1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63

REVIEW ARTICLE The role of genetic testing in unexplained sudden death Q20

CHRIS J. MILES, and ELIJAH R. BEHR LONDON, UK

Most sudden deaths are because of a cardiac etiology and are termed sudden cardiac death (SCD). In younger individuals coronary artery disease is less prevalent and cardiac genetic disorders are more common.1 If sudden death is unexplained despite an appropriate autopsy and toxicologic assessment the term sudden arrhythmic death syndrome (SADS) may be used.2 This is an umbrella term and common underlying etiologies are primary arrhythmia syndromes with a familial basis such as Brugada syndrome, long QT syndrome, and subtle forms of cardiomyopathy. The first clinical presentation of these conditions is often SCD, which makes identification, screening, and risk stratification crucial to avert further deaths. This review will focus on genetic testing in the context of family screening. It will address the role of the ‘‘molecular autopsy’’ alongside current postmortem practices in the evaluation of SADS deaths. We describe the current data underlying genetic testing in these conditions, explore the potential for next-generation sequencing, and discuss the inherent diagnostic problems in determination of pathogenicity. (Translational Research 2015;-:1–15)

Q1

Q2

Abbreviations: AHA ¼ American Heart Association; APHRS ¼ Asia Pacific Heart Rhythm Society; ARVC ¼ arrhythmogenic right ventricular cardiomyopathy; BrS ¼ Brugada syndrome; ChIP ¼ channel interacting protein; CPVT ¼ catecholaminergic polymorphic ventricular tachycardia; DCM ¼ dilated cardiomyopathy; EHRA ¼ European Heart Rhythm Association; ERS ¼ early repolarization syndrome; HCM ¼ hypertrophic cardiomyopathy; HRS ¼ Heart Rhythm Society; IVF ¼ idiopathic ventricular fibrillation; LQTS ¼ long QT syndrome; ONS ¼ Office for National Statistics; PCCD ¼ premature cardiac conduction disease; SADS ¼ sudden arrhythmic death syndrome; SCD ¼ sudden cardiac death; SNR ¼ signal-to-noise ratio; SQTS ¼ short QT syndrome; SUDS ¼ sudden unexpected death syndrome; VUS ¼ variant of unknown significance

Q3

INTRODUCTION

From the Division of Clinical Sciences, Cardiovascular Sciences Research Centre, St George’s University of London, London, UK. Submitted for publication January 1, 2015; revision submitted June 8, 2015; accepted for publication June 11, 2015. Reprint requests: Elijah R. Behr, Division of Clinical Sciences, Cardiovascular Sciences Research Centre, St George’s University of London, Cranmer Terrace, London SW17 0RE, UK; e-mail: ebehr@sgul. ac.uk. 1931-5244/$ - see front matter Ó 2015 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.trsl.2015.06.007

T

he estimated incidence of sudden cardiac death (SCD) in the general population in Europe and the United States (US) is between 50 and 100 per 100,000 per annum.3-5 SCD claims 300,000– 400,000 deaths per annum in the US6,7 and there were an estimated 70,000 SCDs in the UK in 2010, most because of ischemic events.8 SCD is usually defined as an unheralded witnessed instantaneous death but it may also be described as being preceded by a prodrome of acute cardiac symptoms up to 1 hour before death.7 Unwitnessed cases without a prior deterioration in the preceding 12–24 hours may also be included. Estimates vary, however, because of a dependence on the presence 1

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127

2

128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191

Translational Research - 2015

Miles and Behr

of autopsied vs nonautopsied cases and the variability of definitions and the duration of prodromal symptoms before the terminal event. A national prospective survey of English coroners’ cases for more than a 20-month period in the late 1990s evaluated sudden deaths in Caucasians aged 4–64 years with no history of cardiac disease, negative toxicology, and last seen alive within 12 hours of death.2 A normal coroner’s autopsy and subsequent evaluation of the heart by a cardiac pathologist established these as sudden arrhythmic death syndrome (SADS) cases with an estimated incidence of up to 1.38 per 100,000 per annum. SCD in the young. In individuals aged ,35 years inherited cardiac diseases are more prevalent and estimates of the incidence of SCD vary depending on definitions and study site. For example, a Danish nationwide analysis of deaths from 2000 to 2006 revealed an annual incidence of young SCD of 2.8 per 100,000,9 whereas a retrospective US study of .6 million military recruits reported an incidence as high as 13 per 100,000 per annum.10 Puranik et al11 retrospectively reviewed pathologic reports from 427 autopsied sudden death cases aged 5–35 years at a forensic medical facility from 1995 to 2004 in Sydney. The most common cardiac cause of death was presumed arrhythmia in those with no (or minimal) structural heart disease (29%), that is, SADS. Retrospective analysis of death certification, autopsy reports, and registry data estimated that 31% of autopsied SCD cases in Danes aged 1–49 years were unexplained and attributed to SADS.12 Conversely, in 79% of 197 cases of young SCD in Italy, histologic analysis yielded a structural diagnosis such as cardiomyopathy or focal myocarditis; 6% were unexplained and attributable to SADS.13 In the Veneto region of Italy, studies have implicated arrhythmogenic right ventricular cardiomyopathy (ARVC) in 20% of sudden deaths among athletes and the young.14 Maron et al15 described sudden deaths in young competitive athletes for more than a 27-year period in the US: 56% were because of cardiovascular disease, the most common cause (36%) being hypertrophic cardiomyopathy (HCM). In the UK the incidence of cardiac death in the young (#35 years) as determined by analysis of Office for National Statistics data was 1.8 per 100,000 in England and Wales (2002–2005). Critical appraisal estimated the incidence of SADS as 0.24 per 100,000 per annum, significantly higher than the 0.1 per 100,000 reported by the Office for National Statistics as instantaneous unexplained sudden death, but still less than most other estimates.16 This is likely to be because of misclassification of the cause of death as mortality

data are largely derived from death certificate documentation that may under-report the true incidence of cardiac arrhythmia. For example, in one study a significant proportion (23%) of unexplained drowning cases carried mutations associated with arrhythmia syndromes,17 and certification of sudden death in epilepsy may overlook cases, which result from a primary arrhythmic cause.18,19 DEFINING SADS

A consensus statement from the Heart Rhythm Society (HRS), European Heart Rhythm Association (EHRA), and the Asia Pacific HRS defines SADS as a pathologic diagnosis of exclusion after postmortem cardiac investigation and toxicologic analysis.20 Even if nondiagnostic pathology is detected, cases should still be considered as SADS because of the high chance of underlying inherited ion channel disease.21 Expert autopsy is also recommended as general pathologists may misdiagnose cases, over diagnosing ARVC, and under diagnosing SADS.22 Guidelines for autopsy practice exist and include detailed description of postmortem sampling techniques with integration of specialist skills in the evaluation of possible familial disorders.23,24 In the US, the state-wide Sudden Death in the Young Registry collates young SCD case data, using DNA analysis in a subset of cases for the purpose of further evaluation and future research.25 Unfortunately, access to expert cardiac pathology is very much limited internationally. DIAGNOSTIC APPROACHES AND GENETIC TESTING

Two approaches may be taken to make a diagnosis in a family: familial clinical evaluation with genetic testing targeted to phenotype and postmortem genetic testing ‘‘the molecular autopsy.’’ The overall aim is to identify cardiac genetic disease were present and institute preventative treatment were necessary to avert further SCD.26 The clinical role of genetic testing is therefore a diagnostic one and dependent on identifying mutations, that is, disease-causing or pathogenic rare genetic variants. The rapid development in sequencing technology has, however, led to the identification of frequent rare genetic variation in both healthy and affected individuals. Rare variants are often private to a specific family and therefore may be unknown in the literature. Their associated risk for disease causation and therefore their clinical significance is often uncertain and a major challenge as incorrect inferences of causality can have serious implications for diagnosis and management of families. If pathogenicity of a rare variant remains uncertain then it is termed a ‘‘variant of unknown significance’’ (VUS).27

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255

Translational Research Volume -, Number -

3

web 4C=FPO

256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319

Miles and Behr

Fig 1. Guidelines for genetic testing in the channelopathies and diagnostic utility based on the signal-to-noise ratio. Estimates for molecular autopsy are based on the studies analyzed in this review. Adapted from the HRS/EHRA Genetic Testing Guidelines.47 *Yield of diagnostic test—proportion of patients with positive genotyping, derived from unrelated cases. The first number is the yield when a major gene has been targeted. The number in parenthesis is the total when including all disease-associated genes. **Signal-to-noise ratio—estimate of positive predictive value, devised by dividing the case yield of rare variants by the background control rate of rare missense variants among major genes. ^Rare missense variants—possible disease-causing mutations determined by a percentage of rare amino acid substitutions in the major disease-associated genes. BrS, Brugada syndrome; CPVT, catecholaminergic polymorphic ventricular tachycardia; ECG, electrocardiogram; EHRA, European Heart Rhythm Association; HRS, Heart Rhythm Society; LQTS, long QT syndrome; SUDS, sudden unexpected death syndrome.

