Next Article in Journal
Diagnostic Performance of a Deep Learning-Powered Application for Aortic Dissection Triage Prioritization and Classification
Previous Article in Journal
Triple-Negative Breast Cancer: Molecular Particularities Still a Challenge
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Identifying the Pathogenic Variants in Heart Genes in Vietnamese Sudden Unexplained Death Victims by Next-Generation Sequencing

1
Department of Forensic Medicine, Hanoi Medical University, 1 Ton That Tung Str., Dongda, Hanoi 100000, Vietnam
2
Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Caugiay, Hanoi 100000, Vietnam
3
Department of Pathology, National Cancer Hospital, 43 Quan Su Str., Hoan Kiem, Hanoi 100000, Vietnam
4
Cardiovascular Intensive Care Unit, Heart Institute, 108 Military Central Hospital, 1B Tran Hung Dao Str., Hai Ba Trung, Hanoi 100000, Vietnam
5
Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Caugiay, Hanoi 100000, Vietnam
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Diagnostics 2024, 14(17), 1876; https://doi.org/10.3390/diagnostics14171876
Submission received: 18 July 2024 / Revised: 16 August 2024 / Accepted: 17 August 2024 / Published: 27 August 2024
(This article belongs to the Section Pathology and Molecular Diagnostics)

Abstract

:
In forensics, one-third of sudden deaths remain unexplained after a forensic autopsy. A majority of these sudden unexplained deaths (SUDs) are considered to be caused by inherited cardiovascular diseases. In this study, we investigated 40 young SUD cases (<40 years), with non-diagnostic structural cardiac abnormalities, using Targeted NGS (next-generation sequencing) for 167 genes previously associated with inherited cardiomyopathies and channelopathies. Fifteen cases identified 17 variants on related genes including the following: AKAP9, CSRP3, GSN, HTRA1, KCNA5, LAMA4, MYBPC3, MYH6, MYLK, RYR2, SCN5A, SCN10A, SLC4A3, TNNI3, TNNI3K, and TNNT2. Of these, eight variants were novel, and nine variants were reported in the ClinVar database. Five were determined to be pathogenic and four were not evaluated. The novel and unevaluated variants were predicted by using in silico tools, which revealed that four novel variants (c.5187_5188dup, p.Arg1730llefsTer4 in the AKAP9 gene; c.1454A>T, p.Lys485Met in the MYH6 gene; c.2535+1G>A in the SLC4A3 gene; and c.10498G>T, p.Asp3500Tyr in the RYR2 gene) were pathogenic and three variants (c.292C>G, p.Arg98Gly in the TNNI3 gene; c.683C>A, p.Pro228His in the KCN5A gene; and c.2275G>A, p.Glu759Lys in the MYBPC3 gene) still need to be further verified experimentally. The results of our study contributed to the general understanding of the causes of SUDs. They provided a scientific basis for screening the risk of sudden death in family members of victims. They also suggested that the Targeted NGS method may be used to identify the pathogenic variants in SUD victims.

1. Introduction

In forensic medicine, an autopsy has determined the cause of only two-thirds of sudden deaths, and the remaining cases are called sudden unexplained death (SUD) [1]. The majority of SUD cases are believed to be caused by cardiovascular diseases leading to sudden cardiac death (SCD) in young people. Sudden death is usually defined as a person who appeared healthy for 24 h before the onset of symptoms that led to death [2]. SCD accounts for 15–20% of all deaths in the general population, with incidence ranging from 40 to 100 per 100,000 people annually [3]. The main causes of SCD include cardiomyopathies, channelopathies, and ischemic heart diseases. About two-thirds of SCD cases had structural abnormalities that were evident at autopsy [1,4,5]; meanwhile, no structural abnormalities of the heart were found in the remaining cases. SCD may result from complications of cardiomyopathy causing early malignant arrhythmias, leading to death before the development of the cardiomyopathy phenotype. Especially in young people, SCD is often due to cardiomyopathies such as hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM), and arrhythmogenic cardiomyopathy (ACM); channelopathies including ion channel disorders such as long QT syndrome (LQTS), short QT syndrome (SQTS), Brugada syndrome (BrS), catecholaminergic polymorphic ventricular tachycardia (CPVT), progressive cardiac conduction disorder, and early repolarization syndrome; or ischemic heart diseases [6,7,8].
In atrioventricular canal disease, long QT syndrome (LQTS) is caused by a heart’s electrical system that takes too long to recharge. The disease can lead to life-threatening arrhythmias and sudden cardiac death, primarily at a young age [9]. At least 15 genes encoding different ion channels have been identified as causes of LQTS [10]. Short QT syndrome (SQTS) is considered one of the most dangerous diseases associated with sudden cardiac death, characterized by a short QT interval on the electrocardiogram. Potentially pathogenic variants have been reported in five genes (CACNA2D1, KCNH2, KCNJ2, KCNQ1, and SLC4A3); all are inherited in an autosomal dominant [2,11,12]. Brugada syndrome (BrS) is a cardiac channelopathy caused by rare mutations in the SCN5A gene, which encodes the alpha-subunit of the voltage-dependent cardiac Na+ channel protein (Nav1.5) [2,13]. SCN5A-positive BrS patients often exhibit severe conduction abnormalities [14] and have more severe arrhythmic outcomes than SCN5A-negative patients [15,16].
CPVT is a malignant arrhythmia syndrome characterized by bidirectional or polymorphic VT during physical or emotional stress, which leads to a significantly high mortality rate (30% SCD before age 40) if the patient is not detected and treated [17]. Mutations in the RYR2 gene were found in ≈60% of patients with CPVT [18] and concentrated mainly in three specific regions of the ryanodine receptor 2 (RYR2) [18,19]. Additionally, mutations in CASQ2 have been identified as causing a less common but more severe form of CPVT [20]. Mutations in RYR2 and CASQ2 result in diastolic calcium release from the SR and cause arrhythmias. Recently, two other genes, CALM1 and TRDN, involved in calcium balance, have also been identified as causing CPVT [21,22].
In addition to arrhythmias, progressive cardiac conduction disorders, early repolarization syndrome, and ischemic heart disease are also of concern and are considered causes of sudden death. Progressive cardiac conduction disorder (PCCD), characterized by delayed impulse conduction through the His–Purkinje system with right or left bundle branch block, is a heart disease leading to complete atrioventricular (AV) block, syncope, and SCD. Genetic forms of PCCD often overlap or co-occur with other heart diseases. Currently, 20 genes encoding cardiac ion channels and regulatory proteins, protein kinases, structural proteins, and transcription factors are associated with different forms of PCCD [23]. In recent years, early repolarization syndrome (ERS), which is caused by variants in eight ion channel genes (KCNJ8, ABCC9, SCN5A, SCN10A, KCND3, CACNA1C, CACNB2b, and CACNA2D1), has been considered a hereditary SCD syndrome [24,25,26,27,28,29]. In addition, cerebral small vessel disease (CSVD), a clinically and genetically heterogeneous group of disorders, is also considered a leading cause of myocardial infarction and sudden death [30]. Although most cases of CSVD are sporadic, recent reports have identified genes associated with CSVD, including NOTCH3, HTRA1, CTSA, GLA, COL4A1/A2, TREX1, and CSF1R [31].
Other causes of SCD were related to cardiomyopathy including hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM), and arrhythmogenic cardiomyopathy (ACM). HCM is the most common form that leads to left ventricular outflow tract obstruction, coronary ischemia due to the narrowing of small blood vessels, and severe arrhythmias [32], which is the main cause of sudden cardiovascular death [33]. Most of these cases are caused by pathogenic variants in the core sarcomeric genes (MYH7, MYBPC3, TNNT2, TNNI3, MYL2, MYL3, TPM1, ACTC1) [34]. To date, about 100 genes associated with cardiomyopathy have been reported [35].
An autopsy aims to determine the cause of death in the victims and predict the risks to family members. However, until now, in 40% of SUD cases, the causes could not be determined by autopsy [36]. Therefore, genetic testing has been proposed in forensic medicine to investigate sudden death through genetic analysis [37]. It is especially useful in cases of negative traditional autopsies or SUD due to underlying genetic arrhythmic heart disease. Early molecular autopsies relied on Sanger sequencing. Although this is an accurate and easy-to-use method, it is limited because it has low throughput and can only be used to analyze a subset of small genes. Recently, next-generation sequencing (NGS) technologies have allowed exome/genome examination, providing an increase in the detection of pathogenic variants and the discovery of newer genotype–phenotype associations [38,39,40]. NGS can investigate large numbers of genes and thus aid in clinical investigations and increase the likelihood of determining the cause of death.
In this study, we performed Targeted NGS sequencing in autopsy-negative SUD victims to identify genetic alterations associated with cardiomyopathy and channelopathies that may explain their causes of death.

