179455162
A
T
c.56367T > A
5790.41
PASS
0.58
228
167
395
MEH-11
A


179454957
G
A
c.56572C > T
8250.03
PASS
0.52
269
253
522
MDJ-21
A


179454576
G
A
c.56953G > A
5819.28
PASS
0.52
197
184
381
MEW-11
A


179452435
G
A
c.58678C > T
3233.06
PASS
0.58
198
146
344
MAO-92
A


179447207
TG
T
c.60147delC
1144.41
StrandBias
0.23
165
552
717
SS-201
HCM


179444661
C
T
c.62425 + 5G > A
9756.67
FDRtranche10.00
0.24
8
25
33
MAM-12
A







to 50.00+; PASS


179444429
G
A
c.62572C > T
18928.38
PASS
0.57
572
428
1000
UK-A10
B


179443339
T
C
c.63405A > G
1427.03
PASS
0.55
224
187
411
MIS-11
A


179442324
C
G
c.63901 + 5G > C
7488.06
FDRtranche10.00
0.55
343
286
629
MCL-15
A







to 50.00+


179441649
C
T
c.64489 + 1G > A
6822.3
PASS
0.52
255
234
489
UK-B2
B


179441015
CT
C
c.64920delA
5378.99
PASS
0.66
395
201
596
UK-H2
B


179440067
CT
C
c.65867delA
2395.82
PASS
0.13
117
798
915
UK-C8
B


179438874
ATATGC
A
c.67057_67063delG
4467.51
PASS
0.23
165
559
724
MHG-23
A





CATATGinsTA


179434160
CAA
C
c.71774_71775delTT
7059.09
PASS
0.54
306
262
568
A-15
control


179433758
G
GT
c.72178_72179insT
4981.24
PASS
0.55
244
197
441
HFA-57
A


179433213
G
GGTTT
c.72723_72739delins
945.03
PASS
0.33
125
254
379
SS287-15
A




ATCTA
AGA




TCT


179428871
G
A
c.77065C > T
21554.16
FDRtranche10.00
0.61
188
119
307
MID-1
A







to 50.00+; PASS


179428871
G
A
c.77065C > T
21554.16
FDRtranche10.00
0.61
188
119
307
SS725
A







to 50.00+; PASS


179425091
G
A
c.80845C > T
1841.18
PASS
0.14
127
784
911
MGW-11
A


179424496
C
T
c.81440G > A
1670.2
PASS
0.14
148
898
1046
MIP-13
A


179424398
CAG
C
c.81536_81537delCT
6212.74
StrandBias
0.57
283
214
497
UK-C6
B


179424036
A
T
c.81898 + 2T > A
10097.32
PASS
0.47
266
300
566
MIA-1
A


179422457
G
T
c.82701C > A
2923.47
PASS
0.59
219
153
372
UK-B6
B


179417723
ATAAT
A
c.84977_84980delA
8795.62
PASS
0.62
230
142
372
UK-E12
B





TTA


179413187
G
A
c.88242C > T
2808.37
PASS
0.54
150
126
276
UK-C10
B


179412902
C
A
c.88528G > T
1639
FDRtranche10.00
0.53
87
76
163
MBG-121
A







to 50.00+; PASS


179412245
CTTTAA
C
c.89180_89184delA
10083.39
PASS
0.54
140
117
257
HFA-46
A





AATT


179410799
G
A
c.90241C > T
1584.39
PASS
0.12
107
782
889
MHQ-12
A


179410544
T
TGGG
c.90493 + 2insCCT
41982.72
PASS
0.58
862
621
1483
SS723
A


179408239
G
GT
c.91537_91538insA
3613
PASS
0.62
220
136
356
UK-E4
B


179406990
C
G
c.92569 + 1G > C
8827.13
FDRtranche10.00
0.51
180
173
353
HFA-83
A







to 50.00+; PASS


179406990
C
G
c.92569 + 1G > C
8827.13
FDRtranche10.00
0.51
289
274
563
HFA-66
A







to 50.00+; PASS


179404491
CCT
C
c.93376_93377delAG
12491.23
PASS
0.61
562
361
923
UK-G7
B


179404286
G
A
c.93583C > T
8470.72
PASS
0.55
364
299
663
UK-G9
B


179403522
T
A
c.94111A > T
7746.65
PASS
0.50
250
250
500
MHX-11
A


179402067
A
G
c.94942 + 2T > C
7331.01
PASS
0.50
269
265
534
pv-35
control


179401029
G
T
c.95522C > A
2296.56
PASS
0.58
158
115
273
UK-C9
B
















TABLE 7







Numbers of TTN missense variants* and truncating mutations in cohorts.













Cohort







size
Missense
Avg./
Truncating
Avg./


Cohort
(no.)
variants
subject{circumflex over ( )}
mutations
subject{circumflex over ( )}















DCM-A
92
117
1.27
37
0.40


DCM-B
71
103
1.45
17
0.24


DCM-C
149
136
0.91
13
0.087


HCM
231
263
1.14
3
0.013


Control
249
332
1.33
7
0.027





*Missense variants and truncating mutations with a minor allele frequency less than 0.01.


{circumflex over ( )} Average number of missense variants or truncating mutations per subject.













TABLE 8







TTN nonsense and frameshift mutations*












Chr 2







Position
Nucleotide
Amino Acid
Pedigree
ID
Diagnosis





179640342
c.6247delG
p.Arg2083fs
TSSDC011
450
DCM


179604949
c.12059delA
p.Lys4020fs
pv
1
Control


179604264
c.12745C > T
p.Gln4249X
HFA
42
DCM


179591957
c.19183delG
p.Ser6395fs
HFA
9
DCM


179583071
c.23798_23810delGTCAAGATATCTG
p.Gly7933fs
PA
1
HCM



(SEQ ID NO: 723)