The presence in healthy normal people of rare variants, usually missense, is described as genetic ‘‘background noise’’ (ie, the frequency of rare variants within a particular gene in the healthy population).28 The ratio of rare variant yield in cases (also mainly missense) to background noise is known as the ‘‘signal-to-noise ratio’’ (SNR) (Fig 1). Various methods for assessing likelihood of pathogenicity of rare variants have evolved as follows.29,30 Absence population.

of

the

variant

in

a

healthy

control

Before the extent of rare genetic variation in the genome was appreciated, most research studies and laboratory reports presumed pathogenicity on the basis of absence in a control population. Although the presence of a rare variant in the general population is helpful in reducing the chance of it being directly causative, novelty no longer carries much weight. Nonetheless, it is the first step in assessing likelihood of pathogenicity and increasingly large reference databases are available such as the Exome Aggregation Consortium with exome data from more than 60,000 unrelated individuals.31

Cosegregation of phenotype with genotype in large families. This is the process of associating a variant

with disease in a family by identification of the variant in all affected individuals. This assists in determining the likelihood of causality, although if one affected relative does not carry the variant then it cannot be used as a clinical test. In practice, however, this approach is often not feasible because of small family sizes and the possibility of association by chance. The detection of a de novo rare variant in an associated gene in an affected child with unaffected parents does, however, support strongly the likely pathogenicity of the variant. Severity of the type of mutation, that is, nonsense vs missense. In general, the likelihood of a functional ef-

fect of a variant on a protein and/or its reduced expression is increased the more ‘‘radical’’ the associated Q4 DNA change is. Nonsynonymous single nucleotide changes, that is missense mutations, are common and are found in both healthy unaffected and affected individuals and are most difficult to assess (see aforementioned). Mutations because of deletions or insertions and some splice-site mutations may lead to abnormal

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383

4

384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447

Translational Research - 2015

Miles and Behr

protein products and protein truncations. These ‘‘nonsense’’ mutations are less common and less likely to be present in unaffected individuals. Causation is therefore less difficult to determine. For example, premature truncation SCN5A mutations have been linked to severity of disease and are highly likely to be pathogenic because of haploinsufficiency.32 However, although truncating variants of the gene titin (TTN) are associated with dilated cardiomyopathy (DCM),33 they are also present in unaffected controls and represent a diagnostic challenge.34 Prior and reliable description in the literature. It can be helpful if a variant has previously been associated with disease in another proband and their family. Unfortunately not all prior reports are robust. Previously published rare variants postulated to be mutations have since been determined to be present in population databases at too high a frequency to be causative of disease.35,36 They may represent functionally active variants that modify disease expression or otherwise are nonfunctional population variants. For example, one study examined an established exome database and compared it with a catalog of 197 variants reported as causative of DCM; 16.8% of the variants reported as causative were also present in the exome database, although functional data suggested that a significant proportion may have been lower penetrance disease-causing alleles.37 Another study of childhood recessive diseases suggested 27% of mutations previously cited as pathogenic are in fact common polymorphisms or misinterpreted variants.38 Amino methods.

acid

conservation

and

in

silico

Missense mutations result in an amino acid changes that may have varying differences of side chain size, polarity, and lipophilicity compared with the wildtype protein. The greater the difference is the more likely that a structural and/or functional change will occur. In addition, the amino acid may be at a critical part of the protein that is conserved in other human proteins with similar functional domains (paralogs) or the same protein in other species (orthologs). Measures of conservation of amino acids and therefore likelihood of disease causation because of an amino acid change have been assessed by algorithms such as the Grantham conservation score.39 These methods have also been incorporated into a number of different in silico tools that can also use predicted protein structural changes to infer causation. In combination, they can be effective in assisting assessment of pathogenicity of rare variants in KCNQ1 and KCNH2 in long QT syndrome (LQTS)40 and to lesser extent SCN5A variants in LQTS and Brugada syndrome (BrS).41 They are, however, unreliable as sole evidence of pathogenicity in the clinical setting.

Functional expression studies of the variant’s biophysical effect. The consequences of mutations can be studied in

RNA expression systems and functional models such as ion channel expression studies, animal models, and induced pluripotent stem cell–derived cardiomyocytes. Unfortunately, these are generally costly and are not feasible for routine evaluation of genetic findings. To optimize and standardize assessment, Campuzano et al have recently proposed a scoring system to determine the pathogenicity of genetic variants associated with the arrhythmia syndromes combining data from the previously mentioned methods. A scoring range from 0 to 15 categorized variants into separate groups, including benign, VUS, and pathogenic.42 This sort of approach may enable better classification of variants in the future. Determining the causation of disease in families can be further complicated by other genetic variants that can influence phenotype may also be responsible, known as digenic or oligogenic inheritance. For example, 5%–10% of LQTS patients have mutations in 2 or more of the known genes, which can influence disease expression.43 CAUSES OF SADS AND THE ROLE OF GENETIC TESTING

Arrhythmia syndromes (or cardiac channelopathies) are a heterogeneous group of conditions that are usually hereditary and are associated with an apparently structurally normal heart. They are often caused by mutations in genes encoding cardiac ion channel subunits and channel interacting proteins (Table I). The channelopathies include LQTS, BrS, catecholaminergic polymorphic ventricular tachycardia (CPVT), short QT syndrome, idiopathic ventricular fibrillation, early repolarization syndrome, premature cardiac conduction disease, and others. These are described in Table I. LQTS has the highest yield from testing (approximately 75%), with wide variation among the other channelopathies (20% in the BrS).27 In some cases, despite a normal heart being found at autopsy, cardiomyopathy is detected during clinical evaluation of the family. Thus, disease insufficient to cause histopathologic changes detectable even at expert autopsy may still cause or predispose toward SCD.44 We focus on the main phenotypes of interest subsequently. Long QT syndrome. Congenital LQTS has a prevalence of approximately 1:200045 and is characterized by prolongation of the QT interval on the electrocardiogram (ECG) and increased risk of the characteristic ventricular arrhythmia, Torsades de Pointes, and SCD. Hundreds of mutations in 14 Q5 genes27 predominantly encoding components and channel interacting proteins of repolarizing potassium currents and the sodium channel current (Nav1.5) have been identified (Table I).

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511

Translational Research Volume -, Number 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575

Miles and Behr

5

Table I. Genes associated with ion channel diseases and their associated phenotypes and frequencies

Brugada syndrome

Long QT syndrome

CPVT

Other syndromes

Gene

Ion channel component

Effect of mutation

Ion channel disease

SCN5A GPD1L CACNA1C CACNB2 SCN1B KCNE3 SCN3B HCN4 KCNJ8 CACNA2D1 KCND3 MOG1 SLMAP KCNQ1 KCNH2 SCN5A ANK2 KCNE1 KCNE2 KCNJ2 CACNA2D1 CAV3 SCN4B AKAP9 SNTA1 KCNJ5 CALM1/CALM2 RYR2 CASQ2 TRDN CALM1 KCNQ1 KCNH2 KCNE2 KCNJ2 KCNJ8 DPP6 HCN4 CACNA1C CACNB2 CACNA2D1 SCN5A

a Subunit INa INa ChIP a Subunit ICa b Subunit ICa b Subunit INa b Subunit IKs/Ito b subunit INa If a Subunit IKATP a2d subunit ICa a subunit Ito INa ChIP INa ChIP a Subunit IKs a Subunit IKr a Subunit INa INa,K, INCX ChIP b Subunit IKs b Subunit IKr a Subunit IK1 a2d Subunit ICa INa ChIP b Subunit INa IKs ChIP INa ChIP a Subunit IKAch SR Ca regulation a Subunit SR Ca regulation SR Ca regulation SR Ca regulation a Subunit IKs a Subunit IKr b Subunit IKr a Subunit IK1 a Subunit IKATP Ito If a Subunit ICa b Subunit ICa a2d Subunit ICa a Subunit INa

Loss of function Loss of function Loss of function Loss of function Loss of function Gain of function Loss of function Loss of function Gain of function Loss of function Gain of function Loss of function Loss of function Loss of function Loss of function Gain of function Loss of function Loss of function Loss of function Loss of function Gain of function Gain of function Gain of function Loss of function Gain of function Loss of function Loss of function Loss of function Loss of function Loss of function Loss of function Gain of function Gain of function Gain of function Gain of function Gain of function Gain of function Loss of function Loss of function Loss of function Loss of function Loss of function

BrS1 BrS2 BrS3 BrS4 BrS5 BrS6 BrS7 BrS8 BrS9 BrS10 BrS11 BrS12 BrS13 LQT1 LQT2 LQT3 LQT4 LQT5 LQT6 LQT7 (CPVT-like) LQT8 LQT9 LQT10 LQT11 LQT12 LQT13 LQT14 CPVT1 CPVT2 CPVT4 CPVT5 SQT2, FAF SQT1 FAF SQT3, FAF ERS IVF SSS ERS ERS ERS PCCD ERS SSS Overlap MEPPC

Mixed effect Gain of function

Q17

Frequency in phenotype (1reference)

20%–30%41 ,1%64 BrS 2%–12%63,64,110 BrS 2%–12%63,64,110 ,1%64 ,1%64 ,1%64 NA133 ,1%64 BrS 2%–12%63,64,110 ,1%64 ,1%64 NA123 40%–55%107 35%–45%107 2%–8%107 ,1%107 ,1%107 ,1%107 ,1%107 ,1%107 ,1%107 ,0.1%107 ,0.1%107 ,0.1%107 NA117 NA129 50%124 NA126 NA127 NA128 NA112,113 NA114 NA115 NA114,116 NA118,119 NA121 NA132 NA NA NA NA103 66%105 2%–3%104 NA61,108 NA109