2. Materials and Methods

2.1. Subjects

A total of 40 young SUD cases (30 men and 10 women) collected from the Department of Forensic Medicine, Hanoi Medical University, Vietnam, were investigated for molecular forensics. The victims were all <40 years old (from 1 to 40 years old, median age of 28.85) and died suddenly. Autopsy evaluations as well as toxicology screening were negative. The victims were known to be healthy 24 h before death, and none had a family history of SCD (Table 1).

2.2. Targeted Next-Generation Sequencing

To determine the cause of sudden death in the victims, we performed Targeted next-generation sequencing using a gene panel of 167 genes (Table S1) associated with cardiomyopathy and channelopathies. Genomic DNA was extracted from blood samples using the Qiagen DNA mini kit (QIAGEN, Hilden, German) following the manufacturer’s instructions. DNA concentration was determined using a Thermo Scientific NanoDrop spectrophotometer (Waltham, MA, USA). Sequencing was performed on a Nextseq 500 (Illumina, CA, USA). Variants were compared and annotated based on the GRCh38/hg19 reference genome sequence. Variants in the genes associated with cardiomyopathy and channelopathies were identified based on ACMG guidelines (The American College of Medical Genetics and Genomics) [41].

2.3. Sanger Sequencing

Sanger sequencing was performed to confirm the variants were found in these cases. PCR products were purified with the Qiagen PCR Purification kit (QIAGEN, Hilden, Germany) and sequenced on the ABI PRISM 3500 Genetic Analyzer machine (Thermo Fisher Scientific Inc., Waltham, MA, USA) in both directions using the primers that were used in the initial PCR reaction. The sequencing data were analyzed using BioEdit 7.2.5 software.

2.4. In Silico Prediction

The influence of any novel nucleotide changes was evaluated with the following in silico prediction tools: CADD [https://cadd.gs.washington.edu/snv; accessed: 1 July 2024], FATHMM [http://fathmm.biocompute.org.uk/inherited.html; accessed: 1 July 2024], Mutation Taster [https://www.mutationtaster.org/ accessed: 2 July 2024], PhD-SNP [https://snps.biofold.org/phd-snp/phd-snp.html; accessed: 1 July 2024], PolyPhen 2 [http://genetics.bwh.harvard.edu/pph2/; accessed: 1 July 2024], SNP&GO [https://snps-and-go.biocomp.unibo.it/snps-and-go/; accessed: 1 July 2024] for misene variants and EX-SKIP [https://ex-skip.img.cas.cz/; accessed: 1 July 2024], Fruitfly [https://www.fruitfly.org/seq_tools/splice.html; accessed: 1 July 2024], MaxEntScan [http://hollywood.mit.edu/burgelab/maxent/Xmaxentscan_scoreseq.html; accessed: 1 July 2024], NetGene2 v.2.42 [https://services.healthtech.dtu.dk/; accessed: 1 July 2024], and Spliceailookup [https://spliceailookup.broadinstitute.org/; accessed: 1 July 2024] for splicing variants.

3. Results

In this study, 40 Vietnamese SUD victims were investigated through molecular forensics by using Targeted NGS sequencing. The detected variants were confirmed by Sanger sequencing in these cases (Figure 1 and Figure 2). The results showed that 25 (62.5%) victims harbored no variants or benign variants, and 15 (37.5%) victims had 17 variants identified on the genes associated with cardiomyopathy and channelopathies. Among them, eight of these variants were novel, five variants were pathogenic variants in the ClinVar database, and four variants were published in ClinVar but with uncertain significance (Table 2). Seven variants (41%) were detected in genes associated with cardiomyopathies including CSRP3, LAMA4, MYH6, MYBPC3, TNNI3, TNNI3K, and TNNT2. Eight variants (47%) were detected in genes related to cardiac channelopathies such as AKAP9, GSN, KCNA5, SCN5A, SCN10A, SLC4A3, and RYR2. Two other variants were detected in the HTRA1 and MYLK genes that were reported in patients with CSVD and extensive aortic, respectively. The novel variants and variants not evaluated on the ClinVar database were predicted by prediction tools. Prediction results showed that four novel variants including variants c.10498G>T, p.Asp3500Tyr (in the RYR2 gene); c.5187_5188dup, p.Arg1730llefsTer4 (in the AKAP9 gene); c.1454A>T, p.Lys485Met (in the MYH6 gene); and c.2535+1G>A (in the SLC4A3 gene), and an uncertain significance variant c.292C>G, p.Arg98Gly in the TNNI3 gene, are considered likely to cause disease (A and B in Table 3 and Table S2). In addition, variant c.683C>A, p.Pro228His in the KCNA5 gene was predicted as disease-causing by the following in silico tools: CADD (with score 24.1), Mutation taster (with score 77), Polyphen 2 (with score 1.000), and SNP&GO (with score R17). Two variants c.2275G>A, p.Glu759Lys (in the MYBPC3 gene) and c.5025A>T, p.Glu1675Asp (in the LAMA4 gene) were predicted as disease-causing by the following in silico tools: CADD (with score 26.7 and 23.6, respectively), Mutation taster (with score 56 for variant in MYBP3), PhD-SNP (with score R14 for variant in LAMA4), and Polyphen 2 (with score 1.000 and 0.998, respectively). Variants c.298C>T, p.Arg100Cys (in CSRP3), c.4840G>A, p.Glu1614Lys (in MYLK), and c.872T>C, p.Ile291Thr (in GSN) were predicted as probably damaging by the CADD and Polyphen 2 tools. However, variant c.9215G>T, p.Gly3072Val in the AKAP9 gene was assessed by prediction tools as a neutral influence variant. These variants need to be further evaluated to determine the extent of their impact.