179497076
c.38621_38622insA
p.Ala12873fs
MDT
11
DCM


179485012
c.41313C > A
p.Cys13771X
UK
H11
DCM


179478864
c.44336delA
p.Glu14779fs
HFA
68
DCM


179477004
c.45322delT
p.Phe15108fs
UK
G1
DCM


179471841
c.48565G > T
p.Gly16189X
UK
A3
DCM


179469903
c.49077G > A
p.Trp16359X
DNFDC144
05-0444
DCM


179463630
c.51883G > A
p.Arg17295X
DNFDC116
04-1545
DCM


179462477
c.52408C > T
p.Arg17470X
TSFDC050
115
DCM


179459153
c.53145_53146insG
p.Glu17715fs
TSFDC017
253-2
DCM


179458849
c.53347G > T
p.Glu17783X
DNFDC103
03-0941
DCM


179458075
c.53935delC
p.Glu17978fs
HFA
63
DCM


179455162
c.56367T > A
p.Cys18789X
MEH
11
DCM


179454957
c.56572C > T
p.Arg18858X
MDJ
21
DCM


179454575
c.56953C > T
p.Arg18985X
DNFDC142
05-0569
DCM





MEW
11
DCM


179452435
c.58678C > T
p.Arg19560X
MAO
92
DCM


179449211
c.60147delC
p.Pro20049fs
SS
201
HCM


179444429
c.62572C > T
p.Arg20858X
UK
A10
DCM


179441015
c.64925delT
p.Lys21640fs
UK
H2
DCM


179440067
c.65867delA
p.Glu21956fs
UK
C8
DCM


179438874
c.67057_67063delGCATATGinsTA
p.Ala22353fs
MHG
23
DCM


17943819
c.67745delT
p.Pro22582fs
MEK
111
DCM


179434160
c.71774_71775delTT
p.L23925fs
A
015
Control


179433758
c.72178_72179insT
p.Gln24059fs
HFA
57
DCM


179433213
c.72723_72739delinsAGA
p.Ser24241fs
SS
287-15
DCM


179428871
c.77065C > T
p.Gln25689X
MID
1
DCM





SS
725
DCM


179426039
c.79896G > A
p.Trp26632X
DNFDC088
02-1900
DCM


179425091
c.80845C > T
p.Arg26949X
MGW
11
DCM


179424889
c.81046A > T
p.Lys27016X
DNFDC081
02-1564
DCM


179424496
c.81440G > A
p.Trp27147X
MIP
13
DCM


179424398
c.81536_81537delCT
p.Ser27179fs
UK
C6
DCM


179422457
c.82701C > A
p.Tyr27567X
UK
B6
DCM


179417723
c.84977_84980delATTA
p.Tyr28326fs
UK
E12
DCM


179413476
c.87953G > A
p.Trp29318X
TSSDC019
268
DCM


179413187
c.88242C > T
p.Arg29415X
UK
C10
DCM





TSFDC002
23
DCM


179412902
c.88528G > T
p.Glu29510X
MBG
121
DCM


179412245
c.89180_89184delTTAAA
p.Thr29725fs
HFA
46
DCM


179410799
c.90241C > T
p.Gln30081X
MHQ
12
DCM


179408989
c.91043delA
p.Asn30348fs
TSFDC033
434
DCM


179408239
c.91537_91538insA
p.Thr30513fs
UK
E4
DCM


179404491
c.93376_93377delAG
p.Arg31126fs
UK
G7
DCM


179404286
c.93583C > T
p.Arg31195X
UK
G9
DCM


179403522
c.94111A > T
p.Lys31371X
MHX
11
DCM


179401029
c.95522C > A
p.Ser31841X
UK
C9
DCM





*Mutations are annotated using Human Genome Variation Society guidelines: available on the world wide web at http://www.hgvs.org/mutnomen/.













TABLE 9







TTN splicing mutations













Position in
Splicing



Chr 2

splice site
score















Position
Nucleotide
5′
 3′
change
Amino Acid
Pedigree
ID
Diagnosis


















179659280
c.1246 − 3delT

−3
−2.2
p.Val416
pv
75
Control


179647331
c.3101 − 2A > T

−2
−8.37
p.Val1034
N
050
Control


179558736
c.30476 − 1G > A

−1
−8.75
p.Gly10159
D13KD
1
DCM


179535816
c.34186 + 1G > T
+1

−8.5
p.Val11396
SS
333
HCM


179516991
c.34690G > T
+1

−11.03
p.Val11564Phe
12s
C11
Control


179506964
c.35635G > C
−1

−14.19
p.Val11879
UK
B12
DCM


179506779
c.35635 + 1G > A
+1

−8.18
p.Val11879
HFA
26
DCM








HFA
41
DCM


179498055
c.38024 − 2A > G

−2
−7.95
p.Ala12675
MIY
11
DCM


179477885
c.44725 + 2delT
+2

−12.92
p.Asp14909
MDD
22
DCM








MIV
14
DCM


179472127
c.48364 + 1G > T
+1

−8.5
p.Asp16122
HFA
71
DCM


179467006
c.50197 + 3A > G
+3

−5.04
p.Glu16733
12s
G11
Control


179466858
c.50346 + 3A > G
+3

−3.42
p.Lys16782
TSFDC023
300
DCM


179457392
c.54422 − 5T > A

−5
−2.34
p.Glu18141
UK
B1
DCM


179457005
c.54704 − 1G > A

−1
−8.75
p.Asp18235
UK
B12
DCM


179456704
c.55003 + 1G > A
+1

−8.18
p.His18335
MGR
11
DCM


179444661
c.62425 + 5G > A
+5

−12.17
p.Gln20809
MAM
12
DCM


179443339
c.63405A > G
−2

−2.67
p.Thr21135
MIS
11
DCM


179442324
c.63901 + 5G > C
+5

−5.38
p.Tyr21301
MCL
15
DCM


179441649
c.64489 + 1G > A
+1

−8.18
p.Gly21497
UK
B2
DCM


179424036
c.81898 + 2T > A
+2

−8.18
p.Glu27300
MIA
1
DCM


179410544
c.90493 + 2insCCT
+2

−32.13
p.Thr30165
SS
723
DCM


179406779
c.92569 + 1G > C
+1

−8.27
p.Arg30857
HFA
66
DCM








HFA
83
DCM


179402067
c.94942 + 2T > C
+2

−7.75
p.Asp31648
pv
35
Control





*Mutations are annotated using Human Genome Variation Society guidelines: available on the world wide web at http://www.hgvs.org/mutnomen/. Amino acid indicated is the residue juxtaposed to the aberrant splice signal.













TABLE 10 







CNV validation primers











SEQ


Name
Sequence
ID NO





TTN_37_intF1
TGAGGAGCTGTAAGAGAATGTCATCAGA
720





TTN_i21_R1
AGTGCTGGCATTACCACTCCTAGTT
721
















TABLE 11







Clinical characteristics of DCM-A probands






























Age at








Current
Age





VAD,





age
of
Fam-




Tx,




BSA
(yr) or
Dx
ily
LVEF
FS
LVEDD
NYHA
Death


Subject
Sex
(m2)
death
(yr)
Hx
(%)
(%)
(mm)
class
(yr){circumflex over ( )}
Comments□
Gene
Mutation(s)























HFA-42
M
1.84
66
54
No
47.5
27.9
61
4

CHF, Embolic
TTN
p.Gln4249X













CVA, ICD


HFA-9
M
1.89
55
42
Yes
15
15.5
84
3

CHF, ICD(50)
TTN
p.Ser6394fs













NSVT


D13KD-1
F
1.99
51
39
Yes
15
24.3
70
4

Nl Cors
TTN
p.Gly10159


MIY-11
M
2.14
42
41
Yes
20
15.8
65
3


TTN
p.Ala12675


HFA-26
F
1.56
51
45
No
40
29.2
48
2


TTN
p.Val11879


HFA-41
M
2.14
60
57
No
45
24
58
1

Episodic
TTN
p.Val11879













severe













dypsnea


MDT-
F
1.66
30
19
Yes
20
11
52
3

ICD(28)
TTN
p.Ala12873fs


11


MEQ-
M
1.6
25
22
Yes
45
19.3
57
1

Familial
TTN
p.Pro13298_Thr17642dup


132










Screening


HFA-68
F
1.83
40
37
Yes
45
21.4
56
1

CHF, ICD(38)
TTN
p.Glu14779fs













NSVT


MDD-
F
1.88
73
57
Yes
20
18
58
2

ICD(70)
TTN
p.Asp14909


22


MIV-14
M
1.9
50
26
Yes
20
9.5
63
3
49
AF, CHF (35),
TTN
p.Asp14909













ICD(47)


HFA-71
M
2.05
61
40
Yes
15
11.3
80
4

CHF, ICD(56),
TTN
p.Asp16122













NSVT


HFA-63
M
2.25
32
29
Yes
15
15.9
63
4

CHF, ICD,
TTN
p.Glu17978fs













NSVT


MGR-
F
1.58
59
53
Yes
20
9.7
62
2.5

CHF(53)
TTN
p.His18335


11


MEH-
M
2.36
28
25
Yes
11
NA
NA
4
28
CHF, NSVT
TTN
p.Cys18789X


11


MDJ-21
M
2.13
35
NA
NA
40
21
64
3

ICD, NSVT
TTN
p.Arg18858X


MEW-
M
1.97
37
20
Yes
25
NA
50
4
31, 37

TTN
p.Arg18985X


11


MAO-
F
1.6
36
17
Yes
37.5
22.4
63
2

ICD(33)
TTN
p.Trp976Arg/


92












p.Arg19560X


MAM-
M
1.79
23
23
Yes
32.5
25
68
4
23
ICD, NSVT,
TTN
p.Gln20809*


12










SMD


MIS-11
F
1.8
62
59
Yes
20
25.5
61
2

AF, CHF
TTN
p.Thr21135


MCL-15
F
1.53
54
24
Yes
28
18
51
3

ICD(35)
TTN
p.Tyr21301


MHG-
M
1.98
52
31
Yes
29
14.3
77
4
52
AF, CHF,
TTN
p.Ala22353fs


23










ICD(46)


MEK-
M
1.94
24
15
Yes
NA
17
77
4
15, 24
CHF
TTN
p.Pro22582fs


111


HFA-57
F
1.98
55
48
Yes
22.5
13.7
51
2
55
AF, CHF, Nl
TTN
p.Gln24059fs













Cors


SS287-
F
1.97
54
45
Yes
45
36
55
1

Familial
TTN
p.Ser24241fs


15










Screening;