Abbreviations: BrSX, Brugada syndrome subtype X; ChIP, channel interacting protein; CPVT, catecholaminergic polymorphic ventricular tachycardia; ERS, early repolarization syndrome; ICa, depolarizing inward calcium current (slow); If, funny current; IK1, Kir2.1 inward rectifying current; IKATP, ATP-sensitive potassium channel; IKr, repolarizing outward rapid rectifying potassium current; IKs, repolarizing outward slow rectifying potassium current; INa, depolarizing inward sodium current (fast) Nav1.5; Ito, transient outward potassium current; IVF, idiopathic ventricular fibrillation; LQTX, long QT syndrome subtype X; MEPPC, multifocal ectopic Purkinje-related premature contractions; NA, not ascertained or applicable; PCCD, premature cardiac conduction defect; SQTX, short QT syndrome subtype X; SR Ca, sarcoplasmic reticulum calcium; SSS, Q18 Q19 sick sinus syndrome. Adapted from Wilde and Behr.27

Approximately 75% of patients with the syndrome have mutations in 1 of 3 ‘‘major’’ LQTS genes, the potassium channel genes KCNQ1 (LQT1, 35%) and KCNH2 (LQT2, 30%) and the sodium channel gene

SCN5A (LQT3, 10%).46 With a background genetic ‘‘noise’’ level of 2%–4% for major genes, this gives a relatively favorable SNR. The 2011 HRS/EHRA Expert Consensus Statement therefore recommends

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639

6

640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703

Q6

Q7

Translational Research - 2015

Miles and Behr

comprehensive or LQT1–3 targeted LQTS genetic testing for any patient meeting diagnostic criteria (Fig 1).29 The 2013 diagnostic guidelines include carriers of a pathogenic mutation as being affected, regardless of clinical features.47 Genotype can be associated with phenotypic differences in clinical presentation and may be important in the SADS victim’s presentation: LQT1 with exercise (especially swimming) and high emotional states; LQT2 with sudden arousal and acoustic triggers; and LQT3 with rest or sleep.46 The less common or ‘‘minor’’ LQTS genes have been associated with phenotype in small studies with low yields and small control groups. In some cases linkage is not available and causality rests on functional data and absence in a control. For example, the original description of CAV3 as the cause of LQT9 has been questioned in subsequent follow-up studies.48 Background rare variation increases when the minor genes are included in testing panels but with little increase in yield. This makes the SNR less favorable and result less readily interpretable. Brugada syndrome. BrS is diagnosed by the presence of at least 2 mm of coved ST increase with T wave inversion in at least one of the standard and high intercostal space right ventricular ECG leads: the type 1 ECG pattern.20,49 This pattern can be concealed and often dynamic, unmasked by factors such as sleep50 and pyrexia,51 as well as provoked by sodium channel blocking drugs such as ajmaline, procainamide, and flecainide.52 It can manifest with syncope because of polymorphic ventricular tachycardia, ultimately predisposing to ventricular fibrillation and SCD, which is commonly the first presentation.53 The prevalence of a BrS type 1 ECG is higher in Asian countries, approximately 0.15% in adults. This is compared with ,0.02% in a Western population.54 BrS is a heterogeneous condition with reduced genetic penetrance. Recent evidence has suggested variable clinical phenotypes, where the syndrome may overlap with structural conditions such as ARVC.55 Inheritance can be autosomal dominant with variable expressivity or sporadic,56 as such, most individuals can remain asymptomatic throughout their lifetime. Recent evidence has also supported an oligogenic inheritance pattern.57 The most commonly affected gene is the SCN5A gene (approximately 20% of BrS cases, BrS158). More than 300 mutations of SCN5A have been described59 leading to loss of function because of a reduction in the amplitude of the sodium channel current by reduced expression and/or altered voltage-gating properties.60 Conversely, although gain-of-function SCN5A mutations are found in LQT3, an overlap in phenotype exists between BrS

and LQTS, both having been described in large families.61 In addition, there are large SCN5A positive families that have affected individuals who are noncarriers, suggesting a modifier role rather than direct causation in these pedigrees.62 Many other genes have been associated with the syndrome (Table I). One study identified mutations of the L-type calcium channel (encoded by CACNA1C, CACNB2B, and CACNA2D1) in 10%–15% of BrS cases.63 The clinical phenotype also demonstrated a short QT interval. This has not been replicated by a follow-up study and the rest of these candidate genes account for very small numbers of cases.64 Other than GPD1L and SCNB1, these have not demonstrated genetic linkage in pedigrees.24 A recent study has also implicated SCN10A, which encodes the neuronal current Nav1.8, as being responsible for a significant proportion of BrS cases.65 This has not been confirmed in a similar study of SCN5A negative BrS patients. Great caution must therefore be used in assessing rare variants in these low frequency genes. Thus, the overall SNR level and potential for oligogenic inheritance are relatively unfavorable for diagnostic testing and recommendations are not as strong as in LQTS. Genetic testing can be useful in known or suspected cases of BrS, but a positive test adds little to the on-going clinical management. It is, however, recommended for assessing relatives (Fig 1).47 Catecholaminergic polymorphic ventricular tachycardia. CPVT is a rare inherited arrhythmic disor-

der first described in 1975,66 typically presenting within the first decade of life. Prevalence is estimated in 1:10,000 and individuals are at risk of SCD by means of catecholamine-induced polymorphic and bidirectional VT.67 The mechanism of arrhythmia in CPVT is chiefly because of intracellular calcium mishandling, where mutations in the RYR2 gene cause uncontrolled calcium release from the sarcoplasmic reticulum during diastole.68 The initial clinical manifestation is typically syncope associated with exercise or acute stress; however, SCD can be its sole presentation. The heart structure and resting ECG is often unremarkable in CPVT, necessitating use of exercise or catecholamine stress testing to capture arrhythmias, most commonly bidirectional VT.47 CPVT principally arises from mutations in 2 genes: CPVT1-RYR2 (encodes the cardiac ryanodine receptor channel) and CPVT2-CASQ2 (encodes calsequestrin, responsible for buffering calcium in the sarcoplasmic reticulum).69 CPVT has been described in small numbers of KCNJ2, ANKB, and CALM1/2 carriers, genes that have also been associated with LQTS.70 The condition is inherited with high penetrance in either

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767

Translational Research Volume -, Number 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831

Miles and Behr

an autosomal dominant (RYR2) or recessive (CASQ2) fashion, the former being more common affecting 55%–65% of probands, with CASQ2 only affecting a small number of cases but increasing the likelihood of earlier symptom onset.69 More than 100 mutations of RYR2 have been linked to CPVT1, the autosomal dominant form, which affect certain regions of the protein (mainly the FKBP12.6 binding domain, covering most RYR2).71 Because of the large size of the gene, targeted screening has been used in the past and can potentially miss causative mutations.72 Recent studies suggest that mutation location may be associated with severity of the phenotype, suggesting a future role for genetic risk stratification alongside conventional techniques.73 Because of the favorable SNR level targeted CPVT genetic testing is recommended for any patient with clinical suspicion of CPVT based on the clinical findings and electrocardiographic phenotype during stress testing. Mutation-specific genetic testing is also recommended for family members and appropriate relatives after the identification of the CPVT-causative mutation in an index case.47 Like with LQTS, the 2013 diagnostic guidelines include carriers of a pathogenic mutation as being affected, regardless of clinical features.20 Cardiomyopathies. The cardiomyopathies are diseases of the heart muscle associated with mechanical failure of myocardial performance. The American Heart Association describes a heterogeneous group of diseases usually exhibiting ventricular hypertrophy or dilatation, caused by a variety of etiologies that frequently are genetic.74 They can broadly be divided into HCM, DCM, and ARVC. HCM, the most common cardiomyopathy, has a prevalence of approximately 1:500 of the general population.74 It is characterized by unexplained cardiac hypertrophy, often in an asymmetric septal pattern, but not always. Mutations involving sarcomere proteins and other proteins involved in cardiac contraction are responsible. Familial HCM is mostly (.90%) inherited in an autosomal dominant fashion and most mutations involve cardiac b-myosin heavy chain (MYH7) and cardiac myosin binding protein C (MYBPC3). Genes encoding mitochondrial enzymes have also been identified.75 HRS/EHRA guidelines recommend genetic testing after diagnosis, with mutation-specific testing indicated for family members after identification.47 In idiopathic DCM, the heart is dilated and impaired leading to symptoms and signs of congestive cardiac failure but without an apparent underlying cause. The estimated prevalence is approximately 1:2500, affecting both adults and children; inherited forms make up to 50%, where there is a family history in one or more