4. Discussion

In this study, 40 Vietnamese victims who were diagnosed with SUD at ages < 40 years old were sequenced using Targeted NGS with a panel gene consisting of 167 cardiac disease-associated genes. Fifteen (37.5%) victims were identified with 17 variants in genes associated with cardiomyopathy and channelopathies (Table 2). Molecular forensics studies in young individuals with negative autopsies have identified putative pathogenic variants in genes associated with channelopathies in 11 to 26% of cases [11,42]. In 2014, Bagnall et al. [43] first performed whole exome sequencing (WES) in 28 cases of adolescent SUDs and identified three rare variants associated with LQTS and six variants related to channelopathies and cardiomyopathy. In another study, the authors performed gene panel analysis (including 69, 98, or 101 genes) on 51 SUD cases and WES on another 62 SUD cases, finding variants in 31 cases (27%) [36]. Hata et al. [44] used a panel of 70 genes to evaluate 25 SUD cases and identified five known variants and 10 novel variants predicted to be pathogenic by in silico analysis. The variants included three channelopathy-associated genes (RYR2, CACNA1C, and ANK2), three HCM- or DCM-associated genes (MYH7, LDB3, and PRKAG2), five ACM-related genes (PKP2, JUP, DSG2, DSP, and TMEM43), and two cardiac transcription factor genes (TBX5 and GATA4). The authors also identified the simultaneous presence of two heterozygous variants in 3 of 25 cases and 2 cases carrying three or more variants [44]. These data support the hypothesis that the “single gene disease” model may not apply to all cases of SUD, which can sometimes occur because of the interaction of multiple variants [45].
Another study performed with a gene panel of 100 genes in 61 SUD cases found that 21 (34%) individuals carried variants that may have a functional effect. Ten (40%) of these variants were in genes associated with cardiomyopathy and fifteen (60%) were in genes associated with cardiac channelopathies [8]. Previous reports suggested that cardiomyopathy often has a variety of manifestations due to incomplete penetrance, so initial phenotypic changes may not be seen at autopsy or may be considered nonspecific or within the normal range. Variants associated with cardiomyopathy and structural alterations of the heart can give rise to arrhythmias—and in some cases, cause disorders through regulation of cardiac channel function [46,47,48,49]. Primary electrical disorders are thought to be caused by variants in genes encoding ion channels and cardiomyopathy [50].
Cerrone and Priori’s study also provided evidence for an association between channelopathies and hereditary cardiomyopathy with arrhythmogenic substrate-related genes thought to be involved in abnormal structures of the myocardium and primary electrical disorders [51]. In these cases, an autopsy showed that the heart had a normal structure and that the arrhythmia arose from abnormalities in the electrical function of the heart [50]. These primary electrical diseases are mainly caused by variants in the genes encoding the heart’s ion channels and receptors. Most variants that lead to ion channel dysfunction alter the electrical activity in the heart and predispose patients to fatal arrhythmias without changing the morphology of the heart tissue. Identifying genetic factors that lead to SCD is important because genetic testing can contribute to diagnosis and risk prediction.
In our study, five cases carried a known pathogenic variant in the following genes: TNNI3K (c.2302G>C, p.Glu768Gln, RCV000768402.5/Pathogenic, P3); HTRA1 (c.496C>T, p.Arg166Cys, RCV002291765.4/Pathogenic, P6); TNNT2 (c.452G>A, p.Arg151Gln, RCV000796707.6/Pathogenic, P8); SCN10A (c.2158G>A, p.Asp720Asn, VCV000532067.8/Pathogenic, P9); and SCN5A (c.515A>G, p.His184Arg, VCV000201540.5/Likely pathogenic, P12). Another four patients carried known variants in the following genes: KCNA5 (c.683C>A, p.Pro228His, VCV002202820.2, P4); MYBPC3 (c.2275G>A, p.Glu759Lys, VCV000843772.16, P5); CSRP3 (c.298C>T, p.Arg100Cys, VCV000851709.9, P10); and TNNI3 (c.292C>G, p.Arg98Gly, VCV001331910.2, P13). Based on the prediction results using prediction tools, three of these variants (KCNA5, P4; MYBPC3, P5; TNNI3, P13) were predicted to be likely to cause disease in the cases. However, the variant c.298C>T, p.Arg100Cys in the CSRP3 gene was not confirmed as the cause of the disease in case P13 (A in Table 3).
Among four cases that carried novel variants in the following genes: RYR2 (c.10498G>T, p.Asp3500Tyr, P1), AKAP9 (c.5187_5188dup, p.Arg1730llefsTer4, P2), MYH6 (c.1454A>T, p.Lys485Met, P7), and GSN (c.872T>C, p.Ile291Thr, P14), three variants (in the RYR2, AKAP9, and MYH6 genes) were evaluated as like pathogenic variants. Case P11 carried the following novel variants: c.4840G>A, p.Glu1614Lys (in the MYLK gene) and c.9215G>T, p.Gly3072Val (in the AKAP9 gene); however, variant c.4840G>A, p.Glu1614Lys in the MYLK gene was assessed as potentially damaging by CADD, version 1.7 (score: 36.0) and Polyphen version 2 (score: 0.611) software but as neutral by PhD-SNP (score: RI2) software. The variant c.9215G>T, p.Gly3072Val in the AKAP9 gene was evaluated as a neutral variant by CADD (score: 17.6), FATHMM (score: 3.76), and PhD-SNP (score: RI8) software while being evaluated as potentially harmful by Polyphen 2 (score: 0.973). Likewise, case P15 also carried the following novel variants: c.2535+1G>A (in the SLC4A3 gene) and c.5025A>T, p.Glu1675Asp (in the LAMA4 gene). The variant c.2535+1G>A in the SLC4A3 gene was evaluated as “exon-skipping” by the EX-SKIP software, “donor loss” by NetGene 2 version 2.42 and Spliceailookup software and as a damaging variant by MaxEntScan software (B in Table 3 and Table S2). The variant c.5025A>T, p.Glu1675Asp in the LAMA4 gene was assessed as a disease variant by PhD-SNP (score: RI4) and “probably damaging” by CADD (score: 23.6) and Polyphen 2 (score: 0.998), but it was assessed as a “tolerated variant” by FATHMM (score: −1.32) and “benign” by Mutation taster (score: 21) (A in Table 3). Based on the above-predicted results, the c.2535+1G>A variant in the SLC4A3 gene can be considered the cause of the disease in case P15.
Tester et al. [52] also showed that at least one-third of SUD cases in young people were found to be due to variants in the receptor gene, of which variants in cardiac ryanodine (RYR2) account for nearly 14% of cases. AKAP9 variants were associated with types of cardiovascular including LQTS type 11 [53,54,55,56], Brugada syndrome [57,58], sudden death of unknown cause [59,60,61,62], severe ventricular arrhythmias [63,64], and cardiomyopathy [65]. Huynh and colleagues confirmed that AKAP9 variants are associated with fatal arrhythmias and sudden cardiac death [9]. Of interest, some patients may carry the AKAP9 variant and other genes associated with channelopathies, suggesting the complexity of multifactorial and polygenic inheritance in these patients [54,61]. The variant in the SLC4A3 anion exchanger gene is considered a cause of SQTS in the patients [66]. In Christiansen’s study, about a quarter of patients with SQTS carried potentially pathogenic variants in SLC4A3, representing the most common variants and emphasizing the importance of the inclusion of SLC4A3 in the genetic screening of patients with SQTS or onset sudden cardiac death [67].
In addition, stroke is the main cause of death in young people. However, the actual genetic cause of stroke is still unknown. Cerebral small vessel disease (CSVD), often causing stroke, is characterized by young adult onset [68] and is considered to be caused by variants in the high temperature-demanding serine peptidase A1 (HTRA1) gene [69]. The HTRA1 gene encodes a serine enzyme that mediates cell signaling and protein degradation and plays an important role in vascular integrity [70]. Autosomal dominant genetic variants on HTRA1 have been reported in families with CSVD in Italy, Spain, Greece, and China [61,71,72,73]. Heterozygous HTRA1 variants may be responsible for impaired HTRA1 protease activity or form stabilizers leading to the phenotype in patients [74].
In this study, using Targeted NGS sequencing and prediction tools, we identified variations in related genes in 15 SUD cases out of a total of 40 studied cases. Of these 15 cases, the cause of sudden death in 12 cases was identified, and three cases (P10, P11, and P14) required further evaluation to determine the exact cause of sudden death. Forensic molecular autopsy has recently proven valuable in SUD cases and is increasingly used to explain these cases.
There are some limitations in our study. For the first limitation, we used a gene panel with 167 genes related to cardiomyopathy and channelopathies which was reported previously, leading to the possibility of not detecting the variants in other related genes. The second limitation of our study is that we did not conduct studies to evaluate the influence of variants, so we have not yet reached accurate conclusions about the cause of SUD in some cases.