RVD, SVT


MID-1
M
1.83
40
37
Yes
34
15.7
51
2

Familial
TTN
p.Gln25689X













Screening


SS725
M
2.28
62
51
Yes
20
9.5
63
4

CHF, ICD(61)
TTN
p.Gln25689X


MGW-
M
NA
40
35
Yes
12.5
13.3
98
4
40
CHF, ICD
TTN
p.Arg26949X


11


MIP-13
M
2.33
41
33
Yes
30
16.5
51
2

CHF, ICD(33)
TTN
p.Trp27147X













NSVT; Nl Cors


MIA-1
M
2.35
30
14
Yes
32.5
25
68
2

ICD, NSVT
TTN
p.Glu27300


MBG-
M
2.21
34
35
Yes
20
NA
NA
3

CHF, ICD
TTN
p.Glu29510X


121


HFA-46
M
1.9
66
53
Yes
20
57
62
3

AF, AI, ICD
TTN
p.Thr29725fs


MHQ-
M
2.07
42
38
Yes
17.5
9.7
72
1

CHF, ICD(41)
TTN
p.Gln30081X


12


SS723
M
NA
56
51
Yes
15
38
67
3

AF, ICD(52)
TTN
p.Thr30165


HFA-83
M
1.98
45
40
Yes
20
15.2
66
4

AF, CHF, ICD
TTN
p.Arg30857


HFA-66
M
2.04
29
28
Yes
22.5
13
77
3
29, 32
ICD(29), NSVT
TTN
p.Arg30857


MHX-
M
2.24
58
52
Yes
32.5
8.2
61
3.5
58
CHF
TTN
p.Lys31371X


11


HFA-10
M

64
57
NA
15
5.9
84
2
64
ICD(58)


HFA-30
M

62
55
No
25
22.6
62
2
60
IC(54) NSVT


HFA-37
M

38
18
No
35
22.6
53
1

NSVT


HFA-43
M

47
37
No
45
22.2
54
1


HFA-50
M

51
47
No
20
16.7
60
3

ICD(49)


HFA-51
M

43
33
No
30
18.3
71
2

ICD


HFA-53
F

51
47
Yes
55
25
44
1

Familial













Screening; Abn













LV wall













motion; Nl













Cors; NSVT;













ICD(47)


HFA-56
M

37
33
Yes
35
20.4
54
1


HFA-61
F

63
47
Yes
37.5
8.8
57
4

ICD(62)


HFA-67
M

49
43
NA
15
14.5
69
4
45
CAD, ICD(41)


HFA-70
F

74
58
Yes
25
12.7
71
3

AF, ICD, NSVT


JG-32
F

51
43
Yes
45
38
60
3

ICD(45) SM, Nl













Cors


MAB-
F

31
22
Yes
27
21.2
66
3
33
ICD(33)


17


MAE-
M

35
34
Yes
25
57
15.8
3
58
AF, ICD(35),


3111










NSVT


MAG-3
F

69
50
Yes
35
18
56
3


MAK-
M

38
61
Yes
40
22
70
3

ICD(39)


13


MBS-1
M

65
65
Yes
42.5
30.8
65
1

AF


MCH-
M

12
12
Yes
30
15.4
65
1
13
ICD(12)


123


MDK-
M

53
NA
Yes
52
NA
NA
NA


11


MDU-
F

43
31
Yes
NA
NA
NA
NA


111


MDX-
F

40
37
Yes
40
36
61
1

Family


11










Screening


MEE-11
M

44
32
Yes
40
22.6
62
2

ICD(34) VT


MEI-
M

27
17
Yes
NA
NA
NA
NA


112


MEM-
M

18
10
Yes
NA
NA
NA
4
12
CHF


11


MEN-
M

31
12
Yes
42.5
22.6
62
1

AF, ICD, NSVT


111


MEP-
M

46
23
Yes
45
22
61
NA


111


MER-
M

54
38
Yes
18
NA
60
3

ICD(38)


11


MEX-1
M

53
NA
Yes
9
NA
70
NA

VT, CAD


MGL-
F

27
25
Yes
30
16.1
62
2
27
SMD


11


MGM-1
F

73
36
Yes
30
14
50
2

AF, ICD, NSVT


MGP-1
M

58
57
Yes
44
29.7
64
2

ICD(58)


MGS-
M

30
25
No
50
NA
59
1

Nl Cors;


111










Syncope


MGU-
M

41
NA
Yes
NA
NA
NA
NA
32


111


MGY-
M

35
34
Yes
15
12.3
65
NA


123


MHA-
F

37
34
Yes
40
31.5
54
3
34
ICD(34) NSVT


145


MHE-
M

54
NA
Yes
22.5
12.5
64
4
54
ICD, AVB


113


MHF-
F

43
40
Yes
15
8.9
56
1

AF, ICD(41)


11


MHI-11
M

70
56
Yes
NA
NA
NA
3
70
AF


MIF-14
M

42
42
Yes
35
30.6
62
2


MIJ-1
M

49
29
Yes
15
5.2
77
4
47
ICD(40) NSVT


MIK-
F

38
NA
Yes
40
27.6
58
1
37
SCD; ICD(37)


111


MIM-
F

49
33
Yes
20
13.2
68
4
46
ICD(45)


11


MI0-13
M

28
25
Yes
30
NA
55
2

Familial













Screening


MJ-11
M

26
17
Yes
NA
NA
69
4
26
WPW, RVF


MT13
M

48
18
Yes
30
31
56
1

Familial













Screening;













AVB; SVT; LV













biopsy,













myocarditis













negative


MW-11
M

61
43
Yes
20
12.5
64
2

ICD(56)


MYC-
M

53
43
Yes
15
9.9
71
1

AF, ICD(50)


11


SS126
M

38
28
Yes
38
18.8
64
1

ICD


SS705
F

42
33
Yes
15
5
59
4
41
MR


SS719
F

60
45
Yes
20
6.3
64
4

CHF; Nl Cors


SS720
M

67
52
NA
25
10.3
68
2

ICD(66), NSVT


SS731
M

65
65
No
25
16.6
66
3

AF, MR, Nl













Cors


MHM-
M

29
19
Yes
40
28.8
59
1


22


HFA-65
F

54
51
Yes
42.5
14.8
54
1

ICD(50)


MHO-4
M

60
54
Yes
20
13.6
66
1

ICD(54),













NSVT, mild













CAD





BSA, body surface area (m2) is provided for subjects with TTN mutations;


Age of Dx, age of diagnosis;


Family Hx, Familial history of DCM.



Echocardiographic measurements of left ventricular ejection fraction (EF), fractional shortening (FS) and left-ventricular end-diastolic diameter (LVEDD) obtained at time of diagnosis.



NA, not available.



New York Heart Association classifications (NYHA) range from 1 to 4.



{circumflex over ( )}Earliest ages at which subject received a ventricular assist device (VAD) or a cardiac transplant (Tx), or age at death.


|| Clinical data are denoted by: Abn LV wall motion, abnormal left ventricular wall motion identified by echocardiography; AF, atrial fibrillation; AI, aortic insufficiency; AVB, atrio-ventricular block; CHF, hospitalized for congestive heart failure; CVA, cerebral vascular accident; Familial screening, clinical evaluations were prompted by overt DCM in a first-degree relative; ICD, implanted cardiac defibrillator implanted at age (parentheses); MR, mitral regurgitation; PCM, pacemaker; Nl cors, normal coronary artery anatomy defined by cardiac angiography; NSVT, non-sustained ventricular tachycardia; WPW, Wolf Parkinson White; RVD, right ventricular dilation; RVF, right ventricular heart failure, SMD, skeletal muscle disease.


□TTN mutations are detailed in Tables 8 and 9.


*denotes homozygous mutation.