7

relatives.76 Mutations in .30 genes have been associated with DCM, most following an autosomal dominant inheritance pattern.77 The titin gene (TTN) probably accounts for approximately 20% of cases in recent studies but the high frequency of rare variants has limited the clinical utility of assessing risk; other genes have a low yield.78 Extensive or targeted (LMNA and SCN5A) testing is recommended (class I indication) for those patients with significant conduction disease with or without overt DCM as mutations in these genes can indicate an increased risk of SCD.47 ARVC is characterized by fibro-fatty replacement of the right and/or left ventricular myocardium with associated risk of SCD because of ventricular arrhythmias. Most cases are inherited in an autosomal dominant pattern with incomplete penetrance and variable expressivity. Its overall prevalence is approximately 1:10000, often presenting between the second and fourth decade of life.79 Seven genes have been associated with ARVC: JUP, DSP, PKP2, DSG2, DSC2, TGFb3, and TMEM43.80 HRS/EHRA guidelines suggest that comprehensive or targeted genetic testing can be useful in those that meet existing 2010 task force diagnostic criteria (which include evaluation of right ventricular structure and repolarization abnormalities), but is not helpful in those with an uncertain diagnosis.47 FAMILY EVALUATION AND PHENOTYPE TARGETED GENETIC TESTING

The first study of families of SADS victims reported a limited investigative protocol followed by targeted gene testing in 32 families.81 Inherited cardiac disease, mainly LQTS, was diagnosed in 22% of families although testing of the LQTS genes (KCNQ1, KCNH2, SCN5A, KCNE1, and KCNE2) did not contribute to any of the clinical diagnoses. A followup study of 57 families used more comprehensive clinical assessment and targeted genetic evaluation.44 In addition, where available, DNA extracted from formalin-fixed paraffin blocks from 24 SADS cases was used for molecular autopsy of the LQTS genes. Twenty-five families with phenotypic features of cardiac genetic disease underwent targeted mutation analysis. Overall, 53% of families had features of inherited cardiac disease based on the clinical and genetic evaluation, where 70% were diagnosed with LQTS or BrS. Genetic evaluation alone contributed to only 1 (2%) case (identifying a likely LQTS-causing mutation). Other studies have also examined the yield of clinical evaluation and are described in Fig 2. The mean yield of these various studies is 32% with a range of 18%–53%. This variability is very much dependent on the population, the availability of autopsy and expert

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895

Fig 2. Causes of sudden arrhythmic death syndrome as determined by various studies of familial evaluation.44,81,86,135-138 BrS, Brugada syndrome; CPVT, catecholaminergic polymorphic ventricular tachycardia; LQTS, long QT syndrome.

web 4C=FPO

896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959

Translational Research - 2015

Miles and Behr

web 4C=FPO

8

Fig 3. Proportion of molecular diagnoses made in studies of clinical evaluation when testing is targeted to those with a clinical phenotype.44,135-137 LQTS, long QT syndrome.

autopsy, and the rigor of the investigative protocol used. Once a diagnosis is made the actual proportion of molecular diagnoses in those with a clinical phenotype ranges from 23%247% overall (Fig 3). Clinical role. The importance of systematic and comprehensive clinical and genetic evaluation in SADS families is reinforced by recent guidelines.20 Familial evaluation always begins with a detailed analysis of the index case that may give clues to the underlying cause. Pertinent background information includes prior symptoms and clinical encounters (including any previous ECGs) as well as the

circumstances of death and pathologic reports. Firstdegree relatives are then offered full clinical cardiological assessment. Relatives with worrisome symptoms such as syncope, seizures, or palpitations are prioritized for assessment and obligate carriers in a pedigree will also be targeted. This includes history and examination, resting 12-lead ECG (with and without high right ventricular leads), echocardiography, exercise, and ambulatory ECGs, and where indicated signal averaged ECG, provocative testing with sodium channel blockers and/or epinephrine, cardiac imaging, and genetic testing. This

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

Q16

960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023

Translational Research Volume -, Number 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087

Q8

Miles and Behr

is typically conducted in a multidisciplinary environment with access to genetic counseling in a dedicated inherited cardiac disease clinic.82 After comprehensive evaluation, if no abnormalities are detected in first-degree relatives, the risk of future cardiac events related to inherited cardiac disease is generally low.83 In the context of familial assessment, gene testing of SADS relatives is targeted to the phenotype identified in the affected members of the family as described in the previous section. Cascade genetic testing is then performed when a positive clinical diagnosis is made in a family member, enabling identification in other relatives by testing for a specific mutation.84 Coordination of clinical cardiological evaluation and mutation analysis can also help to clarify whether a VUS is likely to be causative that is segregation analysis. The lack of 100% sensitivity of genetic testing must be addressed by appropriate counseling as well as exploration of relatives’ concerns and any psychological implications. In 2014, Erskine et al85 conducted interviews with 50 individuals who had a personal or family history of an arrhythmic syndrome or SCD. Seventy-four percent pursued genetic testing for LQTS or another channelopathy, motivated by factors such as relieving uncertainty surrounding a potential diagnosis, seeking an explanation for a family member’s death, and guiding future medical management. Expectations concerning the scope of genetic testing and individual efficacy in establishing a diagnosis are varied. The main reasons against pursuing testing were fear, denial, and lack of information. Unfortunately a universal and structured approach to deal with the families of young SCD victims is lacking. Indeed, in many countries no formal local guidance exists for evaluating relatives of SADS cases, and an autopsy is often not required. Van der Werf et al86 conducted a community-based intervention study to try to increase autopsy rates of young SCD victims in the Netherlands. Increased awareness in 1 intervention group was promoted via a dedicated helpline for the use of community general practitioners and coroners; information provided by educational meetings and correspondence was also used in another group. The autopsy rate was not significantly improved in either intervention and few families went on to have targeted evaluation in an inherited cardiac disease clinic. This study highlights the difficulties faced in establishing access to suitable services for relatives. Assessment of the cause of death in young SCD cases beyond just exclusion of foul play should be mandatory, as should the onward notification of families to health services. Steps have been taken in Australia, New Zealand, Canada,

9

and the UK, but large gaps in practice remain even in these countries.87-89 MOLECULAR AUTOPSY

Postmortem genetic testing involves the collection of tissue suitable for DNA extraction at autopsy and mutation analysis for specified genes, often described as the ‘‘molecular autopsy.’’ Postmortem sample collection. Postmortem genetic testing has traditionally relied on formalin-fixed paraffin-embedded (FFPE) tissue because of ease of storage and transport. More amenable sample media, such as fresh frozen tissue or EDTA-preserved blood, have not been routinely archived postmortem. In 2008, Carturan et al evaluated different DNA extraction protocols and the feasibility of mutational analysis from archived FFPE tissue. A total of 35 sudden unexpected death syndrome (SUDS) cases were studied; using a number of DNA extraction techniques, nearly one-third of the regions of interest could not be examined.90 In case of SADS, HRS guidelines recommend collection of a tissue sample for subsequent DNA analysis.20 Tissue samples include heart, liver, spleen, and whole EDTA tube or blood spot testing. The National Association of Medical Examiners suggests circumstances in which such specimens should be saved, detailing DNA preservation standards and appropriate counseling of relatives in the context of SCD.91 FFPE should therefore be avoided to allow consistent DNA sampling and more favorable diagnostic yield. Results in research. The first documented report was in 1999 where LQTS was diagnosed from an autopsy specimen of a 19 year-old girl who died after a neardrowning.92 Early work by Tester et al involved mutational analysis of LQTS-associated genes (KCNQ1, KCNH2, and SCN5A)93 and RyR2, implicated in CPVT, in 49 SADS cases.94 Targeted analysis of 18 exons of RyR2 revealed potential CPVT1-causing mutations in 1 of every 7 cases, whereas the yield in LQTS cases was 1 in 5 (20%), giving an overall yield of 35%.47 Further research into molecular autopsy has, however, indicated lower yields. In 2011, Skinner et al95 performed postmortem genetic testing for LQTS in young sudden unexplained death cases. LQT1-6 gene analysis was performed using direct sequencing or denaturing high-performance liquid chromatography. Thirty-three cases underwent testing, and missense mutations in LQTS genes were found in 15%. A more conservative yield was obtained by Winkel et al,96 who examined a young Danish SUDS population of 44

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151

1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215

Translational Research - 2015

Miles and Behr

web 4C=FPO

10

Fig 4. Causes of sudden arrhythmic death syndrome as determined by various molecular autopsy studies.93-95,98,101,134 CPVT, catecholaminergic polymorphic ventricular tachycardia; LQTS, long QT syndrome.

between 2000 and 2006. KCNQ1, KCNH2, and SCN5A were sequenced; 11% carried a mutation in at least 1 of the 3 genes. In a UK study, a molecular diagnosis was made in 14% of 45 cases, and a higher uptake of cascade screening in relatives where a molecular diagnosis had been made at autopsy was also reported.97 The yield of testing LQT and CPVT genes was reexamined by Tester et al98 more recently in an extended series of 173 autopsy-negative SUDS cases that include their original 49 strong cohort. Forty-five likely pathogenic mutations were identified, but the overall yield (25%) was lower than their initial cohort. However, the yield was significantly higher (45%) in those aged ,50 years with a family history of premature SCD. Fig 4 describes the yield of molecular autopsy from this and other recent studies. The mean yield of LQTS and CPVT gene testing was 13% and 11%, respectively, in keeping with more recent conservative estimates. Clinical role. Thus, if the role for the molecular autopsy is to attempt to make a genetic diagnosis in a family then it cannot supplant clinical evaluation given the lower yield. It may complement evaluation by identifying sporadic mutations and focusing evaluation on any offspring of the victim. It may help to confirm or exclude a putative genetic cause in a family especially if a VUS had been found. Occasionally, it may provide a clear well recognized causative variant and direct family evaluation. However, the SNR in molecular autopsy is already as low as 2.25 for the major LQTS and CPVT genes because of the relatively low yields and frequency of rare variation in the general population (see Fig 1). It is therefore not a stand-alone tool at this time. Nonetheless molecular autopsy may have implications for the prevention of additional deaths in other

relatives, and HRS/EHRA guidelines currently recommend genetic testing if circumstantial evidence points toward a clinical diagnosis of LQTS or CPVT44 (Fig 1). This has been reinforced by the more recent clinical testing guidelines that state that an arrhythmia syndrome focused gene panel can be useful.20 NEXT-GENERATION SEQUENCING AND THE FUTURE