5. Conclusions

In our study, 40 SUD cases were investigated with Targeted NGS sequencing, and 17 variants in genes associated with cardiomyopathy and channelopathies in 15 cases were identified. By predicting the pathogenicity of the variants found using prediction software, the cause of death was determined in 12 cases. The obtained results showed that next-generation sequencing is a tool that can be used in molecular forensics to determine the cause of disease in SUD cases.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/diagnostics14171876/s1, Table S1. List of 167 genes in panel genes used in this study. Table S2. Prediction results using in silico tools for splicing variant SLC4A3: c.2535+1G>A.

Author Contributions

Conceptual framework design, T.N.T.; writing—original draft preparation, N.T.K.L.; writing—review and editing, T.N.T., N.T.K.L., H.L.S., T.T.V., D.D.V., L.T.T., N.T.X. and N.H.H.; data curation and investigation, T.N.T., H.N.T. and N.V.T.; methodology and software, N.T.K.L. and N.V.T. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Vietnam Academy of Science and Technology for The Excellent research team of the Institute of Genome Research, NCXS.01.03/23-25.

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of the Hanoi Medical University (Approval No. 913/GCN-HDDDNCYSH-DHYHN).

Informed Consent Statement

This study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of the Hanoi Medical University (Approval No. 913/GCN-HDDDNCYSH-DHYHN). Written informed consent was obtained from the patient’s parents for the publication of any potentially identifiable images or data included in this article.

Data Availability Statement

Data is contained within the article or Supplementary Materials.

Acknowledgments

We would like to thank the patients and their families who participated in this study.

Conflicts of Interest

All authors declare that they have no conflicts of interest.