TABLE 12







Clinical characteristics of DCM-B probands




























Age














at





Current





VAD,





age
Age




Tx,




BSA
(yr) or
of Dx
Family
LVEF
LVEDD
NYHA
Death


Subject
Sex
(m2)
death
(yr)
Hx
(%)
(mm)
Class
(yr){circumflex over ( )}
Comments□
Gene
Mutation(s)






















UK-H11
F
1.74
40
32
No
20
68
3
32

TTN
p.Cys13771X


UK-G1
M
1.9
49
46
NA
35
74
4
49

TTN
p.Phe15108fs


UK-A3
M
1.67
30
18
No
24
79
2
19

TTN
p.Gly16189X


UK-B1
M
2.1
71
50
No
50
55
4
60
RVF
TTN
p.Glu18141












RVEF = 14%


UK-B12
M
1.78
39
30
No
6
81
4
31

TTN
p.Val11879














p.Asp18235


UK-A10
M
1.88
55
49
No
29
64
4
55

TTN
p.Arg20858X


UK-B2
M
NA
42
37
No
23
70
4
42

TTN
p.Gly21497


UK-H2
M
2.09
51
39
Yes
18
80
3
41

TTN
p.Lys21640fs


UK-C8
M
NA
55
37
NA
11
76
4
55

TTN
p.Glu21956fs


UK-C6
M
1.83
41
31
No
29
67
3
31

TTN
p.Ser27179fs


UK-B6
M
2.19
38
30
NA
38
82
4
38

TTN
p.Tyr27567X


UK-E12
M
1.8
57
49
No
21
78
4
51
ICD
TTN
p.Tyr28326fs


UK-C10
M
NA
38
29
No
34
73
4
30

TTN
p.Arg29415X


UK-E4
M
NA
42
32
No
18
61
4
32

TTN
p.Thr30513fs


UK-G7
F
1.78
63
53
Yes
14
59
3
54

TTN
p.Arg31126fs


UK-G9
M
2.2
50
43
No
17
70
3
50

TTN
p.Arg31195X


UK-C9
M
NA
62
49
Yes
32
66
4
54

TTN
p.Ser31841X


UK-A2
M

60
49
NA
44
69
3
49


UK-A4
M

40
29
No
15
87
3
29
AVR


UK-A5
M

40
30
No
16
72
3
31


UK-A7
M

26
17
NA
NA
NA
3
17


UK-A8
M

50
40
No
17
81
3
45


UK-A9
M

69
48
No
20
69
3
60


UK-A12
M

58
48
NA
18
78
2


UK-B3
M

44
32
NA
33
72
4
44


UK-B7
M

52
41
NA
19
80
4
52


UK-B8
M

63
50
NA
8
90
3
54
AF


UK-B9
F

50
49
NA
26
66
4
50
ICD


UK-B10
M

67
53
NA
18
57
3
59
PCM


UK-B11
F

57
51
Yes
34
62
3
52
PCM


UK-C1
F

64
51
Yes
31
61
3
53


UK-C3
M

46
29
NA
27
79
3
35


UK-C4
F

66
55
NA
32
69
2


UK-C5
M

57
46
NA
24
66
3
47


UK-C7
M

60
43
NA
32
78
4
59


UK-C12
F

28
28
NA
15
64
3
28


UK-D1
M

62
46
No
37
78
3
51


UK-D3
F

44
33
Yes
44
69
3
34


UK-D4
M

47
36
No
35
63
3


UK-D6
M

59
57
NA
44
75
4
59
AF


UK-D8
M

43
38
NA
9
98
4
43


UK-D9
M

32
13
NA
20
54
3
23

TTN


UK-D10
M

28
18
No
5
59
3
20


UK-D11
F

65
54
NA
6
74
3
57


UK-D12
M

44
39
Yes
12
76
3
39, 44


UK-E3
F

43
27
No
20
71
3
32


UK-E6
M

69
58
NA
11
74
3
59
PCM


UK-E9
M

50
17
NA
21
65
3
41
PCM


UK-E7
M

32
26
NA
10
91
4
32


UK-E10
M

61
56
NA
30
88
4
61
AF


UK-E11
M

60
52
YES
18
80
3
52


UK-F1
M

71
59
Yes
27
71
3
60
PCM


UK-F3
M

42
29
NA
13
62
4
42


UK-F4
M

57
52
No
45
71
4
57


UK-F5
F

27
13
No
20
94
3
16
PCM


UK-F6
M

55
21
NA
20
87
3
45


UK-F7
F

64
54
No
49
57
2


UK-F9
M

51
50
NA
23
77
3
51


UK-F10
M

50
36
NA
21
91
2

AF, ICD


UK-F11
M

52
52
NA
23
73
4
52


UK-G2
F

44
31
NA
30
63
2


UK-G3
M

58
56
NA
40
76
4
58


UK-G5
F

17
16
No
44
77
4
17


UK-G6
M

46
45
NA
34
67
4
46


UK-G8
F

61
47
No
30
61
3
57


UK-G11
M

67
56
No
28
80
3
59


UK-H3
M

62
51
No
17
72
3
52


UK-H4
M

57
47
No
11
78
3
47


UK-H8
F

66
56
No
30
71
3
57


UK-H9
M

64
54
No
48
93
3


UK-H12
M

46
46
No
10
NA
4
46





Footnotes for symbols ({circumflex over ( )}||□) and abbreviations are defined in Table 11.


Additional abbreviations used: AVR, aortic valve replacement;


RVEF, right ventricular ejection fraction.













TABLE 13







Clinical characteristics of DCM-C probands






























Age















at





Current






VAD,





age






Tx,





(yr) or
Age
Family
LVEF
FS
LVEDD
NYHA
Death


Subject
Sex
BSA (m2)
death
of Dx (yr)
Hx
(%)
(%)
(mm)
class
(yr){circumflex over ( )}
Comments□
Gene
Mutation(s)