Genetic testing for the channelopathies has traditionally involved Sanger sequencing for single gene mutations.99 This method is limited by potential scalability, as more genes with susceptibility to channelopathy have been identified. Advances in technology, massive parallel sequencing or next-generation sequencing, have led to the ability to simultaneously sequence protein coding exons of all genes nucleotide by nucleotide (‘‘the exome,’’ which is approximately 1% of the genome) from much smaller quantities of DNA.100 Bagnall et al101 performed exome sequencing on a subgroup of 28 SUDS cases referred for autopsy in Australia between 2005 and 2009. Three rare variants were discovered in the 3 most common LQTS genes when a narrow panel of the 4 major genes (KCNQ1, KCNH2, SCN5A, and RYR2) was studied. The scope of the molecular autopsy was also expanded to include a wider selection of .70 arrhythmia and cardiomyopathy genes, subsequently revealing a variant in the CAC NA1C gene of a 20 year-old man who died in his sleep. This variant had previously been identified in a LQTS family,102 highlighting that screening additional genes may offer causality beyond the ‘‘usual suspects.’’ Unfortunately two-thirds of the other putative mutations that were identified were VUSs.

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279

Translational Research Volume -, Number -

11

web 4C=FPO

1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343

Miles and Behr

Fig 5. Recommended diagnostic pathway after a sudden unexplained death (adapted from Wong and Behr139). ECG, electrocardiogram.

This highlights the concern over increasing gene panel sizes. The SNR will become even lower for the expanded ‘‘next-generation’’ arrhythmia syndrome and cardiac genetic disease panels because of the enhanced background variation and minimal increase in yield. The family context is currently the only way to extract additional value from the high yield of VUSs by molecular autopsy. This situation will only improve when more is understood about how to assess the likely functional effects of mutations detected in these disorders.

CONCLUSIONS

The molecular autopsy can only occasionally diagnose disease in an index case and direct subsequent family evaluation. Because of methodological issues surrounding determination of variant pathogenicity, the comprehensive next-generation sequencing panel will have to complement careful phenotypic evaluation of family members to inform a targeted genotyping strategy (see Fig 5). Indeed, the utility of genetic testing is only really appreciated in such a context and represents the best chance of developing clinically meaningful results. Only once robust methods are in place for assessing the pathogenicity or disease susceptibility of putative mutations from a large range of potential genetic causes can the molecular autopsy be a truly diagnostic tool. In the interim most of its clinical utility will

be provided by analyzing the 4 major LQTS, BrS, and CPVT genes that we already know and understand best. UNCITED REFERENCES

106,111,120,122,125,130,131. ACKNOWLEDGMENTS

Conflicts of Interest: All authors have read the journal’s authorship agreement and policy on disclosure of potential conflicts of interest and have none to declare. There are no financial or personal relationships with organizations that could potentially be perceived as influencing the research. The manuscript has been read and approved by all the named authors. REFERENCES

1. Deo R, Albert C. Epidemiology and genetics of sudden cardiac death. Circulation 2012;125:620–37. 2. Behr ER, Casey A, Sheppard M, et al. Sudden arrhythmic death syndrome: a national survey of sudden unexplained cardiac death. Heart 2007;93:601–5. 3. Nichol G, Thomas E, Callaway CW, et al. Regional variation in out-of-hospital cardiac arrest incidence and outcome. JAMA 2008;300:1423–31. 4. Chugh SS, Jui J, Gunson K, et al. Current burden of sudden cardiac death: multiple source surveillance versus retrospective death certificate-based review in a large U.S. Community. J Am Coll Cardiol 2004;44:1268–75. 5. Byrne R, Constant O, Smyth Y, et al. Multiple source surveillance incidence and aetiology of out-of-hospital sudden cardiac

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

Q9

1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402 1403 1404 1405 1406 1407

12

1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440 1441 1442 1443 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470 1471

6.

7.

8.

9. 10.

11. 12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

Translational Research - 2015

Miles and Behr

death in a rural population in the west of Ireland. Eur Heart J 2008;29:1418–23. Engelstein ED, Zipes DP. Sudden cardiac death. In: Alexander RW, Schlant RC, Fuster V, eds. The heart, arteries and veins. New York, NY: McGraw-Hill, 1998:1081–112. Myerburg RJ. Cardiac arrest and sudden cardiac death’ in heart disease: a textbook of cardiovascular medicine. 7th ed. Philadelphia: WB Saunders, 2005. NICE Technology Appraisal—implantable cardioverter defibrillators and cardiac resynchronisation therapy for arrhythmias and heart failure. 2014. Available at: http://www.nice.org.uk/ guidance/ta314/chapter/2-clinical-need-and-practice. Accessed September 2014. Winkel BG. Sudden cardiac death in young Danes. Dan Med J 2012;59:B4403. Eckart RE, Scoville SL, Campbell CL, et al. Sudden death in young adults: a 25-year review of autopsies in military recruits. Ann Intern Med 2004;141:829–34. Puranik R, Chow CK, Duflou JA, Kilborn MJ, McGuire MA. Sudden death in the young. Heart Rhythm 2005;2:1277–82. Risgaard B, Winkel BG, Jabbari R, et al. Burden of sudden cardiac death in persons aged 1 to 49 years: nationwide study in Denmark. Circ Arrhythm Electrophysiol 2014;7:205–11. Corrado D, Basso C, Thiene G. Sudden cardiac death in young people with apparently normal heart. Cardiovasc Res 2001;50: 399–408. Corrado D, Fontaine G, Marcus FI, et al. Arrhythmogenic right ventricular dysplasia/cardiomyopathy: need for an international registry. Study Group on Arrhythmogenic Right Ventricular Dysplasia/Cardiomyopathy of the Working Groups on Myocardial and Pericardial Disease and Arrhythmias of the European Society of Cardiology and of the Scientific Council on Cardiomyopathies of the World Heart Federation. Circulation 2000; 101:E101–6. Maron BJ, Doerer JJ, Haas TS, Tierney DM, Mueller FO. Sudden deaths in young competitive athletes: analysis of 1866 deaths in the United States, 1980-2006. Circulation 2009;119: 1085–92. Papadakis M, Sharma S, Cox C, Sheppard MN, Panoulas VF, Behr ER. The magnitude of sudden cardiac death in the young: a death certificate-based review in England and Wales. Europace 2009;11:1353–8. Tester DJ, Medeiros-Domingo A, Will ML, Ackerman MJ. Unexplained drownings and the cardiac channelopathies: a molecular autopsy series. Mayo Clin Proc 2011;86:941–7. Raji H, Behr ER. Unexplained sudden death, focussing on genetics and family phenotyping. Curr Opin Cardiol 2013;28: 19–25. Sandorfi G, Clemens B, Csanadi Z. Electrical storm in the brain and in the heart: epilepsy and Brugada syndrome. Mayo Clin Proc 2013;88:1167–73. Priori SG, Wilde AA, Horie M, et al. HRS/EHRA/APHRS expert consensus statement on the diagnosis and management of patients with inherited primary arrhythmia syndromes: document endorsed by HRS, EHRA, and APHRS in May 2013 and by ACCF, AHA, PACES, and AEPC in June 2013. Heart Rhythm 2013;10:1932–63. Papadakis M, Hariharan R, Behr ER, et al. Sudden cardiac death with autopsy findings of uncertain significance. Circ Arrhythm Electrophysiol 2013;6:588–96. de Noronha SV, Behr ER, Papadakis M, et al. The importance of specialist cardiac histopathological examination in the investigation of young sudden cardiac deaths. Europace 2014;16: 899–907.