References

  1. Winkel, B.G.; Holst, A.G.; Theilade, J.; Kristensen, I.B.; Thomsen, J.L.; Ottesen, G.L.; Bundgaard, H.; Svendsen, J.H.; Haunsø, S.; Tfelt-Hansen, J. Nationwide Study of Sudden Cardiac Death in Persons Aged 1–35 Years. Eur. Heart J. 2011, 32, 983–990. [Google Scholar] [CrossRef] [PubMed]
  2. Priori, S.G.; Blomstrom-Lundqvist, C.; Mazzanti, A.; Bloma, N.; Borggrefe, M.; Camm, J.; Elliott, P.M.; Fitzsimons, D.; Hatala, R.; Hindricks, G.; et al. 2015 ESC Guidelines for the Management of Patients with Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death the Task Force for the Management of Patients with Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death of the Europea. Eur. Heart J. 2015, 36, 2793–2867. [Google Scholar] [CrossRef]
  3. Wong, C.X.; Brown, A.; Lau, D.H.; Chugh, S.S.; Albert, C.M.; Kalman, J.M.; Sanders, P. Epidemiology of Sudden Cardiac Death: Global and Regional Perspectives. Heart Lung Circ. 2019, 28, 6–14. [Google Scholar] [CrossRef]
  4. Wisten, A.; Forsberg, H.; Krantz, P.; Messner, T. Sudden cardiac death in 15–35-year olds in Sweden during 1992–99. J. Intern. Med. 2002, 252, 529–536. [Google Scholar] [CrossRef] [PubMed]
  5. Puranik, R.; Chow, C.K.; Duflou, J.A.; Kilborn, M.J.; McGuire, M.A. Sudden Death in the Young. Heart Rhythm 2005, 2, 1277–1282. [Google Scholar] [CrossRef] [PubMed]
  6. Eckart, R.E.; Shry, E.A.; Burke, A.P.; McNear, J.A.; Appel, D.A.; Castillo-Rojas, L.M.; Avedissian, L.; Pearse, L.A.; Potter, R.N.; Tremaine, L.; et al. Sudden Death in Young Adults: An Autopsy-Based Series of a Population Undergoing Active Surveillance. J. Am. Coll. Cardiol. 2011, 58, 1254–1261. [Google Scholar] [CrossRef]
  7. Tester, D.J.; Medeiros-Domingo, A.; Will, M.L.; Haglund, C.M.; Ackerman, M.J. 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, 524–539. [Google Scholar] [CrossRef]
  8. Christiansen, S.L.; Hertz, C.L.; Ferrero-Miliani, L.; Dahl, M.; Weeke, P.E.; Camp, L.; Ottesen, G.L.; Frank-Hansen, R.; Bundgaard, H.; Morling, N. Genetic Investigation of 100 Heart Genes in Sudden Unexplained Death Victims in a Forensic Setting. Eur. J. Hum. Genet. 2016, 24, 1797–1802. [Google Scholar] [CrossRef]
  9. Huynh, M.T.; Proust, A.; Bouligand, J.; Popescu, E. AKAP9-Related Channelopathy: Novel Pathogenic Variant and Review of the Literature. Genes 2022, 13, 2167. [Google Scholar] [CrossRef]
  10. Wu, J.; Ding, W.G.; Horie, M. Molecular Pathogenesis of Long QT Syndrome Type 1. J. Arrhythmia 2016, 32, 381–388. [Google Scholar] [CrossRef]
  11. Hedley, P.L.; Jorgensen, P.; Schlamowitz, S.; Wangari, R.; Moolman-Smook, J.; Brink, P.A.; Kanters, J.K.; Corfield, V.A.; Christiansen, M. The Genetic Basis of Long QT and Short QT Syndromes: A Mutation Update. Hum. Mutat. 2009, 30, 1486–1511. [Google Scholar] [CrossRef]
  12. Tester, D.J.; Ackerman, M.J. Cardiomyopathic and Channelopathic Causes of Sudden Unexplained Death in Infants and Children. Annu. Rev. Med. 2009, 60, 69–84. [Google Scholar] [CrossRef]
  13. Wijeyeratne, Y.D.; Tanck, M.W.; Mizusawa, Y.; Batchvarov, V.; Barc, J.; Crotti, L.; Bos, J.M.; Tester, D.J.; Muir, A.; Veltmann, C.; et al. SCN5A Mutation Type and a Genetic Risk Score Associate Variably With Brugada Syndrome Phenotype in SCN5A Families. Circ. Genom. Precis. Med. 2020, 13, E002911. [Google Scholar] [CrossRef] [PubMed]
  14. Sacilotto, L.; Scanavacca, M.I.; Olivetti, N.; Lemes, C.; Pessente, G.D.; Wulkan, F.; Hachul, D.T.; Krieger, J.E.; Pereira, A.C.; Darrieux, F.C.C. Low Rate of Life-Threatening Events and Limitations in Predicting Invasive and Noninvasive Markers of Symptoms in a Cohort of Type 1 Brugada Syndrome Patients: Data and Insights From the GenBra Registry. J. Cardiovasc. Electrophysiol. 2020, 31, 2920–2928. [Google Scholar] [CrossRef] [PubMed]
  15. Di Resta, C.; Berg, J.; Villatore, A.; Maia, M.; Pili, G.; Fioravanti, F.; Tomaiuolo, R.; Sala, S.; Benedetti, S.; Peretto, G. Concealed Substrates in Brugada Syndrome: Isolated Channelopathy or Associated Cardiomyopathy? Genes 2022, 13, 1755. [Google Scholar] [CrossRef]
  16. Wilde, A.A.M.; Semsarian, C.; Márquez, M.F.; Sepehri Shamloo, A.; Ackerman, M.J.; Ashley, E.A.; Sternick Eduardo, B.; Barajas-Martinez, H.; Behr, E.R.; Bezzina, C.R.; et al. Expert Consensus Statement on the state of genetic testing for cardiac diseases. J. Arrhythmia 2022, 38, 491–553. [Google Scholar] [CrossRef] [PubMed]
  17. Hayashi, M.; Denjoy, I.; Extramiana, F.; Maltret, A.; Buisson, N.R.; Lupoglazoff, J.M.; Klug, D.; Hayashi, M.; Takatsuki, S.; Villain, E.; et al. Incidence and Risk Factors of Arrhythmic Events in Catecholaminergic Polymorphic Ventricular Tachycardia. Circulation 2009, 119, 2426–2434. [Google Scholar] [CrossRef]
  18. Medeiros-Domingo, A.; Bhuiyan, Z.A.; Tester, D.J.; Hofman, N.; Bikker, H.; van Tintelen, J.P.; Mannens, M.M.; Wilde, A.A.; Ackerman, M.J. 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, 2065–2074. [Google Scholar]
  19. Priori, S.G.; Chen, S.R. Inherited Dysfunction of Sarcoplasmic Reticulum Ca2+ Handling and Arrhythmogenesis. Circ. Res. 2011, 108, 871–883. [Google Scholar] [CrossRef]
  20. Lahat, H.; Pras, E.; Olender, T.; Avidan, N.; Ben-Asher, E.; Man, O.; Levy-Nissenbaum, E.; Khoury, A.; Lorber, A.; Goldman, B.; 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, 1378–1384. [Google Scholar] [CrossRef]
  21. Nyegaard, M.; Overgaard, M.T.; Søndergaard, M.T.; Vranas, M.; Behr, E.R.; Hildebrandt, L.L.; Lund, J.; Hedley, P.L.; Camm, A.J.; Wettrell, G.; et al. Mutations in Calmodulin Cause Ventricular Tachycardia and Sudden Cardiac Death. Am. J. Hum. Genet. 2012, 91, 703–712. [Google Scholar] [CrossRef]
  22. Roux-Buisson, N.; Cacheux, M.; Fourest-Lieuvin, A.; Fauconnier, J.; Brocard, J.; Denjoy, I.; Durand, P.; Guicheney, P.; Kyndt, F.; Leenhardt, 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, 2759–2767. [Google Scholar] [CrossRef]
  23. Asatryan, B.; Medeiros-Domingo, A. Molecular and Genetic Insights into Progressive Cardiac Conduction Disease. Europace 2019, 21, 1145–1158. [Google Scholar] [CrossRef]
  24. Medeiros-Domingo, A.; Tan, B.; Crotti, L.; Tester, D.J.; Eckhardt, L.; Cuoretti, A.; Kroboth, S.L.; Song, C.; Zhou, Q.; Kopp, D.; 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, 1466–1471. [Google Scholar] [CrossRef]
  25. Noseworthy, P.; Tikkanen, J.; Porthan, K.; Oikarinen, L.; Pietila, A.; Harald, K.; Peloso, G.M.; Merchant, F.M.; Jula, A.; Väänänen, H.; et al. The Early Repolarization Pattern in the General Population: Clinical Correlates and Heritability. J. Am. Coll. Cardiol. 2011, 57, 2284–2289. [Google Scholar] [CrossRef] [PubMed]
  26. Reinhard, W.; Kaess, B.; Debiec, R.; Nelson, C.P.; Stark, K.; Tobin, M.D.; Macfarlane, P.W.; Tomaszewski, M.; Samani, N.J.; Hengstenberg, C. Heritability of Early Repolarization: A Population-based Study. Circ. Cardiovasc. Genet. 2011, 4, 134–138. [Google Scholar] [CrossRef]