TSSDC011-
M
2.1
53
45
No
37
12
82
2


TTN
p.Arg2083fs


450


DNFDC144-
F
2.18
54
32
Yes
45
NA
57
2


TTN
p.Trp16359X


05-0444


TSFDC023-
F
1.75
38
20
Yes
26
15
60
1


TTN
p.Lys16782


300


DNFDC116-
M
1.96
33
21
Yes
40
15
54
1


TTN
pArg17295X


04-1545


TSFDC050-
M
1.74
53
35
Yes
24
13
70
3


TTN
p.Arg17470X


115


TSFDC017-
M
2.13
72
65
Yes
40
22
59
1


TTN
p.Glu17715fs


253-2


DNFDC103-
M
2.48
57
45
Yes
30
10
74
2.5


TTN
p.Glu17783X


03-0941


DNFDC142-
M
1.79
30
23
Yes
20
15
69
2.5


TTN
p.Arg18985X


05-0569


DNFDC088-
F
2.04
40
38
Yes
25
NA
50
2


TTN
p.Trp26632X


02-1900


DNFDC081-
M
2.13
45
43
Yes
20
13
63
4
45

TTN
p.Lys27016X


02-1564


TSSDC019-
F
1.45
67
60
No
21
13
70
4


TTN
p.Trp29318X


268


TSFDC002-
M
1.79
57
43
Yes
29
10
72
1


TTN
p.Arg29415X


23


TSFDC033-
M
2.0
40
19
Yes
24
10
72
4


TTN
p.Asn30348fs


434


TSFDC004-
M

41
37
Yes
18
10
57
3
41


118


DNFDC050-
F

36
29
Yes
43
22
56
2


02-2210


DNFDC096-
F

37
36
No
37
NA
NA
NA


03-0005


TSFDC049-4
M

64
45
Yes
29
18
79
2


TSXLC002-
M

38
19
Yes
25
7
83
2


22


TSFDC007-
F

42
20
Yes
20
11
73
2


28


TSFDC047-
M

35
30
Yes
39
12
52
4
35


120


TSXLC001-
M

31
31
Yes
28
15
60
4
42


131


TSFDC010-
M

46
36
No
29
15
71
3


134


TSFDC003-
F

51
54
Yes
34
NA
62
1


136


TSLVN002-
M

24
18
Yes
17

74
1


156


TSLVN001-
F

57
41
Yes
35
18
56
1


172


TSFDC013-
F

26
25
Yes
NA

51
4
26


206


TSFDC014-
F

60
43
No
41
21
62
1


208


TSFDC015-
M

49
34
Yes
38
24
63
1


230-2


TSFDC016-
M

59
44
Yes
32
20
66
2


254-2


TSFDC026-
M

27
14
Yes
27
16
53
4
27


311


TSFDC029-
F

59
46
Yes
42
20
56
2


409


TSFDC027-
M

52
35
Yes
22
17
69
4
52


419


TSFDC032-
M

39
22
Yes
23
8
76
3


430


TSFDC031-
M

48
44
Yes
17
5
75
4
48


438


DNFDC020-
M

60
55
Yes
45
20
59

3


99-0824


DNFDC010-
M

58
56
Yes
45
NA
71
3


01-0045


DNFDC053-
M

42
41
Yes
60
34
60
1

Abn LV wall


01-0081










motion


DNFDC058-
F

37
31
Yes
32
NA
NA
2


01-0341


DNFDC055-
F

39
38
Yes
10
5
90
2


01-0439


TSFDC036-
M

50
40
Yes
48
27
63
4
50


01-0729


TSFDC039-
F

55
54
Yes
23
26
69
4
55


01-0731


DNFDC054-
M

70
56
Yes
28.5
NA
NA
2


01-0937


DNFDC004-
M

35
21
Yes
NA
NA
NA
2


01-1624


DNFDC065-
M

66
55
Yes
37
18
55
2


01-2260


DNFDC066-
F

39
34
Yes
50
23
57
1


01-2393


DNFDC068-
M

47
45
Yes
50
18
67
4
47


02-0053


TSFDC040-
M

37
28
Yes
52
25
61
1


02-0169


TSFDC041-
M

NA
45
Yes
24
6
63
2


02-0176


TSFDC042-
F

60
46
Yes
22
15
72
4
46


02-0191


TSFDC038-
F

62
48
Yes
46
20
58
1


02-0225


DNFDC071-
F

76
58
Yes
45
29
41
2


02-0394


DNFDC073-
M

19
18
Yes
25
11
56
2


02-0502


DNFDC072-
M

44
32
No
19
12
89
4
44


02-0503


DNFDC069-
F

36
28
Yes
30
22
62
2


02-0646


DNFDC074-
F

53
46
Yes
35
NA
NA
2


02-0911


DNFDC077-
M

46
44
Yes
10
NA
NA
46


02-0954


TSFDC018-
M

74
33
Yes
40
19
61
2


02-1027


TSFDC045-
M

60
49
Yes
40
20
64
2


02-1089


TSFDC025-
M

41
28
Yes
22
21
70
2


02-1092


TSFDC044-
M

34
26
Yes
43
18
56
1


02-1097


DNFDC078-
M

48
39
Yes
20
NA
NA
3
48


02-1192


DNFDC079-
F

34
33
Yes
20
15
55
3


02-1517


DNFDC029-
F

76
62
Yes
25
13
NA
2


02-1532


DNFDC083-
F

69
38
Yes
30
20
74
2


02-1566


DNFDC030-
M

40
29
Yes
21
9
75
3


02-1612


DNFDC087-
F

48
22
Yes
11
9
81
3


02-1758


DNFDC013-
F

44
37
Yes
37
11
62
2


02-1902


DNFDC042-
F

53
52
Yes
20
11
64
4
53


02-2037


DNFDC034-
M

64
53
No
35
NA
74
3


02-2053


DNFDC090-
M

47
47
Yes
41
22
84
2


02-2104


DNFDC089-
F

38
31
Yes
25
14
80
3


02-2151


TSFDC009-
M

67
44
Yes
26
13
83
1


02-2276


TSFDC022-
F

60
44
No
40
28
56
1


02-2304


DNFDC092-
F

39
38
Yes
45
NA
NA
2


02-2349


DNFDC091-
M

43
35
Yes
31
20
54
2.5


02-2555


DNFDC099-
F

32
30
Yes
17
16
66
3


03-0432


DNFDC100-
M

49
39
Yes
27
13
80
3
49


03-0433


DNFDC108-
F

61
49
Yes
29
14
NA
2


03-2056


DNFDC016-
F

38
36
Yes
15
15
60
3


04-0243


DNFDC109-
F

59
58
Yes
30
13
67
2


04-0326


DNFDC014-
M

50
34
Yes
17
NA
NA
3


04-0567


DNFDC114-
F

42
41
Yes
40
23
47
2


04-0871


DNFDC112-
F

55
38
Yes
19
12
66
3


04-0906


DNFDC117-
M

60
50
Yes
15
11
55
2.5


04-1674


DNFDC123-
F

65
63
Yes
30
26
49
2.5


05-0110


DNFDC133-
M

50
47
No
17
5
66
2


05-0199


DNFDC039-
M

54
50
Yes
34
17
64
2.5


05-0234


DNFDC131-
F

28
24
Yes
25
11
64
3


05-0381


DNFDC139-
M

45
30
No
33
NA
NA
3


05-0394


DNFDC147-
F

36
33
Yes
25
NA
64
2


05-0548


DNFDC155-
F

49
43
Yes
47
21
50
2


05-0698


DNFDC141-
F

22
18
Yes
18
14
64
2.5


05-0716


TSAR004-6
M

73
53
Yes
50
32
61
1


TSAR003-
M

14
12
Yes
31
7
56
2
14


35-2


TSAR005-
M

37
19
Yes
37
18
65
1


125


TSFDC001-
M

51
29
Yes
21
11
87
1


98-38


TSSDC128-
M

42
38
No
37
24
59
4
42


220


TSSDC012-
M

69
53
Yes
38
19
64
2


273


TSSDC116-
M

24
23
No
18
13
61
4
24


292


TSSDC020-
F

56
39
No
33
20
59
2


395


TSSDC009-
F

59
46
Yes
18
16
89
4
59


398-2


TSSDC024-
M

50
38
No
17
15
89
4
50


401


TSSDC026-
F

71
51
No
25
10
68
2


433


TSSDC027-
M

50
44
No
29
9
78
2


435


TSSDC028-
M

32
25
No
22
10
79
2
32


436


TSSDC030-
M

61
44
No
28
10
79
3


439


TSSDC031-
M

69
56
No
29
19
67
2
69


441


TSSDC032-
M

74
57
Yes
37
11
77
2


442


TSSDC033-
M

58
46
No
26
9
64
1


443


TSSDC002-
M

51
39
No
17
6
77
4
51


445-2


TSFDC005-
F

52
34
Yes
35
16
60
4


127


TSFDC012-
M

58
41
Yes
61
24
50
1

Familial


148-2










Screening;













NSVT, AF


TSSDC004-
M

76
55
No
45
16
83
3


447


TSSDC005-
M

65
53
No
NA
12
67
4
65


448


TSSDC007-
M

72
53
No
25
13
70
2


449


TSSDC014-
M

72
48
No
42
14
65
3
72


451


TSSDC015-
M

48
48
No
40
30
63
1


452


DNFDC052-
M

21
17
Yes
NA
27
57
2
21


01-0131


DNFDC057-
F

59
48
Yes
30
25
NA
2
59


01-0987


DNFDC084-
F

52
42
No
10
NA
69

2


02-1567


DNFDC095-
M

54
44
Yes
10
6
78
2


02-2523


DNFDC113-
F

45
44
Yes
25
NA
NA
NA
NA


04-0881


DNFDC138-
F

27
21
No
20
5
53
3


05-0395


DNFDC080-
M

52
33
Yes
30
7
53
3
52


02-1405


DNFDC094-
M

68
52
yes
21
20
63
1


02-2524


DNFDC101-
F

61
50
No
25
14
60
2


03-0942


DNFDC104-
F

37
27
No
30
16
60
2


03-2029


DNFDC110-
M

59
58
No
28
12
58
2


04-0347


DNFDC119-
M

55
49
Yes
15
23
70
2


04-1877


DNFDC127-
M

55
44
No
18
NA
NA
3


05-0160


DNFDC140-
F

39
31
No
40
25
60
2


05-0355


DNFDC075-
M


31
Yes
52


02-0970


DNFDC136-
F


23
No
50


05-0301


TSSDC010-
M


56
No
49


01-0745-2


DNFDC102-
F


27
No
48


03-2097


TSFDC067-
M


13
No
47


02-2315


DNFDC093-
M


58
No
47


03-0646


DNFDC047-
F


49
No
46


02-0952


TSFDC054-
M


27
No
46


03-0818


DNFDC107-
F


40
Yes
42


03-1827


DNFDC026-
M


63
No
36


02-1565


TSFDC069-
F


50
No
35


02-2322


DNFDC059-
M


58
No
33


01-2291


TSSDC058-
M


20
No
30


03-0817


DNFDC118-
M


22
No
29


04-1787


TSSDC056-
F


47
No
28


02-0174


TSSDC060-
M


49
No
13


01-0725


DNFDC008-
F

66
61
Yes
55
29
NA
4
66
Rapid


02-2451










Progression


DNFCD003-
F

59
41
Yes
56
35
NA


Familial


02-01704










Screening





Footnotes for symbols ({circumflex over ( )}||□) and abbreviations are defined in Table 11.


Additional abbreviations used: AVR. aortic valve replacement;


RVEF, right ventricular ejection fraction.













TABLE 14







Clinical findings in DCM families with TTN truncating mutations*





















Current
Age at



Age of








age (yr)
diag-



VAD/





or
nosis
FS
LVEDD
NYHA
Tx/Death

Clinical
TTN


Pedigree
ID
Sex
deatht
(yr)
(%)
(mm)
class
(yr){circumflex over ( )}
Comment□
Status
Genotype(s)□





















DNFDC081
02-
M
45
43
13
63
4
Tx(45)
EF = 20%
A
p.Lys27016X



1564



02-
M
33
33
30.2
53
1

IVCD
A
+



1663


DNFDC088
02-
F
40
38
16
50
2

EF = 25%
A
p.Trp26632X



1900



2395
M
70
61
39
54
1


I




1901
F
58
58
33
55
1

Diastolic
I
+











Dysfunction,











TWA



05-
M
47
45
NA
NA
4
Tx(47)
ST,
A
+



0427







RBBB,











LAFB


DNFDC103
03-
M
57
45
10
74
2.5

TWA,
A
p.Glu17783X



0941







1AVB











EF = 30%



05-
M
51
48
NA
NA
2

EF = 10%
A
+



1266


DNFDC142
05-
M
30
23
15
69
2.5

TWA,
A
p.Arg18985X



0569







LAFB,











IVCD,











EF = 20%



0568
F
53
53
NA
NA
2


I
+


MAO
92
F
36
17
22.4
63
2

EF = 37%
A
p.Trp976Arg











ICD(33)

p.Arg19560X



9
M
49
47
32
50
1
49
Septic
U
p.Trp976Arg











Shock



90
F
49
NA
NA
NA
NA


I
p.Arg19560X



91
F
40
40
35.3
51
1


I
p.Arg19560X



93
M

19


1


I
pArg19560X


MDD
22
F
73
57
18
58
2

EF = 20%
A
p.Asp14909











ICD(70)



21
M
31
31




CHF
A
NA



23
F
72
58




EF = 20%
A
NA(+)



231
M
45
45




AF(39)
I
+



2311
M
18
18
35.7
56
1

AF(16)
I
+



2312
M
16
16
33.3
48


NSR
I



MEK
111
M
24
17
17
77
4
15, 24
Tx(15)
A
p.Pro22582fs



1
F
72
50
34
50
NA

STTWA;
A
+











Q-











waves, CHF



11
M
41
25
8
75
4
36
1AVB,
A
+











LAFB,











TWA











Tx(36)



110
F
43
NA
NA
NA
NA


U




112
F
19
NA
NA
NA
NA


I
+



12
M
33
30
NA
NA
4
33
CHF(33)
A
NA


MEQ
132
M
25
22
19.3
57
1

SB,
A
p.Pro13298_Thr17642dup











EF = 45%



12
M
56
33
NA
NA
4
50
NSVT,
A
NA











CHF(49)











Tx(50)



13
M
46
41
NA
NA
NA
42
SCD(46)
A
NA(+)



131
M
33
33
38.8
54



I



MEW
11
M
37
20
NA
50
4
31
EF = 25%
A
p.Arg18985X











Tx(30)



1
M
63
62
NA
NA
4
43
Tx(43)
A
NA(+)



21
M
32
23
NA
NA
NA

ICD
A
+


MHG
23
M
52
31
14.3
77
4
52
AF,
A
p.Ala22353fs











EF = 29%











ICD(46)



1
F
57
NA
NA
NA
NA
58
Lung Ca
U
NA



12
M
65
62
43.2
44
1

SB
U




15
F
57
41
32.8
61
2

Morbid
I












Obesity











Type II











DM, PCM



152
F
27
27
50
46



I




2
F
66
59
NA
NA
NA
66
CHF
A
NA(+)



21
F
63
48
31.9
38.3
2

LBBB,
A
+











ICD



211
M
41
37
28.2
44.7



I
+



212
M
41
37
32.8
45.4



I
+



22
F
61
42
30.1
55.2
2

1AVB,
A
+











LBBB











EF = 22%



221
F
35
32
30.1
52.2
1

NSR
I
+



222
M
32
29




NSR
I




231
M
33
33




NSR
I




3
F
25





Leukemia
I
NA



31
M
52
52
46.4
48.1
1

IVCD
U




311
F
28
28
38
47
1


I




4
F
55
50
NA
NA
NA
54
CHF
A
NA(+)



41
F
56
NA
NA
NA
NA


I




43
F
50
46
12.2
57.3
3

NSVT,
A











STTWA











ICD(50)



46
F
48
48
32
50
1


I




47
M
42
NA
26
53.5
1

NSR
A
+



48
M
42
43
16.7
72
NA

ICD(42)
A
NA



6
M
41
37
NA
NA
3
41
NSVT,
A
NA











CHF(37)



61
M
49
49
30.5
40.6
NA

NSR
U




62
F
47
47
41.7
34.3
NA

TWA
U




63
F
45
45
24.7
38.1
NA

TWA
I




64
M
42
NA
38.1
40.4
NA

NSR
U




7
M
61
56
NA
68
NA
61
ST, CHF
A
NA(+)



70
F
65
NA
NA
NA
NA


U




71
F
45
45
34.9
47
1

NSR
U




72
M
44
44
30.9
57.2
1

NSR
A
+



73
F
43
NA
36.3
38.6
1

SB, long
I












QTc,











TWA



74
M
42
NA
19.2
49.9
NA

SB
I
+



8
F
63
59
NA
NA
2
63
Long
A
+











QTc,











TWA,











CHF











Nl Cors



82
M
40
31
9.6
55.3
2

LVH,
A
+











LAFB,











TWA



83
F
42
38
43.2
45.5
2


I
+



84
F
48
44
40.6
43.3
2

SB, TWA
I
+



9
F
66
62
41.6
39.9
1

NSR
U




98
M
53
NA
NA
NA
NA
53
SCD(53)
I
NA



981
F
42
39
33
43



U




99
F
59
56
33.8
41.7
1

PVC, MR
I
+


MHQ
12
M
42
38
9.7
72
3

EF = 17.5%
A
p.Gln30081X











CHF, NSR











ICD(41)



1
F
64
64
37.2
43
1

PAC
A
NA(+)



11
F
44
38
26.5
49
1


U



MHX
11
M
58
52
8.2
61
3.5

EF = 32.5%
A
p.Lys31371X











CHF,











IVCD



1
M
59
50
NA
NA


DCM CHF
A
NA(+)



10
F
90
90





U




12
F
59
NA
41.3
46
1

TWA
U




13
M
51
50
27
63
NA

PVC,
A
+











LVH,











RBBB,











ICD(51)



14
M
53
NA
36
50
NA

NSR
U




15
F
61
54
40
48


ICD(56)
I
+











MRI:GdE



16
M
63





Nl Echo
U




17
M
61
56
34
58
4
61
CHF SCD
A
NA



18
F
66
52
NA
NA
NA

CHF; ICD
A
NA


MID
1
M
40
37
15.7
51
2

PVC,
A
p.Gln25689X











TWA,











EF = 34%



2
M
48
41
19.6
56
1


A
+



21
M
19
19
25.5
55
1

JR
A
+



22
M
17
NA
38.3
60
1


A
+


MIP
13
M
41
33
16.5
51
2

EF = 25%
A
p.Trp27147X











CHF,











NSVT,











ICD(33)



1
M
65
42
19.7
76
4
50 (Tx)
ST,
A
+











1AVB,











ICD



11
M
45
44
9
64
2

STTWA,
A
+











ICD



111
M
27
27


1


I




112
M
15
15
27.1
48
1

NSR
I
+



113
M
12
12
33.3
48
1

LVH, long
I
+











QTc



12
F
44
44
27.7
47
1

STTWA,
I
+











PVC



121
M
19
19
34.6
52
1

NSR
I




122
M
18
18
30.8
52
1

NSR
I




123
M
16
16
38
50
1

NSR
I




131
M
10
10
45.2
42
1

NSR
I
+



132
M
7
7
43.6
39
1

NSR
I




14
F
40
40
40
50
1

NSR
U




141
F
14
14
30.2
43
1

NSR
I




142
F
10
10
37.8
45
1

NSR
I




2
M
60
60
32.1
53
1

IVCD
U




3
M
63
56
5.6
54
4

AF
A
+


MIS
11
F
62
59
25.5
61
2
62
EF = 20%
A
p.Thr21135











AF, CHF



1
F
65
65




CHF, ICD
A
NA(+)



2
F
79
79





U




3
F
85
85





U




12
M
61
55
18
57
1


A
+



13
M
59
59
19
48.9


CAD/MI
I












PCIX3



132
M
35
35
28
51.6
1


I




14
M
37
27
13
76
4

Tx(37)
A
NA



141
M
30
30
32.6
52
1

EF = 60%
I




142
M
29
29
27
46.7
2

Epilepsy
I




19
M
50





DCM
A
NA











reported


MIV
14
M
50
26
9.5
63
3
49
EF = 20%
A
p.Asp14909











CHF, AF,











ICD(47)



11
M
56
54
31.1
54.3
1

NSR,
I












CAD



12
F
53
52
22.3
54.3
1

Long
A
+











QTc, VT











ablation











X2



121
M
22
20
13
76
4

VAD(20)
A
+



13
F
52
52
34.2
44.7
1

NRS
U




16
M
43
43
25.8
53.5
1

NSVT, RV
A
+











dilated











(55 mm)



2
F
81
81
12.4
49.2
1


A
+



21
F
51
51
35
53.2
1

AVR/MVR
I
+



23
F
48
48
33.3
55.5
1

NSR
I




24
M
46
46
34
54.5
1


I



TSFDC002
23
M
57
43
10
72
1
57
PVC,
A
p.Arg29415X











LAFB,











EF = 29%



116
M
51
42
8.6
70
1

TWA,
A
+











LAFB



190
F
72
66
22.4
58
3

TWA,
A
+











PVC,











RBBB,











LAFB



205
F
52
47
NA
NA
3

LAFB, ST
A
+



26
F
18
13
32
50
1

NSR, Abn
I
+











LV wall











motion,











MR


TSFDC017
253-2
M
72
65
22
59
1

EF = 40%
A
p.Glu17715fs



318
M
54
34
15
75
2
54
AF,
A
+











EF = 17%


TSFDC033
434
M
40
19
10
72
4

AF, TWA,
A
p.Asn30348fs











ICD











EF = 24%



0173
M
35
26
16.2
68
1
35
NSR
A
+


TSFDC050
115
M
53
35
13
70
3

NSR, ICD
A
p.Arg17470X











EF = 24%



1
M
50
48
NA
NA
3
50

A
NA(+)



113
F
21
18
31
48
1


I




114
F
61
40
10.3
68
2

1AVB,
A
+











TWA ICD





*Pedigrees are provided in FIG. 5. Probands and the TTN truncating mutation are listed first among family members.


Footnotes for symbols ({circumflex over ( )}||) are defined in Table 11.



Non-cardiac causes of death are indicated.



Clinical status is denoted: A, affected; U, unaffected; I, indeterminate.


□TTN genotypes are +, mutation present; −, mutation absent; NA, genotype not available; NA(+), obligate carrier based on pedigree position, but genotype not available.


Abbreviations used are defined in Table 11. Additional abbreviations used to describe electrophysiology are: NSR, normal sinus rhythm, LVH, electrocardiographic criteria for left ventricular hypertrophy; ST, sinus tachycardia; SB, sinus bradycardia; Q, Q waves; STTWA, ST and T-wave abnormality; TWA: T-wave abnormality; 1AVB, first degree atrioventricular block; LAFB, left anterior fasicular block; IVCD, intraventricular conduction delay; JR, junctional rhythm; RBB, right bundle branch block; LBBB, left bundle block; VT, ventricular tachycardia; PAC, premature atrial contractions; PVC, premature ventricular contractions; long QTc, prolonged corrected QT interval. Additional abbreviations to denote clinical findings are: Abn LV wall motion; abnormal left ventricular wall motion identified by echocardiography; CAD/MI, coronary artery disease/myocardial infarction; Diastolic Dysfunction, physician reported abnormal LV relaxation parameters; DM, diabetes mellitus; EF, ejection fraction; PCI, percutaneous coronary intervention SCD, sudden cardiac death; MRI:GdE, LV fibrosis identified by gadolinium enhance-MRI; RV, right ventricle; VT ablation, ventricular tachycardia treated by ablation.













TABLE 15







Likelihood of the odds (LOD) scores reflecting linkage between TTN


mutation and DCM in individual families*













TTN
LOD score
LOD score



Family
variant
(pen = 0.95)
(pen = 0.80)
















MEK
Frameshift
0.29
0.29



MEW
Nonsense
0.17
0.17



MHQ
Nonsense
0.16
0.12



MIP
Nonsense
1.16
1.04



MID
Nonsense
0.59
0.59



MHG
Frameshift
3.57
3.21



MEQ
Duplication
0.00
0.00



DNFDC081
Nonsense
0.17
0.17



DNFDC088
Nonsense
0.84
0.84



DNFDC103
Nonsense
0.17
0.17



DNFDC142
Nonsense
0.00
0.00



TSFDC002
Nonsense
0.59
0.59



TSFDC017
Frameshift
0.00
0.00



TSFDC033
Frameshift
0.17
0.17



TSFDC050
Nonsense
0.30
0.30



TSSDC019
Nonsense
0.00
0.00



MHX
Nonsense
1.14
0.96



MDD
Splice
0.17
0.17



MIV
Splice
1.47
1.40



MIS
Splice
0.14
0.09




total
11.1
10.3







*LOD score for each family calculated at θ = 0 and indicated penetrance. Pedigrees and clinical data are provided in FIG. 5 and Table 15. See Methods for details on the assignment of affection status.













TABLE 16







Previously published TTN truncation mutations*













AA


Mutation





position
NT name
AA name
Type
Genotype{circumflex over ( )}
Diagnosis
Source
















2484
c.7450G > A
p.Gln2484X
nonsense
Hetero
Cancer
Greenman








200720


4053
c.12156C > T
p.Gln4053X
nonsense
Hetero
Heart failure
Itoh-Satoh 2002


15465
c.46395C > T
p.Trp15465X
nonsense
Somatic
Cancer
Greenman








200720


15700
c.47098G > A
p.Arg15700X
nonsense
Somatic
Cancer
Greenman








200720


21924
c.65766_65767insAT
p.Thr21924fs
frameshift
Hetero
DCM
Gerull 20021


28388
c.85161delG
p.Glu28388fs
frameshift
Hetero
DCM
Gerull 200621


33534
c.100600_100600
p.His33534fs
frameshift
Homo
Congenital Myopathy
Carmignac



delACCAAGTG




200722


33915
c.101744delA
p.Lys33915fs
frameshift
Homo
Congenital Myopathy
Carmignac








200722


34242
c.102723delT
p.Ser34242fs
frameshift
Hetero
Severe tibial
Hackman 200823







muscular dystrophy


34322
c.102965delA
p.Lys34322fs
frameshift
Hetero
Severe tibial
Hackman 200823







muscular dystrophy


34323
c.102967G > A
p.Gln4322X
nonsense
Hetero
Severe tibial
Hackman 200823







muscular dystrophy





*Positions are for UniProt titin (Q8WZ42);


{circumflex over ( )}Subjects are reported to have germline (hetero, heterozygous or homo, homozygous) or somatic TTN mutations.













TABLE 17







TTN exons comprising the standard UniProt titin protein (Q8WZ42)


that have minimal or no evidence for cardiac expression














Q8WZ42
Q8WZ42


Chrom
hg19 start
hg19 end
start
end





2
179,535,817
179,537,208
11245
11396


2
179,540,648
179,542,644
11029
11139


2
179,544,066
179,545,898
10766
10931


2
179,549,057
179,549,716
10507
10591









All patents and other publications identified in the specification and examples are expressly incorporated herein by reference for all purposes. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents.


Although preferred embodiments have been depicted and described in detail herein, it will be apparent to those skilled in the relevant art that various modifications, additions, substitutions, and the like can be made without departing from the spirit of the invention and these are therefore considered to be within the scope of the invention as defined in the claims which follow. Further, to the extent not already indicated, it will be understood by those of ordinary skill in the art that any one of the various embodiments herein described and illustrated can be further modified to incorporate features shown in any of the other embodiments disclosed herein.

Claims
  • 1.-44. (canceled)
  • 45. An assay comprising: (i) contacting a nucleic acid sample obtained from a subject with a probe, wherein the probe is capable of detecting a mutation resulting in a truncated TITIN polypeptide;(ii) detecting the presence of the mutation in the nucleic acid sample, and(iii) determining the subject has an increased risk for developing dilated cardiomyopathy (DCM) or in need of treatment for DCM if at least one mutation is detected in (ii).
  • 46. The assay of claim 45, wherein the probe is a sequencing primer.
  • 47. The assay of claim 11, wherein detecting the presence of the mutation comprises: (i) sequencing at least a portion of TTN gene in a sample from a human subject;(ii) comparing the sequence obtained in step (i) with wild type TTN gene sequence having a nucleic acid sequence set forth in SEQ ID NO: 1.
  • 48. The assay of claim 45, wherein the truncated TITIN polypeptide lacks part of A-band having a wild-type sequence set forth in SEQ ID NO: 725.
  • 49. The assay of claim 45, wherein the mutation is selected from the group consisting of 6247—6247delG, 12745C>T, 14470—14471insCACACTCCATA (SEQ ID NO: 722), 19183—19183delG, 23798—23810de1GTCAAGATATCTG (SEQ ID NO: 723), 38621—38622insA, 44336—44336delA, 45322—45322delT, 49077G>A, 51883C>T, 52408C>T, 53145—53146insG, 53347G>T, 53935—53935delC, 56367T>A, 56572C>T, 56953C>T, 58678C>T, 59530C>T, 61046—61046delC, 65867—65867delA, 67057—67063de1GCATATGinsTA, 67745—67745delT, 72178—72179insT, 72723—72739delinsAGA, 77065C>T, 79896G>A, 80845C>T, 81046A>T, 81440G>A, 81536—81537delCT, 82701C>A, 84977—84980delATTA, 87953G>A, 88242C>T, 88528G>T, 89177—89181delAAATT, 90241C>T, 91042—91042delA, 91537—91538insA, 94111A>T, 95522C>A, 30476-1G>A, 34186+1G>T, 35635G>C, 35635+1G>A, 44725+2delT, 48364+1G>T, 50346—+3A>G, 54422-5T>A, 54704-1G>A, 55003+1G>A, 62425+5G>A, 63405A>G, 64489+1G>A, 81898+2T>A, 92569+1G>C, and any combination thereof, wherein the mutation location is determined based upon the wildtype TTN sequence having a nucleic acid sequence set forth in SEQ ID NO: 1.
  • 50. The assay of claim 45, wherein dilated cardiomyopathy is idiopathic dilated cardiomyopathy or heart failure or any other related cardiomyopathy.
  • 51. A method of treating a subject for dilated cardiomyopathy (DCM), comprising: a. Selected a subject in need of treatment for DCM using an assay comprising: i. contacting a nucleic acid sample obtained from a subject with a probe, wherein the probe is capable of detecting a mutation resulting in a truncated TITIN polypeptide;ii. detecting the presence of the mutation in the nucleic acid sample, andb. administering a treatment for DCM if at least one mutation is detected in (ii).
  • 52. The method of claim 51, wherein the treatment is selected from the group consisting of angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers, beta blockers, diuretics, aldosterone antagonists, digoxin (Lanoxin), blood thinning medications, biventricular pacemakers, implantable cardioverter-defibrillators (ICDs), heart pumps (left ventricular assist devices, or LVADs), heart transplant, gene therapy, calcium channel blockers, tissue growth factor inhibitors, and any combinations thereof.
  • 53. The method of claim 51, wherein the treatment comprises administering to said subject a polypeptide comprising amino acid SEQ ID NO: 2 or a nucleic acid encoding a polypeptide comprising amino acid SEQ ID NO: 2.
  • 54. The method of claim 53, wherein the nucleic acid encoding the polypeptide of amino acid SEQ ID NO: 2 is operably linked to a vector.
  • 55. The method of claim 51, wherein the truncated TITIN polypeptide lacks part of the A-band, having a wild-type sequence set forth in SEQ ID NO: 725.
  • 56. The method of claim 51, wherein the mutation is selected from the group consisting of 6247—6247delG, 12745C>T, 14470—14471insCACACTCCATA (SEQ ID NO: 722), 19183—19183delG, 23798—23810de1GTCAAGATATCTG (SEQ ID NO: 723), 38621—38622insA, 44336—44336delA, 45322—45322delT, 49077G>A, 51883C>T, 52408C>T, 53145—53146insG, 53347G>T, 53935—53935delC, 56367T>A, 56572C>T, 56953C>T, 58678C>T, 59530C>T, 61046—61046delC, 65867—65867delA, 67057—67063de1GCATATGinsTA, 67745—67745delT, 72178—72179insT, 72723—72739delinsAGA, 77065C>T, 79896G>A, 80845C>T, 81046A>T, 81440G>A, 81536—81537delCT, 82701C>A, 84977—84980delATTA, 87953G>A, 88242C>T, 88528G>T, 89177—89181delAAATT, 90241C>T, 91042—91042delA, 91537—91538insA, 94111A>T, 95522C>A, 30476-1G>A, 34186+1G>T, 35635G>C, 35635+1G>A, 44725+2delT, 48364+1G>T, 50346—+3A>G, 54422-5T>A, 54704-1G>A, 55003+1G>A, 62425+5G>A, 63405A>G, 64489+1G>A, 81898+2T>A, 92569+1G>C, and any combination thereof, wherein the mutation location is determined based upon the wildtype TTN sequence having a nucleic acid sequence set forth in SEQ ID NO: 1.
  • 57. The method of claim 51, wherein detection of the mutation is by nucleic acid sequencing.
  • 58. The method of claim 51, wherein dilated cardiomyopathy is idiopathic dilated cardiomyopathy.
  • 59. A method of treating a subject for dilated cardiomyopathy (DCM), comprising administering a treatment for DCM to a subject determined to have a mutation resulting in a truncated TITIN polypeptide and not administering a treatment for DCM to a subject determined not to have a mutation resulting in a truncated TITIN polypeptide.
RELATED APPLICATIONS

This application claims benefit under 35 U.S.C. §119(e) of the U.S. Provisional Application No. 61/474,106, filed Apr. 11, 2011, the content of which is incorporated herein by reference in its entirety.

GOVERNMENT SUPPORT

This invention was made with government support under grant no. N01-HV-48194 awarded by the National Heart, Lung, and Blood Institute. The government has certain rights in the invention.

PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US12/33122 4/11/2012 WO 00 2/24/2014
Provisional Applications (1)
Number Date Country
61474106 Apr 2011 US