23. Trans-Tasman Response Against Sudden Death in the Young (TRAGADY). Post-mortem in sudden unexpected death in the young: guidelines on autopsy practice. 2008. Available at: https://www.cidg.org/webcontent/LinkClick.aspx?fileticket5D O9YIQWqegI%3D&tabid5161. Accessed June 5, 2015. 24. Office of the Chief Coroner for Ontario. Guidelines for the investigation of sudden cardiac death. Memo #08–01. Available at: http://nsgc.org/d/do/978. Accessed June 5, 2015. 25. Sudden Death in the Young Case Registry. University of Michigan. National Heart, Lung, and Blood Institute (NHLBI), the National Institute of Neurological Disorders and Stroke (NINDS), and the Centers for Disease Control and Prevention (CDC). Available at: http://www.nhlbi.nih.gov/news/spotlight/factsheet/frequently-asked-questions-about-sudden-death-young-ca se-registry. Accessed June 5, 2015. 26. Coronary Heart Disease National Service Framework, . Chapter 8. Arrhythmias and sudden cardiac death. London: Department of Health, 2005. 27. Wilde AA, Behr ER. Genetic testing for inherited cardiac disease. Nat Rev Cardiol 2013;10:571–83. 28. Landstrom AP, Ackerman MJ. The Achilles’ heel of cardiovascular genetic testing: distinguishing pathogenic mutations from background genetic noise. Clin Pharmacol Ther 2011;90:496–9. 29. Willerson J, Cohn J, Wellens H, Holmes D. Cardiovascular medicine. 3rd ed. 2007 ISBN-10: 1846281881. P2592. Q10 30. Tester DJ, Ackerman MJ. Genetic testing for potentially lethal, highly treatable inherited cardiomyopathies/channelopathies in clinical practice. Circulation 2011;123:1021–37. 31. Exome Aggregation Consortium (ExAC), Cambridge, MA. Available at: http://exac.broadinstitute.org. Accessed June 3, 2015. 32. Tfelt-Hansen J, Jespersen T, Hofman-Bang J, et al. Ventricular tachycardia in a Brugada syndrome patient caused by a novel deletion in SCN5A. Can J Cardiol 2009;25:156–60. 33. Herman DS, Lam L, Taylor MR, et al. Truncations of titin causing dilated cardiomyopathy. N Engl J Med 2012;366: 619–28. 34. Roberts AM, Ware JS, Herman DS, et al. Integrated allelic, transcriptional, and phenomic dissection of the cardiac effects of titin truncations in health and disease. Sci Transl Med 2015;7: 270ra6. 35. Jabbari J, Jabbari R, Nielsen MW, et al. New exome data question the pathogenicity of genetic variants previously associated with catecholaminergic polymorphic ventricular tachycardia. Circ Cardiovasc Genet 2013;6:481–9. 36. Refsgaard L, Holst AG, Sadjadieh G, Haunsø S, Nielsen JB, Olesen MS. High prevalence of genetic variants previously associated with LQT syndrome in new exome data. Eur J Hum Genet 2012;20:905–8. 37. Norton N, Robertson PD, Rieder MJ, et al. Evaluating pathogenicity of rare variants from dilated cardiomyopathy in the exome era. Circ Cardiovasc Genet 2012;5:167–74. 38. Bell CJ, Dinwiddie DL, Miller NA, et al. Carrier testing for severe childhood recessive diseases by next-generation sequencing. Sci Transl Med 2011;3:65ra4. 39. Grantham R. Amino acid difference formula to help explain protein evolution. Science 1974;185:862–4. 40. Giudicessi JR, Kapplinger JD, Tester DJ, et al. Phylogenetic and physicochemical analyses enhance the classification of rare nonsynonymous single nucleotide variants in type 1 and 2 long-QT syndrome. Circ Cardiovasc Genet 2012;5:519–28. 41. Kapplinger JD, Giudicessi JR, Ye D, et al. Enhanced classification of Brugada syndrome- and long QT syndrome-associated genetic variants in the SCN5A-encoded Nav1.5 cardiac sodium channel. Circ Cardiovasc Genet 2015. Q11

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484 1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500 1501 1502 1503 1504 1505 1506 1507 1508 1509 1510 1511 1512 1513 1514 1515 1516 1517 1518 1519 1520 1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1533 1534 1535

Translational Research Volume -, Number 1536 1537 1538 1539 1540 1541 1542 1543 1544 1545 1546 1547 1548 1549 1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564 1565 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 1580 1581 1582 1583 1584 1585 1586 1587 1588 1589 1590 1591 1592 1593 1594 1595 1596 1597 1598 1599

Miles and Behr

42. Campuzano O, Allegue C, Fernandez A, Iglesias A, Brugada R. Determining the pathogenicity of genetic variants associated with cardiac channelopathies. Sci Rep 2015;5:7953. 43. Itoh H, Shimizu W, Hayashi K, et al. Long QT syndrome with compound mutations is associated with a more severe phenotype: a Japanese multicenter study. Heart Rhythm 2010;7: 1411–8. 44. Behr ER, Dalageorgou C, Christiansen M, et al. Sudden arrhythmic death syndrome: familial evaluation identifies inheritable heart disease in the majority of families. Eur Heart J 2008; 29:1670–80. 45. Schwartz PJ, Stramba-Badiale M, Crotti L, et al. Prevalence of the congenital long-QT syndrome. Circulation 2009;120: 1761–7. 46. Mizusawa Y, Horie M, Wilde AA. Genetic and clinical advances in congenital long QT syndrome. Circ J 2014;78:2827–33. 47. Ackermann MJ, Priori SG, Willems S, et al. HRS/EHRA expert consensus statement on the state of genetic testing for the channelopathies and cardiomyopathies. Europace 2011;13: 1077–109. 48. Hedley PL, Kanters JK, Dembic M, et al. The role of CAV3 in long-QT syndrome: clinical and functional assessment of a caveolin-3/Kv11.1 double heterozygote versus caveolin-3 single heterozygote. Circ Cardiovasc Genet 2013;6:452–61. 49. Wilde A, Antzelevitch C, Borggrefe M, et al. Proposed diagnostic criteria for the Brugada syndrome. Eur Heart J 2002;23: 1648–54. 50. Matsuo K, Kurita T, Inagaki M, et al. The circadian pattern of the development of ventricular fibrillation in patients with Brugada syndrome. Eur Heart J 1999;20:465–70. 51. Amin AS, Klemens CA, Verkerk AO, et al. Fever-triggered ventricular arrhythmias in Brugada syndrome and type 2 long-QT syndrome. Neth Heart J 2010;18:165–9. 52. Wolpert C, Echternach C, Veltmann C, et al. Intravenous drug challenge using flecainide and ajmaline in patients with Brugada syndrome. Heart Rhythm 2005;2:254–60. 53. Raju H, Papadakis M, Govindan M, et al. Low prevalence of risk markers in cases of sudden death due to Brugada syndrome: relevance to risk stratification in Brugada syndrome. J Am Coll Cardiol 2011;57:2340–5. 54. Kamakura S. Epidemiology of Brugada syndrome in Japan and the rest of the world. J Arrhythmia 2013;29:52–5. 55. Gray B, Semsarian C, Sy RW. Brugada syndrome: a heterogeneous disease with a common ECG phenotype? J Cardiovasc Electrophysiol 2014;25:450–6. 56. Schulze-Bahr E, Eckardt L, Breithardt G, et al. Sodium channel gene (SCN5A) mutations in 44 index patients with Brugada syndrome: different incidences in familial and sporadic disease. Hum Mutat 2005;26:61. 57. Bezzina CR, Barc J, Mizusawa Y, et al. Common variants at SCN5A-SCN10A and HEY2 are associated with Brugada syndrome, a rare disease with high risk of sudden cardiac death. Nat Genet 2013;45:1044–9. 58. Antzelevitch C, Brugada P, Borggrefe M, et al. Brugada syndrome: report of the Second Consensus Conference Endorsed by the Heart Rhythm Society and the European Heart Rhythm Association. Circulation 2005;111:659–70. 59. Kapplinger D, Tester DJ, Alders M, et al. An international compendium of mutations in the SCN5A-encoded cardiac sodium channel in patients referred for Brugada syndrome genetic testing. Heart Rhythm 2010;7:33–46. 60. Bezzina CR, Rook MB, Wilde AA. Cardiac sodium channel and inherited arrhythmia syndromes. Cardiovasc Res 2001;49: 257–71.

13

61. Makita N, Behr E, Shimizu W, et al. The E1784K mutation in SCN5A is associated with mixed clinical phenotype of type 3 long QT syndrome. J Clin Invest 2008;118:2219–29. 62. Probst V, Wilde AA, Barc J, et al. SCN5A mutations and the role of genetic background in the pathophysiology of Brugada syndrome. Circ Cardiovasc Genet 2009;2:552–7. 63. Burashnikov E, Pfeiffer R, Barajas-Martinez H, et al. Mutations in the cardiac L-type calcium channel associated with inherited J-wave syndromes and sudden cardiac death. Heart Rhythm 2010;7:1872–82. 64. Crotti L, Marcou CA, Tester DJ, et al. Spectrum and prevalence of mutations involving BrS1-12-susceptibility genes in a cohort of unrelated patients referred for Brugada syndrome genetic testing: implications for genetic testing. J Am Coll Cardiol 2012;60:1410–8. 65. Hu D, Barajas-Martınez H, Pfeiffer R, et al. Mutations in SCN10A are responsible for a large fraction of cases of Brugada syndrome. J Am Coll Cardiol 2014;64:66–79. 66. Reid DS, Tynan M, Braidwood L, Fitzgerald GR. Bidirectional tachycardia in a child: a study using His bundle electrography. Br Heart J 1975;37:339–44. 67. Napolitano C, Bloise R, Memmi M, Priori S. Clinical utility gene card for: catecholaminergic polymorphic ventricular tachycardia (CPVT). Eur J Hum Genet 2014;22 http://dx.doi.org/10.1 038/ejhg.2013.55. published online April 3, 2013. Q12 68. Kumar D, Elliot P. Principles and practice of clinical cardiovascular genetics. Oxford University Press, 2010:P257. ISBN 978-0-19-536895-6. Q13 69. Bastiaenen R, Behr E. Sudden death and ion channel disease: pathophysiology and implications for management. Heart 2011;97:1365–72. 70. Napolitano C, Priori SG, Bloise R. Catecholaminergic polymorphic ventricular tachycardia. In: Pagon RA, Adam MP, Ardinger HH, et al., eds. GeneReviewsÒ [Internet]. Seattle, WA: University of Washington, Seattle:1993–2014. Available at: http://www.ncbi.nlm.nih.gov/books/NBK1289/; 2004. Accessed March 6, 2014. Q14 71. George CH, Jundi H, Thomas NL, Fry DL, Lai FA. Ryanodine receptors and ventricular arrhythmias: emerging trends in mutations, mechanisms and therapies. J Mol Cell Cardiol 2007;42: 34–50. 72. Medeiros-Domingo A, Bhuiyan ZA, Tester DJ, et al. The RYR2-encoded ryanodine receptor/calcium release channel in patients diagnosed previously with either catecholaminergic polymorphic ventricular tachycardia or genotype negative, exercise-induced long QT syndrome: a comprehensive open reading frame mutational analysis. J Am Coll Cardiol 2009;54: 2065–74. 73. Van der Werf C, Nederend I, Hofman N, et al. Familial evaluation in catecholaminergic polymorphic ventricular tachycardia: disease penetrance and expression in cardiac ryanodine receptor mutation-carrying relatives. Circ Arrhythm Electrophysiol 2012; 5:748–56. 74. Maron BJ, Towbin JA, Thiene G, et al. Contemporary definitions and classification of the cardiomyopathies: an American Heart Association Scientific Statement from the Council on Clinical Cardiology, Heart Failure and Transplantation Committee; Quality of Care and Outcomes Research and Functional Genomics and Translational Biology Interdisciplinary Working Groups; and Council on Epidemiology and Prevention. Circulation 2006;113:1807–16. 75. Roma-Rodrigues C, Fernandes A. Genetics of hypertrophic cardiomyopathy: advances and pitfalls in molecular diagnosis and therapy. Appl Clin Genet 2014;7:195–208.

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

1600 1601 1602 1603 1604 1605 1606 1607 1608 1609 1610 1611 1612 1613 1614 1615 1616 1617 1618 1619 1620 1621 1622 1623 1624 1625 1626 1627 1628 1629 1630 1631 1632 1633 1634 1635 1636 1637 1638 1639 1640 1641 1642 1643 1644 1645 1646 1647 1648 1649 1650 1651 1652 1653 1654 1655 1656 1657 1658 1659 1660 1661 1662 1663

14

1664 1665 1666 1667 1668 1669 1670 1671 1672 1673 1674 1675 1676 1677 1678 1679 1680 1681 1682 1683 1684 1685 1686 1687 1688 1689 1690 1691 1692 1693 1694 1695 1696 1697 1698 1699 1700 1701 1702 1703 1704 1705 1706 1707 1708 1709 1710 1711 1712 1713 1714 1715 1716 1717 1718 1719 1720 1721 1722 1723 1724 1725 1726 1727

Translational Research - 2015

Miles and Behr

76. Sheppard M. Practical cardiovascular pathology. 2nd ed. CRC Press, 2011. ISBN-10: 0340981938. 77. van Spaendonck-Zwarts KY, van Rijsingen IA, van den Berg MP, et al. Genetic analysis in 418 index patients with idiopathic dilated cardiomyopathy: overview of 10 years’ experience. Eur J Heart Fail 2013;15:628–36. 78. Mestroni L, Taylor M. Genetics and genetic testing of dilated cardiomyopathy: a new perspective. Discov Med 2013;15:43–9. 79. Sen-Chowdhry S, Morgan RD, Chambers JC, McKenna WJ. Arrhythmogenic cardiomyopathy: etiology, diagnosis, and treatment. Annu Rev Med 2010;61:233–53. 80. Marcus FI, McKenna WJ, Sherrill D, et al. Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia. Circulation 2010;121:1533–41. 81. Behr E, Wood DA, Wright M, et al. Cardiological assessment of first-degree relatives in sudden arrhythmic death syndrome. Lancet 2003;362:1457–9. 82. Kauferstein S, Kiehne N, Jenewein T, et al. Genetic analysis of sudden unexplained death: a multidisciplinary approach. Forensic Sci Int 2013;229:122–7. 83. van der Werf C, Stiekema L, Tan HL, et al. Low rate of cardiac events in first-degree relatives of diagnosis-negative young sudden unexplained death syndrome victims during follow-up. Heart Rhythm 2014;11:1728–32. 84. Garratt CJ, Elliott P, Behr ER, et al. Clinical indications for genetic testing in familial sudden cardiac death syndromes: an HRUK position statement. Heart 2008;94:502–7. 85. Erskine KE, Hidayatallah NZ, Walsh CA, et al. Motivation to pursue genetic testing in individuals with a personal or family history of cardiac events or sudden cardiac death. J Genet Couns 2014;23:849–59. 86. Van der Werf C, Hofman N, Tan HL, et al. Diagnostic yield in sudden unexplained death and aborted cardiac arrest in the young: the experience of a tertiary referral center in The Netherlands. Heart Rhythm 2010;7:1383–9. 87. UK Department of Health. National Service Framework for Coronary Heart Disease—Chapter 8: arrhythmias and sudden cardiac death. 2005. Available at: http://webarchive.nationalarchi ves.gov.uk/20130107105354/http://www.dh.gov.uk/prod_consu m_dh/groups/dh_digitalassets/@dh/@en/documents/digitalasse t/dh_4105280.pdf. Accessed June 4, 2016. 88. Pilmer CM, Porter B, Kirsh JA, et al. Scope and nature of sudden cardiac death before age 40 in Ontario: a report from the cardiac death advisory committee of the office of the chief coroner. Heart Rhythm 2013;10:517–23. 89. Skinner JR, Duflou JA, Semsarian C. Reducing sudden death in young people in Australia and New Zealand: the TRAGADY initiative. Med J Aust 2008;189:539–40. 90. Carturan E, Tester DJ, Brost BC, Basso C, Thiene G, Ackerman MJ. Postmortem genetic testing for conventional autopsy-negative sudden unexplained death: an evaluation of different DNA extraction protocols and the feasibility of mutational analysis from archival paraffin-embedded heart tissue. Am J Clin Pathol 2008;129:391–7. 91. Middleton O, Baxter S, Demo E, et al. National Association of Medical Examiners Position Paper: retaining postmortem samples for genetic testing. Acad Forensic Pathol 2013;3:191–4. 92. Ackerman MJ, Tester DJ, Coburn JP, Edwards WD. Molecular diagnosis of the inherited long-QT syndrome in a woman who died after near-drowning. N Engl J Med 1999;341:1121–5. 93. Tester DJ, Ackerman MJ. Postmortem long QT syndrome genetic testing for sudden unexplained death in the young. J Am Coll Cardiol 2007;49:240–6.

94. Tester DJ, Spoon DB, Valdivia HH, Makielski JC, Ackerman MJ. Targeted mutational analysis of the RyR2encoded cardiac ryanodine receptor in sudden unexplained death: a molecular autopsy of 49 medical examiner/coroner’s cases. Mayo Clin Proc 2004;79:1380–4. 95. Skinner JR, Crawford J, Smith W, et al. Prospective, population-based long QT molecular autopsy study of postmortem negative sudden death in 1 to 40 year olds. Heart Rhythm 2011;8:412–9. 96. Winkel B, Larsen M, Olesen M, Tfelt-Hansen J, Banner J. The prevalence of mutations in KCNQ1, KCNH2, and SCN5A in an unselected national cohort of young sudden unexplained death cases. J Cardiovasc Electrophysiol 2012;23:1092–8. 97. Dean J, Cann F, Corbett M, et al. Molecular autopsy for sudden cardiac death—outcome of a practical approach. Heart Lung Circ 2014;23:e3. 98. Tester DJ, Medeiros-Domingo A, Will ML, Haglund CM, Ackerman MJ. Cardiac channel molecular autopsy: insights from 173 consecutive cases of autopsy-negative sudden unexplained death referred for postmortem genetic testing. Mayo Clin Proc 2012;87:524–39. 99. Churko J, Mantalas G, Synder M, Wu J. Overview of high throughput sequencing technologies to elucidate molecular pathways in cardiovascular diseases. Circ Res 2013;112:1613–23. 100. Chugh SS, Huertas-Vazquez A. Inherited arrhythmia syndromes: exome sequencing opens a new door to diagnosis. J Am Coll Cardiol 2014;63:267–8. 101. Bagnall R, Das KJ, Duflou J, Semsarian C. Exome analysisbased molecular autopsy in cases of sudden unexplained death in the young. Heart Rhythm 2014;11:655–62. 102. Laish-Farkash A, Nof E, Luria D, et al. Incessant bidirectional ventricular tachycardia as the presenting arrhythmia of Andersen-Tawil syndrome in two families. J Interv Card Electrophysiol 2009;24. 277–229. Q15 103. Schott JJ, Alshinawi C, Kyndt F, et al. Cardiac conduction defects associate with mutations in SCN5A. Nat Genet 1999;23: 20–1. 104. Benson DW, Wang DW, Dyment M, et al. Congenital sick sinus syndrome caused by recessive mutations in the cardiac sodium channel gene (SCN5A). J Clin Invest 2003;112:1019–28. 105. Watanabe H, Nogami A, Ohkubo K, et al. Electrocardiographic characteristics and SCN5A mutations in idiopathic ventricular fibrillation associated with early repolarization. Circ Arrhythm Electrophysiol 2011;4:874–81. 106. Hershberger RE, Norton N, Morales A, Li D, Siegfried JD, Gonzalez-Quintana J. Coding sequence rare variants identified in MYBPC3, MYH6, TPM1, TNNC1, and TNNI3 from 312 patients with familial or idiopathic dilated cardiomyopathy. Circ Cardiovasc Genet 2010;3:155–61. 107. Hedley PL, Jørgensen P, Schlamowitz S, Wangari R, MoolmanSmook J, Brink PA. The genetic basis of long QT and short QT syndromes: a mutation update. Hum Mutat 2009;30:1486–511. 108. Bezzina C, Veldkamp MW, van Den Berg MP, et al. A single Na(1) channel mutation causing both long-QT and Brugada syndromes. Circ Res 1999;85:1206–13. 109. Laurent G, Saal S, Amarouch MY, et al. Multifocal ectopic Purkinje-related premature contractions: a new SCN5A-related cardiac channelopathy. J Am Coll Cardiol 2012;60:144–56. 110. Antzelevitch C, Pollevick GD, Cordeiro JM, et al. Loss-of-function mutations in the cardiac calcium channel underlie a new clinical entity characterized by ST-segment elevation, short QT intervals, and sudden cardiac death. Circulation 2007;115: 442–9.

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

1728 1729 1730 1731 1732 1733 1734 1735 1736 1737 1738 1739 1740 1741 1742 1743 1744 1745 1746 1747 1748 1749 1750 1751 1752 1753 1754 1755 1756 1757 1758 1759 1760 1761 1762 1763 1764 1765 1766 1767 1768 1769 1770 1771 1772 1773 1774 1775 1776 1777 1778 1779 1780 1781 1782 1783 1784 1785 1786 1787 1788 1789 1790 1791

Translational Research Volume -, Number 1792 1793 1794 1795 1796 1797 1798 1799 1800 1801 1802 1803 1804 1805 1806 1807 1808 1809 1810 1811 1812 1813 1814 1815 1816 1817 1818 1819 1820 1821 1822 1823 1824 1825 1826 1827 1828 1829 1830 1831 1832 1833 1834 1835 1836 1837 1838 1839 1840 1841 1842 1843 1844 1845 1846 1847 1848 1849 1850 1851 1852 1853 1854 1855

Miles and Behr

111. Ohno S, Zankov DP, Ding WG, et al. KCNE5 (KCNE1L) variants are novel modulators of Brugada syndrome and idiopathic ventricular fibrillation. Circ Arrhythm Electrophysiol 2011;4: 352–61. 112. Bellocq C, van Ginneken AC, Bezzina CR, et al. Mutation in the KCNQ1 gene leading to the short QT-interval syndrome. Circulation 2004;109:2394–7. 113. Bartos DC, Anderson JB, Bastiaenen R, et al. A KCNQ1 mutation causes a high penetrance for familial atrial fibrillation. J Cardiovasc Electrophysiol 2013;24:562–9. 114. Priori SG, Pandit SV, Rivolta I, et al. A novel form of short QT syndrome (SQT3) is caused by a mutation in the KCNJ2 gene. Circ Res 2005;96:800–7. 115. Yang Y, Xia M, Jin Q, et al. Identification of a KCNE2 gain-offunction mutation in patients with familial atrial fibrillation. Am J Hum Genet 2004;75:899–905. 116. Xia M, Jin Q, Bendahhou S, et al. A Kir2.1 gain-of-function mutation underlies familial atrial fibrillation. Biochem Biophys Res Commun 2005;332:1012–9. 117. Yang Y, Liang B, Liu J, et al. Identification of a Kir3.4 mutation in congenital long QT syndrome. Am J Hum Genet 2010;86: 872–80. 118. Medeiros-Domingo A, Tan BH, Crotti L, et al. Gain-of-function mutation S422L in the KCNJ8-encoded cardiac K(ATP) channel Kir6.1 as a pathogenic substrate for J-wave syndromes. Heart Rhythm 2010;7:1466–71. 119. Ha€ıssaguerre M, Chatel S, Sacher F, et al. Ventricular fibrillation with prominent early repolarization associated with a rare variant of KCNJ8/KATP channel. J Cardiovasc Electrophysiol 2009;20:93–8. 120. Bienengraeber M, Olson TM, Selivanov VA, et al. ABCC9 mutations identified in human dilated cardiomyopathy disrupt catalytic KATP channel gating. Nat Genet 2004;36:382–7. 121. Alders M, Koopmann TT, Christiaans I, et al. Haplotype-sharing analysis implicates chromosome 7q36 harboring DPP6 in familial idiopathic ventricular fibrillation. Am J Hum Genet 2009;84: 468–76. 122. Hayashi T, Arimura T, Ueda K, et al. Identification and functional analysis of a caveolin-3 mutation associated with familial hypertrophic cardiomyopathy. Biochem Biophys Res Commun 2004;313:178–84. 123. Ishikawa T, Sato A, Marcou CA, et al. A novel disease gene for Brugada syndrome: sarcolemmal membrane-associated protein gene mutations impair intracellular trafficking of hNav1.5. Circ Arrhythm Electrophysiol 2012;5:1098–107. 124. Priori SG, Napolitano C, Memmi M, et al. Clinical and molecular characterization of patients with catecholaminergic polymorphic ventricular tachycardia. Circulation 2002;106:69–74. 125. Koop A, Goldmann P, Chen SR, Thieleczek R, Varsanyi M. ARVC-related mutations in divergent region 3 alter functional

126.

127.

128.

129.

130.

131.

132.

133.

134.

135.

136.

137.

138.

139.

15

properties of the cardiac ryanodine receptor. Biophys J 2008; 94:4668–77. Lahat H, Pras E, Olender T, et al. A missense mutation in a highly conserved region of CASQ2 is associated with autosomal recessive catecholamine-induced polymorphic ventricular tachycardia in Bedouin families from Israel. Am J Hum Genet 2001;69:1378–84. Roux-Buisson N, Cacheux M, Fourest-Lieuvin A, et al. Absence of triadin, a protein of the calcium release complex, is responsible for cardiac arrhythmia with sudden death in human. Hum Mol Genet 2012;21:2759–67. Nyegaard M, Overgaard MT, Søndergaard MT, et al. Mutations in calmodulin cause ventricular tachycardia and sudden cardiac death. Am J Hum Genet 2012;91:703–12. Crotti L, Johnson CN, Graf E, et al. Calmodulin mutations associated with recurrent cardiac arrest in infants. Circulation 2013; 127:1009–17. Kruse M, Schulze-Bahr E, Corfield V, et al. Impaired endocytosis of the ion channel TRPM4 is associated with human progressive familial heart block type I. J Clin Invest 2009;119:2737–44. Liu H, Chatel S, Simard C, et al. Molecular genetics and functional anomalies in a series of 248 Brugada cases with 11 mutations in the TRPM4 channel. PLoS One 2013;8:e54131. Schulze-Bahr E, Neu A, Friederich P, et al. Pacemaker channel dysfunction in a patient with sinus node disease. J Clin Invest 2003;111:1537–45. Ueda K, Hirano Y, Higashiuesato Y, et al. Role of HCN4 channel in preventing ventricular arrhythmia. J Hum Genet 2009;54: 115–21. Larsen MK, Berge KE, Leren TP, et al. Postmortem genetic testing of the ryanodine receptor 2 (RYR2) gene in a cohort of sudden unexplained death cases. Int J Legal Med 2013;127: 139–44. Tan HL, Hofman N, van Langen IM, van der Wal AC, Wilde AA. Sudden unexplained death: heritability and diagnostic yield of cardiological and genetic examination in surviving relatives. Circulation 2005;112:207–13. McGorrian C, Constant O, Harper N, et al. Family-based cardiac screening in relatives of victims of sudden arrhythmic death syndrome. Europace 2013;15:1050–8. Kumar S, Peters S, Thompson T, et al. Familial cardiological and targeted genetic evaluation: low yield in sudden unexplained death and high yield in unexplained cardiac arrest syndromes. Heart Rhythm 2013;10:1653–60. Mellor G, Raju H, de Noronha SV, et al. Clinical characteristics and circumstances of death in the sudden arrhythmic death syndrome. Circ Arrhythm Electrophysiol 2014;7:1078–83. Wong LC, Behr ER. Sudden unexplained death in infants and children: the role of undiagnosed inherited cardiac conditions. Europace 2014c;16:1706–13.

REV 5.2.0 DTD  TRSL924_proof  2 July 2015  4:46 pm  ce

1856 1857 1858 1859 1860 1861 1862 1863 1864 1865 1866 1867 1868 1869 1870 1871 1872 1873 1874 1875 1876 1877 1878 1879 1880 1881 1882 1883 1884 1885 1886 1887 1888 1889 1890 1891 1892 1893 1894 1895 1896 1897 1898 1899 1900 1901 1902 1903 1904 1905 1906 1907 1908 1909 1910 1911 1912 1913 1914 1915 1916 1917 1918 1919

The role of genetic testing in unexplained sudden death.

Most sudden deaths are because of a cardiac etiology and are termed sudden cardiac death (SCD). In younger individuals coronary artery disease is less...
2MB Sizes 5 Downloads 11 Views