  27. Watanabe, H.; Nogami, A.; Ohkubo, K.; Kawata, H.; Hayashi, Y.; Ishikawa, T.; Makiyama, T.; Nagao, S.; Yagihara, N.; Takehara, N.; et al. Electrocardiographic Characteristics and SCN5A Mutations in Idiopathic Ventricular Fibrillation Associated with Early Repolarization. Circ. Arrhythmia Electrophysiol. 2011, 4, 874–881. [Google Scholar] [CrossRef] [PubMed]
  28. Hu, D.; Barajas-Martínez, H.; Terzic, A.; Park, S.; Pfeiffer, R.; Burashnikov, E.; Wu, Y.; Borggrefe, M.; Veltmann, C.; Schimpf, R.; et al. ABCC9 is a Novel Brugada and Early Repolarization Syndrome Susceptibility Gene. Int. J. Cardiol. 2014, 171, 431–442. [Google Scholar] [CrossRef]
  29. Takayama, K.; Ohno, S.; Ding, W.; Ashihara, T.; Fukumoto, D.; Wada, Y.; Makiyama, T.; Kise, H.; Hoshiai, M.; Matsuura, H.; et al. A de novo Gain-of-function KCND3 Mutation in Early Repolarization Syndrome. Heart Rhythm 2019, 16, 1698–1706. [Google Scholar] [CrossRef] [PubMed]
  30. Pantoni, L. Cerebral Small Vessel Disease: From Pathogenesis and Clinical Characteristics to Therapeutic Challenges. Lancet Neurol. 2010, 9, 689–701. [Google Scholar] [CrossRef]
  31. Mancuso, M.; Arnold, M.; Bersano, A.; Burlina, A.; Chabriat, H.; Debette, S.; Enzinger, C.; Federico, A.; Filla, A.; Finsterer, J.; et al. Monogenic Cerebral Smallvessel Diseases: Diagnosis and Therapy. Consensus Recommendations of the European Academy of Neurology. Eur. J. Neurol. 2020, 27, 909–927. [Google Scholar]
  32. Ullal, A.J.; Abdelfattah, R.S.; Ashley, E.A.; Froelicher, V.F. Hypertrophic Cardiomyopathy as a Cause of Sudden Cardiac Death in the Young: A Meta-analysis. Am. J. Med. 2016, 129, 486–496. [Google Scholar] [CrossRef]
  33. Veselka, J.; Anavekar, N.S.; Charron, P. Hypertrophic Obstructive Cardiomyopathy. Lancet 2017, 389, 1253–1267. [Google Scholar] [CrossRef]
  34. Abbas, M.T.; Baba Ali, N.; Farina, J.M.; Mahmoud, A.K.; Pereyra, M.; Scalia, I.G.; Kamel, M.A.; Barry, T.; Lester, S.J.; Cannan, C.R.; et al. Role of Genetics in Diagnosis and Management of Hypertrophic Cardiomyopathy: A Glimpse into the Future. Biomedicines 2024, 12, 682. [Google Scholar] [CrossRef]
  35. Bonaventura, J.; Polakova, E.; Vejtasova, V.; Veselka, J. Genetic Testing in Patients with Hypertrophic Cardiomyopathy. Int. J. Mol. Sci. 2021, 22, 10401. [Google Scholar] [CrossRef]
  36. Bagnall, R.D.; Weintraub, R.G.; Ingles, J.; Duflou, J.; Yeates, L.; Lam, L.; Davis, A.M.; Thompson, T.; Connell, V.; Wallace, J.; et al. A Prospective Study of Sudden Cardiac Death among Children and Young Adults. N. Engl. J. Med. 2016, 374, 2441–2452. [Google Scholar] [CrossRef]
  37. Miura, A.; Yamamoto, T.; Funayama, K.; Koyama, A.; Takatsuka, H.; Sato, T.; Nishio, H. Postmortem Identification of Genetic Variations Associated with Sudden Unexpected Death in Young People. Int. Heart J. 2024, 65, 55–62. [Google Scholar] [CrossRef]
  38. Hertz, C.L.; Christiansen, S.L.; Ferrero-Miliani, L.; Fordyce, S.L.; Dahl, M.; Holst, A.G.; Ottesen, G.L.; Frank-Hansen, R.; Bundgaard, H.; Morling, N. Next-generation Sequencing of 34 Genes in Sudden Unexplained Death Victims in Forensics and in Patients with Channelopathic Cardiac Diseases. Int. J. Legal Med. 2015, 129, 793–800. [Google Scholar] [CrossRef]
  39. Hertz, C.L.; Christiansen, S.L.; Ferrero-Miliani, L.; Dahl, M.; Weeke, P.E.; LuCamp, P.E.; Ottesen, G.L.; Frank-Hansen, R.; Bundgaard, H.; Morling, N. Next-generation Sequencing of 100 Candidate Genes in Young Victims of Suspected Sudden Cardiac Death with Structural Abnormalities of the Heart. Int. J. Legal Med. 2016, 130, 91–102. [Google Scholar] [CrossRef]
  40. Castiglione, V.; Modena, M.; Aimo, A.; Chiti, E.; Botto, N.; Vittorini, S.; Guidi, B.; Vergaro, G.; Barison, A.; Rossi, A.; et al. Molecular Autopsy of Sudden Cardiac Death in the Genomics Era. Diagnostics 2021, 11, 1378. [Google Scholar] [CrossRef]
  41. Richards, S.; Aziz, N.; Bale, S.; Bick, D.; Das, S.; Gastier-Foster, J.; Grody, W.W.; Hegde, M.; Lyon, E.; Spector, E.; et al. Standards and Guidelines for the Interpretation of Sequence Variants: A Joint Consensus Recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet. Med. 2015, 17, 405–424. [Google Scholar] [CrossRef]
  42. Hedley, P.L.; Jørgensen, P.; Schlamowitz, S.; Moolman-Smook, J.; Kanters, J.K.; Corfield, V.A.; Christiansen, M. The Genetic Basis of Brugada Syndrome: A Mutation Update. Hum. Mutat. 2009, 30, 1256–1266. [Google Scholar] [CrossRef] [PubMed]
  43. Bagnall, R.D.; Jipin Das, K.; Duflou, J.; Semsarian, C. Exome Analysis–Based Molecular Autopsy in Cases of Sudden Unexplained Death in the Young. Heart Rhythm 2014, 11, 655–662. [Google Scholar] [CrossRef]
  44. Hata, Y.; Kinoshita, K.; Mizumaki, K.; Yamaguchi, Y.; Hirono, K.; Ichida, F.; Takasaki, A.; Mori, H.; Nishida, N. Postmortem Genetic Analysis of Sudden Unexplained Death Syndrome under 50 Years of Age: A next-Generation Sequencing Study. Heart Rhythm 2016, 13, 1544–1551. [Google Scholar] [CrossRef] [PubMed]
  45. Modena, M.; Castiglione, V.; Aretini, P.; Mazzanti, C.M.; Chiti, E.; Giannoni, A.; Emdin, M.; Di Paolo, M. Unveiling a Sudden Unexplained Death Case by Whole Exome Sequencing and Bioinformatic Analysis. Mol. Genet. Genom. Med. 2020, 8, e1182. [Google Scholar] [CrossRef]
  46. Rizzo, S.; Lodder, E.M.; Verkerk, A.O.; Wolswinkel, R.; Beekman, L.; Pilichou, K.; Basso, C.; Remme, C.A.; Thiene, G.; Bezzina, C.R. Intercalated Disc Abnormalities, Reduced Na(+) Current Density, and Conduction Slowing in Desmoglein-2 Mutant Mice Prior to Cardiomyopathic Changes. Cardiovasc. Res. 2012, 95, 409–418. [Google Scholar] [CrossRef]
  47. Zhang, M.; Tavora, F.; Oliveira, J.B.; Li, L.; Franco, M.; Fowler, D.; Zhao, Z.; Burke, A. PKP2 Mutations in Sudden Death From Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC) and Sudden Unexpected Death with Negative Autopsy (SUDNA). Circ. J. 2012, 76, 189–194. [Google Scholar] [CrossRef]
  48. Zhang, Q.; Deng, C.; Rao, F.; Modi, R.M.; Zhu, J.; Liu, X.; Mai, L.; Tan, H.; Yu, X.; Lin, Q.; et al. Silencing of Desmoplakin Decreases Connexin 43/Nav1.5 Expression and Sodium Current in HL1 Cardiomyocytes. Mol. Med. Rep. 2013, 8, 780–786. [Google Scholar]
  49. Cerrone, M.; Lin, X.; Zhang, M.; Agullo-Pascual, E.; Pfenniger, A.; Gusky, H.C.; Novelli, V.; Kim, C.; Tirasawadichai, T.; Judge, D.P.; et al. Missense Mutations in Plakophilin-2 Cause Sodium Current Deficit and Associate with a Brugada Syndrome Phenotype. Circulation 2014, 129, 1092–1103. [Google Scholar] [CrossRef]
  50. Bezzina, C.R.; Lahrouchi, N.; Priori, S.G. Genetics of Sudden Cardiac Death. Circ. Res. 2015, 116, 1919–1936. [Google Scholar] [CrossRef]
  51. Cerrone, M.; Priori, S.G. Genetics of Sudden Death: Focus on Inherited Channelopathies. Eur. Heart J. 2011, 32, 2109–2118. [Google Scholar] [CrossRef]
  52. Tester, D.J.; Spoon, D.B.; Valdivia, H.H.; Makielski, J.C.; Ackerman, M.J. Targeted Mutational Analysis of the RyR2-encoded Cardiac Ryanodine Receptor in Sudden Unexplained Death: A Molecular Autopsy of 49 Medical Examiner/coroner’s Cases. Mayo Clin. Proc. 2004, 79, 1380–1384. [Google Scholar] [CrossRef]
  53. Chen, L.; Marquardt, L.M.; Tester, D.J.; Sampson, K.J.; Ackerman, M.J.; Kass, R.S. Mutation of an A-kinase Anchoring Protein Causes Long-QT Syndrome. Proc. Natl. Acad. Sci. USA 2007, 104, 20990–20995. [Google Scholar] [CrossRef]
  54. Maltese, E.P.; Orlova, N.; Krasikova, E.; Emelyanchik, E.; Cheremisina, A.; Kuscaeva, A.; Salmina, A.; Miotto, R.; Bonizzato, A.; Guerri, G.; et al. Gene-targeted Analysis of Clinically Diagnosed Long QT Russian Families. Int. Heart J. 2017, 58, 81–87. [Google Scholar] [CrossRef]
  55. Bottigliero, D.; Monaco, I.; Santacroce, R.; Casavecchia, G.; Correale, M.; Guastafierro, F.; Leccese, A.; Cordisco, G.; Leva, R.; Trunzo, R.; et al. Novel AKAP9 Mutation and Long QT Syndrome in a Patient with Torsade de Pointes. J. Interv. Card. Electrophysiol. 2019, 56, 171–172. [Google Scholar] [CrossRef]
  56. Tse, G.; Lee, S.; Zhou, J.; Liu, T.; Wong, C.K.I.; Mak, C.; Mok, N.S.; Jeevaratnam, K.; Zhang, Q.; Cheng, S.H.; et al. Territory-wide Chinese Cohort of Long QT Syndrome: Random Survival Forest and Cox Analyses. Front. Cardiovasc. Med. 2021, 8, 608592. [Google Scholar] [CrossRef]
  57. Allegue, C.; Coll, M.; Mates, J.; Campuzano, O.; Iglesias, A.; Sobrino, B.; Brion, M.; Amigo, J.; Carracedo, A.; Brugada, P.; et al. Genetic Analysis of Arrythmogenic Disease in the Era of NGS: The Complexity of Clinical Decision-Making in Brugada Syndrome. PLoS ONE 2015, 10, e0133037. [Google Scholar] [CrossRef]
  58. Garris, R.; Vasudev, R.; Gupta, P.; Tiyyagura, S.; Shamoon, F.; Bikkina, M. Brugada Syndrome and AKAP9: Reconciling Clinical Findings with Diagnostic Uncertainty. J. Electrocardiol. 2019, 57, 119–121. [Google Scholar] [CrossRef]
  59. Campuzano, O.; Allegue, C.; Sarquella-Brugada, G.; Coll, M.; Mates, J.; Alcalde, M.; Ferrer-Costa, C.; Iglesias, A.; Brugada, J.; Brugada, R. The Role of Clinical, Genetic and Segregation Evaluation in Sudden Infant Death. Forensic Sci. Int. 2014, 242, 9–15. [Google Scholar] [CrossRef]
  60. Neubauer, J.; Lecca, R.M.; Russo, G.; Bartsch, C.; Medeiros-Domingo, A.; Berger, W.; Haas, C. Exome Analysis in 34 Sudden Unexplained Death (SUD) Victims Mainly Identified Variants in Channelopathy-associated Genes. Int. J. Legal Med. 2018, 132, 1057–1065. [Google Scholar] [CrossRef]
  61. Li, J.L.; Wang, B.Y.; Qu, F.P.; Ma, L.; Liu, K.; Yang, L.; Nie, J.S.; Xi, M.Y.; Jia, L.P.; Tang, X.; et al. Genetic Analysis of Yunnan Sudden Unexplained Death by Whole Exome Sequencing in Southwest of China. J. Forensic Leg. Med. 2020, 70, 101896. [Google Scholar] [CrossRef]
  62. Jaouadi, H.; Bouyacoub, Y.; Chabrak, S.; Kraoua, L.; Zaroui, A.; Elouej, S.; Nagara, M.; Dallali, H.; Delague, V.; Levy, N.; et al. Multiallelic Rare Variants Support an Oligogenic Origin of Sudden Cardiac Death in the Young. Herz 2021, 46, 94–102. [Google Scholar] [CrossRef] [PubMed]
  63. Tan, H.V.; Duff, H.; Gerull, B.; Sumner, G. Early Repolarization Syndrome: A Case Report Focusing on Dynamic Electrocardiographic Changes Before Ventricular Arrhythmias and Genetic Analysis. Heart Rhythm Case Rep. 2015, 1, 213–216. [Google Scholar] [CrossRef]
  64. Qui, H.; Li, W.H.; Zhang, H.S.; Zhou, G.X.; Li, P.W. Torsades de Pointes Episode in a Woman with High-grade Fever and Inflammatory Activation: A Case Report. World J. Clin. Cases 2021, 9, 2899–2907. [Google Scholar]
  65. Forleo, C.; D’Erchia, M.A.; Sorrentino, S.; Manzari, C.; Chiara, M.; Iacoviello, M.; Guaricci, I.A.; Santis, D.D.; Musci, L.R.; La Spada, A.; et al. Targeted Next-Generation Sequencing Detects Novel Gene-phenotype Associations and Expands the Mutational Spectrum in Cardiomyopathies. PLoS ONE 2017, 12, e0181842. [Google Scholar] [CrossRef] [PubMed]
  66. Thorsen, K.; Dam, V.S.; Kjaer-Sorensen, K.; Pedersen, L.N.; Arvydas Skeberdis, V.; Jurevičius, J.; Treinys, R.; Petersen, I.M.B.S.; Nielsen, M.S.; Oxvig, C.; et al. Loss-of-activity-mutation in the Cardiac Chloride-bicarbonate Exchanger AE3 Causes Short QT Syndrome. Nat. Commun. 2017, 8, 1696. [Google Scholar] [CrossRef]
  67. Christiansen, M.K.; Kjær-Sørensen, K.; Clavsen, N.C.; Dittmann, S.; Jensen, M.F.; Guldbrandsen, H.O.; Pedersen, L.N.; Sørensen, R.H.; Lildballe, D.L.; Müller, K.; et al. Genetic Analysis Identifies the SLC4A3 Anion Exchanger as a Major Gene for Short QT Syndrome. Heart Rhythm 2023, 20, 1136–1143. [Google Scholar] [CrossRef]
  68. Bianchi, S.; Di Palma, C.; Gallus, G.N.; Taglia, I.; Poggiani, A.; Rosini, F.; Rufa, A.; Muresanu, D.F.; Cerase, A.; Dotti, M.T.; et al. Two Novel HTRA1 Mutations in a European CARASIL Patient. Neurology 2014, 82, 898–900. [Google Scholar] [CrossRef]
  69. Hara, K.; Shiga, A.; Fukutake, T.; Nozaki, H.; Miyashita, A.; Yokoseki, A.; Kawata, H.; Koyama, A.; Arima, K.; Takahashi, T.; et al. Association of HTRA1 Mutations and Familial Ischemic Cerebral Small-vessel Disease. N. Engl. J. Med. 2009, 360, 1729–1739. [Google Scholar] [CrossRef]
  70. Ito, J.; Nozaki, H.; Toyoshima, Y.; Abe, T.; Sato, A.; Hashidate, H.; Igarashi, S.; Onodera, O.; Takahashi, H.; Kakita, A. Histopathologic Features of an Autopsied Patient with Cerebral Small Vessel Disease and a Heterozygous HTRA1 Mutation. Neuropathology 2018, 38, 428–432. [Google Scholar] [CrossRef] [PubMed]
  71. Bougea, A.; Velonakis, G.; Spantideas, N.; Anagnostou, E.; Paraskevas, G.; Kapaki, E.; Kararizou, E. The First Greek Case of Heterozygous Cerebral Autosomal Recessive Arteriopathy with Subcortical Infarcts and Leukoencephalopathy: An Atypical Clinico-radiological Presentation. Neuroradiol. J. 2017, 30, 583–585. [Google Scholar] [CrossRef] [PubMed]
  72. Favaretto, S.; Margoni, M.; Salviati, L.; Pianese, L.; Manara, R.; Baracchini, C. A New Italian Family with HTRA1 Mutation Associated with Autosomal-dominant Variant of CARASIL: Are We Pointing Towards a Disease Spectrum? J. Neurol. Sci. 2019, 396, 108–111. [Google Scholar] [CrossRef] [PubMed]
  73. Zhang, J.; Zhang, X.; Nie, S.; Li, Z.; Zhang, Z.; Xiao, T. A New Chinese Family of Autosomal Dominantly inherited Cerebral Small Vessel Disease Related to A Heterozygous HTRA1 Mutation. Res. Sq. 2021. [Google Scholar] [CrossRef]
  74. Nozaki, H.; Kato, T.; Nihonmatsu, M.; Saito, Y.; Mizuta, I.; Noda, T.; Koike, R.; Miyazaki, K.; Kaito, M.; Ito, S.; et al. Distinct Molecular Mechanisms of HTRA1 Mutants in Manifesting Heterozygotes with CARASIL. Neurology 2016, 86, 1964–1974. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Sanger sequencing results confirming the novel variants in victims. Red letters are the positions of the altered amino acids.
Figure 1. Sanger sequencing results confirming the novel variants in victims. Red letters are the positions of the altered amino acids.
Diagnostics 14 01876 g001
Figure 2. Sanger sequencing results confirming the known variants in victims.
Figure 2. Sanger sequencing results confirming the known variants in victims.
Diagnostics 14 01876 g002
Table 1. Characteristics of sudden unexplained cardiac death cases.
Table 1. Characteristics of sudden unexplained cardiac death cases.
IDSex/AgeEvent at SUDMedical HistoryAutopsy Findings
Height (cm)Health
P1F/14Sleep at night160 NormalNegative
P2M/29Sleep at night178NormalNegative
P3M/25Driving/daytime158NormalNegative
P4M/19Sleep at night175NormalNegative
P5M/28Sleep at night174NormalNegative
P6M/40Eating at night152NormalNegative
P7M/39Sleep at daytime170NormalNegative
P8M/22Rest at daytime162NormalNegative
P9M/29Sleep at night168NormalNegative
P10F/33Sleep at night158NormalNegative
P11F/25Rest at daytime156NormalNegative
P12M/35Sleep at night170NormalNegative
P13M/39Sleep at night165NormalNegative
P14M/24Working/daytime180NormalNegative
P15M/23Sleep at night168NormalNegative
P16M/27Working/daytime160NormalNegative
P17M/30Working/daytime172NormalNegative
P18F/1Rest at daytime76NormalNegative
P19M/30Sleep at night169NormalNegative
P20M/40Sleep at night172NormalNegative
P21F/32Rest at daytime159NormalNegative
P22M/27Working/daytime160NormalNegative
P23M/34Sleep at night 167NormalNegative
P24M/40Sleep at night162NormalNegative
P25M/38Sleep at night170NormalNegative
P26M/40Rest at daytime162NormalNegative
P27M/35Sleep at daytime164NormalNegative
P28F/2Rest at daytime80NormalNegative
P29M/33Rest at daytime162NormalNegative
P30M/27Sleep at night173NormalNegative
P31M/35Sleep at night170NormalNegative
P32M/32Sleep at night162NormalNegative
P33M/33Sleep at night167NormalNegative
P34M/39Playing a sport166NormalNegative
P35M/24Sleep at night179NormalNegative
P36M/19Working/daytime169NormalNegative
P37F/29Rest at daytime155NormalNegative
P38F/20Sleep at night158NormalNegative
P39M/23Sleep at night168NormalNegative
P40F/40Sleep at night159NormalNegative
Table 2. Variants found in SUD victims.
Table 2. Variants found in SUD victims.
IDGenecDNA/ProteindbSNP/MAF/ClinVar/ExACZygosity
P1
(F/14)
RYR2
(NM_001035.3)
c.51C>G
p.Phe17Leu
novelhet
P2
(M/29)
AKAP9
(NM_005751.4)
c.5187_5188dup
p.Arg1730llefsTer4
novelhet
P3
(M/25)
TNNI3K
(NM_015978.3)
c.2302G>C
p.Glu768Gln
rs202238194/0.00000
RCV000768402.5/pathogenic
het
P4
(M/19)
KCNA5
(NM_002234.4)
c.683C>A
p.Pro228His
VCV002202820.2
uncertain significance
het
P5
(M/28)
MYBPC3
(NM_000256.3)
c.2275G>A
p.Glu759Lys
rs750810342/0.00002487
VCV000843772.16/uncertain significance
het
P6
(M/40)
HTRA1
(NM_002775.5)
c.496C>T
p.Arg166Cys
rs2097494390/VCV001325819.5
RCV002291765.4/pathogenic
het
P7
(M/39)
MYH6
(NM_002471.4)
c.1454A>T
p.Lys485Met
novelhet
P8
(M/22)
TNNT2
(NM_001276345.2)
c.452G>A
p.Arg151Gln
rs730881101/0.00000
RCV000796707.6/pathogenic
het
P9
(M/29)
SCN10A
(NM_006514.4)
c.2158G>A
p.Asp720Asn
rs781354273/0.00006/VCV000532067.8
pathogenic
het
P10
(F/33)
CSRP3
(NM_003476.5)
c.298C>T
p.Arg100Cys
rs201214593/0.00004/VCV000851709.9
uncertain significance
het
P11
(F/25)
MYLK
(NM_053025.4)
c.4840G>A
p.Glu1614Lys
novelhet
AKAP9
(NM_005751.4)
c.9215G>T
p.Gly3072Val
novelhet
P12
(M/35)
SCN5A
(NM_00335.5)
c.515A>G
p.His184Arg
rs794728898/0.000102/VCV000201540.5
likely pathogenic
het
P13
(M/39)
TNNI3
(NM_000363.5)
c.292C>G
p.Arg98Gly
rs730881068/0.00005/VCV001331910.2
uncertain significance
het
P14
(M/24)
GSN
(NM_198252.3)
c.872T>C
p.Ile291Thr
novelhet
P15
(M/23)
SLC4A3
(NM_005070.4)
c.2535+1G>Anovelhet
LAMA4
(NM_001105206.3)
c.5025A>T
p.Glu1675Asp
novelhet
Table 3. (A) Predictions from in silico software for variants. (B) Predictions from in silico software for the splice variant SLC4A3: c.2535+1G>A.
Table 3. (A) Predictions from in silico software for variants. (B) Predictions from in silico software for the splice variant SLC4A3: c.2535+1G>A.
(A)
Gene/Variants ClinVarCADD
Score/Prediction
FATHMM
Score/Prediction
Mutation Taster
Score/Prediction
PhD-SNP
Score/Prediction
Polyphen 2
Score/Prediction
SNP&GO
Score/Prediction
RYR2
c.10498G>T
p.Asp3500Tyr
Novel25.5
Probably damaging
−4.21
Damaging
160
Deleterious
Neutral RI81.000
Possibly damaging
Disease RI1
KCNA5
c.683C>A
p.Pro228His
Uncertain significance24.1
Probably damaging
−0.21
Tolerated
77
Deleterious
Neutral RI51.000
Possibly damaging
Disease RI7
MYBPC3
c.2275G>A
p.Glu759Lys
Uncertain significance26.7
Probably damaging
−0.20
Tolerated
56
Deleterious
Neutral RI51.000
Possibly damaging
-
MYH6
c.1454A>T
p.Lys485Met
Novel28.9
Probably damaging
−2.60
Damaging
-Disease RI61.000
Possibly damaging
Disease RI9
CSRP3
c.298C>T
p.Arg100Cys
Uncertain significance28.5
Probably damaging
−0.28
Tolerated
89
Benign
-0.110
Benign
Disease RI2
MYLK
c.4840G>A
p.Glu1614Lys
Novel36.0
Probably damaging
--Neutral RI20.611
Possibly damaging
-
AKAP9
c.9215G>T
p.Gly3072Val
Novel17.6
Tolerated
3.76
Tolerated
-Neutral RI80.973
Possibly damaging
-
TNNI3
c.292C>G
p.Arg98Gly
Uncertain significance26.3
Probably damaging
−3.49
Damaging
125
Deleterious
Neutral RI11.000
Possibly damaging
Disease RI10
GSN
c.872T>C
p.Ile291Thr
Novel29.1
Probably damaging
−0.34
Tolerated
BenignNeutral RI71.000
Possibly damaging
-
LAMA4
c.5025A>T
p.Glu1675Asp
Novel23.6
Probably damaging
−1.32
Tolerated
21
Benign
Disease RI40.998
Possibly damaging
Neutral RI7
(B)
In Silico Prediction ToolsWildtypeMutantPrediction
EX-SKIP −218.867Exon-skipping
FruitflyNANA-
MaxEntScan1.51−6.67Damage variant
NetGene20.600.00Donor loss
Spliceailookup0.980.00Donor loss
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nguyen Tat, T.; Lien, N.T.K.; Luu Sy, H.; Ta Van, T.; Dang Viet, D.; Nguyen Thi, H.; Tung, N.V.; Thanh, L.T.; Xuan, N.T.; Hoang, N.H. Identifying the Pathogenic Variants in Heart Genes in Vietnamese Sudden Unexplained Death Victims by Next-Generation Sequencing. Diagnostics 2024, 14, 1876. https://doi.org/10.3390/diagnostics14171876

AMA Style

Nguyen Tat T, Lien NTK, Luu Sy H, Ta Van T, Dang Viet D, Nguyen Thi H, Tung NV, Thanh LT, Xuan NT, Hoang NH. Identifying the Pathogenic Variants in Heart Genes in Vietnamese Sudden Unexplained Death Victims by Next-Generation Sequencing. Diagnostics. 2024; 14(17):1876. https://doi.org/10.3390/diagnostics14171876

Chicago/Turabian Style

Nguyen Tat, Tho, Nguyen Thi Kim Lien, Hung Luu Sy, To Ta Van, Duc Dang Viet, Hoa Nguyen Thi, Nguyen Van Tung, Le Tat Thanh, Nguyen Thi Xuan, and Nguyen Huy Hoang. 2024. "Identifying the Pathogenic Variants in Heart Genes in Vietnamese Sudden Unexplained Death Victims by Next-Generation Sequencing" Diagnostics 14, no. 17: 1876. https://doi.org/10.3390/diagnostics14171876

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop