Skip to main content

Analysis of the genetic contribution to thoracic aortic aneurysm or dissection in a prospective cohort of patients with familial and sporadic cases in East China

Abstract

Background

Thoracic aortic aneurysm or dissections (TAADs) represent a group of life-threatening diseases. Genetic aetiology can affect the age of onset, clinical phenotype, and timing of intervention. We conducted a prospective trial to determine the prevalence of pathogenic variants in TAAD patients and to elucidate the traits related to harbouring the pathogenic variants. One hundred and one unrelated TAAD patients underwent genetic sequencing and analysis for 23 TAAD-associated genes using a targeted PCR and next-generation sequencing-based panel.

Results

A total of 47 variants were identified in 52 TAAD patients (51.5%), including 5 pathogenic, 1 likely pathogenic and 41 variants of uncertain significance. The pathogenic or likely pathogenic (P/LP) variants in 4 disease-causing genes were carried by 1 patient with familial and 5 patients with sporadic TAAD (5.9%). In addition to harbouring one variant causing familial TAAD, the FBN1 gene harboured half of the P/LP variants causing sporadic TAAD. Individuals with an age of onset less than 50 years or normotension had a significantly increased genetic risk.

Conclusions

TAAD patients with a younger age at diagnosis or normotension were more likely to carry a P/LP variant; thus, routine genetic testing will be beneficial to a better prognosis through genetically personalized care prior to acute rupture or dissection.

Background

Thoracic aortic diseases that progress to dissection or rupture manifest a clinical disastrous course with unacceptably high morbidity and mortality [1]. Approximately 21% of patients presenting with aortic dissection die suddenly before seeking medical care [2], and 51% pass away in or en route to the facility to which they are transferred [3]. Acute surgery is the only option for in-hospital patients with type A aortic dissection; however, its overall mortality rate ranges from 8 to 24% [4]. Compared to acute treatment, thoracic aorta dissection repair has a less than 3% perioperative mortality rate and an over 85% 5-year survival rate [5] [6]. Therefore, diagnosis and treatment prior to acute rupture or dissection are likely to have better clinical outcomes.

Currently, the American and European guidelines recommend that the threshold for considering surgical intervention be a thoracic ascending aortic diameter of 5.5 cm [7, 8]. However, acute aortic events occur in more than half of patients who do not meet the criteria for prophylactic surgery, as widely reported [9] [10]. In response to this, a lower bound less than 5.0 cm was given for the timing of surgery for syndromic disorders such as Marfan syndrome (MFS), Loeys‒Dietz syndrome (LDS) and Ehlers‒Danlos syndrome (EDS) [11]. However, some individuals with nonsyndromic familial thoracic aortic aneurysm and dissections (ns-FTAADs) with manifestations restricted to the aorta are also known to have a high risk of dissection at diameters less than 5.0 cm [12, 13]. Therefore, genetic indicators might be required to choose the optimal timing for surgical repair to avoid acute aortic events.

Genetic studies over the past two decades have identified that both ns-FTAADs and syndromic thoracic aortic aneurysm or dissections (s-TAADs) are predominantly single-gene hereditary disorders inherited in an autosomal dominant manner [12]. Syndromic TAADs are caused by pathogenic variants in genes associated with the dysfunction of the extracellular matrix and TGF-β signalling and include Marfan syndrome (FBN1), Loeys‒Dietz syndrome (TGFBR1, TGFBR2, SMAD2, SMAD3, TGFB2, TGFB3), Ehlers‒Danlos syndrome (COL1A1, COL1A2, COL3A1, COL5A1 and COL5A2), arterial tortuosity syndrome (SLC2A10), Shprintzen-Goldberg syndrome (SKI) and cutis laxa (EFEMP2, ELN) [12] [14]. In comparison, the majority of ns-FTAADs arise from pathogenic variants in genes with arterial functionality. Five causative genes were identified in TAAD families: ACTA2, MYH11, MYLK and PRKG1 encode components of the contractile apparatus in vascular smooth muscle cells, and LOX encodes an extracellular matrix cross-linking enzyme [15,16,17,18]. In addition, the ns-FTAADs of some families can be explained by variants in s-TAAD causative genes, such as FBN1, TGFBR1, TGFBR2, SMAD3, and TGFB2 [11]. Genetic testing for mutations in these genes can thus contribute to the making of a definitive diagnosis, the choice of the timing of prophylactic surgery, and the optimization of clinical management.

Herein, we analysed 23 known causative genes of TAAD in a cohort of 101 individuals from East China using multiplex PCR targeted amplicon enrichment and deep-coverage massively parallel DNA sequencing, providing insight into the prevalence and clinical characteristics of causative gene carriers.

Results

Characteristics of patients

For this study, 101 consecutive patients who presented with thoracic aortic diseases, including 7 thoracic aortic aneurysms (6.8%) and 94 acute aortic syndromes (AASs; 93.1%), were enrolled; the AASs of these patients consisted of 89 aortic dissections, 2 intramural haematomas and 3 penetrating aortic ulcers (Table 1). Approximately 63.7% of patients with thoracic aortic dissections (TADs) had concurrent thoracic aortic aneurysms (TAAs). According to the Stanford classification, 60 out of 89 cases involved the ascending aorta (type A), and the remaining cases involved the descending aorta (type B). Risk factors for TAAD were investigated, including sex, blood pressure and family history (Table 1). Males composed 81.2% (82) of the cohort. Seventy-three patients (72.3%) had hypertension; 78.6% of them were males. Only one patient had a positive family history of an aortic disease, bicuspid valve or coronary artery disease.

Table 1 Baseline characteristics of the cohort (n = 101)

Variants identified by IMPATT assay

The study revealed that 79 out of 101 patients (78.2%) carried missense variants (Table 2). Fifty-six patients (55.4%) harboured missense variants with minor allele frequency (MAF) thresholds less than 0.1% worldwide (Exac) or in East Asia (Exac). Of them, 6 patients carried more than 1 variant, and 17 patients shared 4 variants (Tables S3 and S4). Thus, the total number of analysed variants in the whole cohort was 69, including 5 pathogenic variants, 1 likely pathogenic variant and 63 variants of uncertain significance (VUS) (Table 3). Sixteen of 23 targeted genes harboured alterations, of which the largest number of VUS and P/LP variants were in the FBN1 gene, and the highest overall proportion of P/LP variants per gene was also in the FBN1 gene (27.3%) (Fig. 1A).

Table 2 Summary of the variants harboured by TAAD patients
Table 3 Summary of patients with the variants
Fig. 1
figure 1

Summary of sequencing results. (A) Numbers of all variants for each gene and percentages of P/LP variants for each gene in the cohort are shown. Impact of age at diagnosis (B, C), blood pressure (D) and arotic diameter (E) on variant category in the whole cohort. Abbreviations: P/LP - pathogenic or likely pathogenic; VUS - variant of uncertain significance; NV/LB – no variant or likely benign

Six variants were considered to be P/LP: 3 variants in the FBN1 gene and 1 in each of the MYH11, MYLK, and SMAD3 genes (Table 4). Except for two in FBN1, the P/LP variants were associated with only isolated aortic disorders. Four VUS were repeatedly identified in TAAD patients: FBN1 c.1217T > A (p. Leu406His) in 8 patients (6 Type A, 2 Type B), SMAD3 c.1180T > A (p. Cys394Ser) in 4 (Type A), MYH11 c. 1729 A > C (p. Lys577Gln) in 3 (Type A), and SKI c.2189 C > T (p. Pro730Leu) in 2 (1 Type A, 1 Type B) (Table S3); in sharp contrast, these variants had never been found in healthy Chinese individuals [19]. According to protein function prediction from Provean, SIFT, PolyPhen2 and MutationTaster, all variants had a deleterious effect on protein function except for FBN1 c.1217T > A (p.Leu406His). Despite this, FBN1 c.1217T > A might be associated with TAADs if considering the median age at diagnosis of 48.5 years, which was significantly less than the 57 years in the no variant (NV) group (P = 0.0273).

Table 4 Pathogenic or likely pathogenic variants identified by the IMPATT assay

Genotype-phenotype correlation with P/LP variants

To determine which TAAD patients could benefit from genetic testing, we took the clinical characteristics of the P/LP carriers into consideration (Fig. 1B and E). Noteworthy, a P/LP variant was more likely to be identified in younger patients (Fig. 1B). The median age at diagnosis was 31.5 years for P/LP carriers, 57 years for VUS carriers (P = 0.0006) and 57 years in the NV group (P = 0.0005). A P/LP alteration was more likely to be found in the patients under the age of 50 years (5 of 32) than in those 50 years or older (1 of 69) (P = 0.0182) (Fig. 1C). In addition to the age of onset, three cardiovascular risk factors were analysed for the probability of identifying a P/PL alteration among TAAD patients. Genetic alterations contributed to a much larger percentage of TAAD cases in normotensive patients (6 of 22) than in those with hypertension (0 of 77) (P = 0.0002, Fig. 1D). However, the probability of detecting P/LP variants was not associated with normal blood lipid and glucose levels. In addition, several traits of aortic pathology were considered, including the aortic diameter, presence of an aneurysm, and dissection location. We did not find that the likelihood of carrying a P/LP variant was associated with Z scores of a aortic diameter greater than 2 (Fig. 1E), the presence of an aneurysm or Stanford Type A classification. Moreover, no significant difference was found in the percentage of P/LP variants according to the sex of the patients.

Except for the possibility of harbouring a P/LP variant based on those traits, the relative risk was also calculated as described in Table 5. Overall, our results suggested that genetic testing should be performed in TAAD patients who met either of two conditions: an age of onset less than 50 years and normotension. Additionally, when a patient is identified as carrying a pathogenic variant, genetic testing should be recommended for his or her blood relatives.

Table 5 Relative risk of carrying a pathogenic or likely pathogenic variant according to diverse phenotypes

Discussion

The present study was a one-year prospective trial aimed at determining the prevalence of genetic abnormalities in thoracic aortic disease according to the data of 101 consecutively enrolled unrelated patients from the Department of Cardiovascular Surgery. Since the cost was free, all patients in whom the IMPATT assay was recommended were able to undergo genetic testing; subsequently, the total frequency of P/LP variants among 23 known TAAD-associated genes was found to be 5.9%. This frequency is similar to the 3.9% and 4.9% reported previously, respectively, in a whole-exome sequencing study of 102 TAAD patients [20] and in a large cohort study of 1025 TAAD patients [21]. Even though we met the most stringent criteria of the ClinGen framework, including only 9 definitive causes[22], the percentage of P/LP variants was still 5.9%, close to that of the above two studies [20, 21]. A mixed cohort of sporadic and familial cases in South China had been previously reported to have a total frequency of P/LP variants of 22.5% (34/151) [23], which was far greater than the prevalence of P/LP variants in our study and the two other studies as described above [21] [20]. However, given that the P/LP variants were identified from 129 candidate genes with little evidence reported for being TAAD-causing variants in the majority, the total frequency could remain enormously overestimated in South China [22]. In addition, we repeatedly identified some VUS in TAAD patients but never in local healthy people, indicating the importance of genetic factors in thoracic aortic disease.

The FBN1 gene was the greatest contributor to TAAD cases in patients harbouring 3 out of 6 P/LP variants. Three carriers of a P/LP variant in FBN1 met the clinical criteria for a diagnosis of MFS according to the 2010 revised Ghent nosology criteria [24], of whom two displayed typical MFS manifestations involving multiple organ systems and one manifested signs isolated strictly to the ascending aorta with the dilatation of the aortic root. Notably, one of two patients with typical MFS carried a de novo variant that was not present in his parents or four siblings [25]. Consistent with our observation, FBN1 pathogenic variants have been demonstrated to be associated with a wide range of phenotypic variabilities from single organ involvement to a multiorgan dysfunction syndrome; the dysfunction of FBN1 is a common pathogenesis of aortic disease in MFS, FTAADs and sporadic TAADs[26] [27] [28]. The type and location of pathogenic FBN1 variants affect TAAD progression [27], consequently providing some insight into the prognostic stratification of TAAD cases based on the variants. Moreover, it is very likely that a variant in FBN1 arises spontaneously, sometimes without full penetrance, and involves different organs among carriers of the same variant [25] [29], indicating the difficulty of family history tracking.

As an important risk factor for TAAD [30], genetic testing is required to identify pathogenic variants in patients. Studying the clinical features of high-risk individuals harbouring a variant prone to dissection would be beneficial to improve the efficiency. Two risk factors were significantly associated with genetic disorders, including an age at diagnosis less than 50 years and normotension (Table 5). The median age of onset of P/LP carriers was much lower than that of the rest of the cohort, consistent with an observation in a large cohort [21]. Compared with the age at diagnosis in patients with familial (56.8 years) and sporadic (64.3 years) cases, the lower age of onset identified in P/LP carriers again proved that genetic defects accelerated TAAD progression[21]. Correspondingly, in this study, genetic defects were 13.1 times (95% CI: 1.6474-104.5677, P = 0.0150) more likely to be identified in the patients with an age of onset before 50 years than in those with an age of onset after 50 years.

Hypertension was strongly associated with incident thoracic aortic dissection [30]. The wall tension of the aorta is directly proportional to blood pressure, thus explaining why hypertension is a major risk factor for TAAD [31]. Hypertension accelerates the progression of the usual histopathologic changes in the aorta associated with TAAD [7]. In our study, in TAAD patients with normal circumferential stress on the aorta, the possibility of a vascular inborn error was over 44.1 times (2.579-753.565, P = 0.0089) higher than in those with hypertension.

Although the association of aortic root enlargement with aortic dissection seems straightforward, we found no significant association between variant categories and three aortic traits, including Z scores of the aortic diameter, presence of aneurysm, and dissection location. It has been proven that some pathogenic variants in genes such as MYLK [32] are not always preceded by obvious aortic dilatation. Moreover, arterial hypertension, as a predisposing condition for the development of thoracic aorta aneurysms, further makes the association more complex [7].

Our study is subject to a number of limitations. First, the number of P/LP variants might be underestimated if stringent criteria on variant classification in the guidelines of the American College of Medical Genetics and Genomics (ACMG) are considered. Several VUS carried by more than 1 patient in this study were not present in population databases, yet some of them were classified as VUS associated with ns-FTAAD in previous reports. Although extremely rare variants in causative genes are often disease related, diagnostic uncertainty is apparent solely based on the genetic technology itself, indicating the need for high-throughput functional assays to systematically classify genetic alterations [33] [34]. Moreover, our IMPATT assay specifically analysed the coding sequences and intron/exon boundaries of 23 TAAD-related genes (Table S1) but neglected noncoding variants despite their known small effects on disease; subsequently, genome-wide sequencing could enrich our understanding of TAAD-related genetic factors. Furthermore, we were unable to track the cause of death, and thus accurately track the family history of aortic disease, for the deceased parents of the patients due to limited local medical resources before 2000. Future studies will aim to correlate VUS carriers’ family histories based on further follow-up.

Conclusions

The study identified that the frequency of genetic variants causing TAAD was 5.9% among 101 affected patients in this prospective cohort. Positive carriers had nonsyndromic manifestations of TAAD except for two patients who had typical manifestations of MFS. Two clinical features, age under 50 years at diagnosis and normotension, were found to increase the likelihood of harbouring P/LP variants. If the potential for de novo variants and unknown family history are taken into consideration, the high probability of a P/LP variant among these individuals suggests that routine genetic screening is still worth recommending.

Methods

Ethical compliance

All procedures performed in this study involving human participants were in accordance with the guidelines outlined in the Declaration of Helsinki (as revised in 2013).

Patient profile

Patients who were diagnosed with TAAD by cardiovascular surgeons at the Heart Medical Centre of the First Affiliated Hospital of Gannan Medical University, Jiangxi, China, between August 2020 and August 2021 were included in this study (clinical trial registration no. ChiCTR2000034841 [clinicaltrials.gov]). Patients were included if he or she had TAAD and were aged no more than 80 years and were excluded when one of the following existed: pseudoaneurysm, heart surgery history, bone marrow transplantation, exogenous transfusion in the past six months, and pregnancy. Peripheral blood samples were collected from all participants before surgery. MFS diagnoses were made according to the 2010 revised Ghent nosology criteria [24]. Family history inquiry showed that no kinship was found in any reported individuals.

Targeted exon sequencing

The alterations in the known TAAD-associated genes were profiled using our IMPATT assay (Integrated Mutation Profiling of Actionable TAAD Targets), which utilizes multiplex PCR targeted amplicon enrichment and deep-coverage massively parallel DNA sequencing. The coordinates of 23 targeted genes in Table S1 were determined using the human reference genome (NCBI GRCh37) [35]. Custom primers for the multiplexed PCR approach were designed to enrich 754 amplicons covering all protein-coding exons plus 15 bp adjacent intron padding of the targeted genes using Illumina’s DesignStudio Sequencing Assay Designer. The total targeted DNA length was 80.86 kb. The average amplicon length was 123 bp with 48% GC content. The library was prepared using two primer pools for multiplex PCR, indexed using index adapters, further amplified, and combined in preparation for PE150 sequencing in the HiSeq XTen platform as described in the manufacturer’s AmpliSeq for Illumina workflow (Illumina, San Diego, CA).

Read mapping, variant calling & annotation

The sequencing read quality was assessed using FASTQC. Primer sequences were trimmed from FASTQ files using cutPrimers [36] prior to read mapping to the human GRCh37/hg reference sequence using the Burrows‒Wheeler Aligner V0.5.17 [37]. After the removal of potential PCR duplicates (Picard, http://picard.sourceforge.net) and realignment (GATK) [38], mutations were called and filtered using SAMtools [39], GATK3.7 [38], Pindel [40], BreakDancer [41] and CNVnator [42]. All variants were annotated according to the control population of the 1000 Genomes Project (2014 October release) [43], ExAC r0.3.1 [44], EVS (https://evs.gs.washington.edu/EVS/), the disease databases of ClinVar [45], and OMIM [46].

Variant and sample filtering

Synonymous variants and intronic variants outside splice sites were not considered for downstream analysis only when they were annotated as pathogenic or disease-causing in ClinVar [45] or HMGD [47]. Variants with MAF thresholds greater than 0.1% in either the worldwide superpopulation or the East Asian population ExAC r0.3.1 [44] datasets were excluded. Samples with less than 80% of target bases covered by more than 60 reads were excluded from downstream analysis.

Pathogenicity assignment

Variants passing filters were assigned to one of four categories, ‘pathogenic’, ‘likely pathogenic’, ‘VUS’ or ‘likely benign’, based on ACMG criteria [48].

In particular, a variant was considered a pathogenic variant if it (I) had been reported as disease causing in HGMD or pathogenic in ClinVar, (II) resulted in the same amino acid substitution as an HGMD disease-causing variant or ClinVar pathogenic variant, (III) was a de novo variant if nonsense (i.e., had an in-frame and out-of-frame deletion/insertion, the splice site variants affected the canonical splice sequence, or was shown to alter splicing on an mRNA level), (IV) was missense and either affected/created cysteine residues or affected conserved residues of the EGF consensus sequence in FBN1 [49], (V) had a substitution of a glycine residue within a GlyXY repeat in collagen triple helical domain, (VI) had an insertion of amino acids disrupting the GlyXY repeat sequence in collagens, or (VII) had an alteration of a key residue in a protein feature in keeping with previously ascribed molecular mechanisms for a given gene [21]. The pathogenic or likely pathogenic (P/LP) variants and four VUS were confirmed by Sanger sequencing (Table S2).

Statistical analysis

Fisher’s exact test was used for the assessment of categorical variables between different groups. Nonparametric phenotypic continuous measurements were analysed using the unpaired Wilcoxon rank-sum test. P values less than 0.05 were considered statistically significant (two-sided). All calculations were performed using SPSS 24.0 software.

Data Availability

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

AAS:

acute aortic syndromes

EDS:

Ehlers-Danlos Syndrome

ns-FTAAD:

non-syndromic familial thoracic aortic aneurysm and dissection

MFS:

Marfan Syndrome

LDS:

Loeys-Dietz Syndrome

NV:

no variation

P/LP:

pathogenic or likely pathogenic

s-TAAD:

syndromic thoracic aortic aneurysm or dissection

TAA:

thoracic aortic aneurysm

TAAD:

thoracic aortic aneurysm or dissection

VUS:

variant of uncertain significance

References

  1. Nienaber CA, Clough RE. Management of acute aortic dissection. Lancet. 2015;385(9970):800–11.

    PubMed  Google Scholar 

  2. Saeyeldin AA, Velasquez CA, Mahmood SUB, Brownstein AJ, Zafar MA, Ziganshin BA, Elefteriades JA. Thoracic aortic aneurysm: unlocking the “silent killer” secrets. Gen Thorac Cardiovasc Surg. 2019;67(1):1–11.

    PubMed  Google Scholar 

  3. Tanaka Y, Sakata K, Sakurai Y, Yoshimuta T, Morishita Y, Nara S, Takahashi I, Hirokami M, Yamagishi M. Prevalence of type a Acute Aortic dissection in patients with Out-Of-Hospital cardiopulmonary arrest. Am J Cardiol. 2016;117(11):1826–30.

    PubMed  Google Scholar 

  4. Gregory SH, Yalamuri SM, Bishawi M, Swaminathan M. The Perioperative Management of ascending aortic dissection. Anesth Analg. 2018;127(6):1302–13.

    PubMed  Google Scholar 

  5. Goodney PP, Travis L, Lucas FL, Fillinger MF, Goodman DC, Cronenwett JL, Stone DH. Survival after open versus endovascular thoracic aortic aneurysm repair in an observational study of the Medicare population. Circulation. 2011;124(24):2661–9.

    PubMed  PubMed Central  Google Scholar 

  6. Hellgren T, Wanhainen A, Steuer J, Mani K. Outcome of endovascular repair for intact and ruptured thoracic aortic aneurysms. J Vasc Surg. 2017;66(1):21–8.

    PubMed  Google Scholar 

  7. Erbel R, Aboyans V, Boileau C, Bossone E, Bartolomeo RD, Eggebrecht H, Evangelista A, Falk V, Frank H, Gaemperli O, et al. 2014 ESC Guidelines on the diagnosis and treatment of aortic diseases: document covering acute and chronic aortic diseases of the thoracic and abdominal aorta of the adult. The Task Force for the diagnosis and treatment of aortic Diseases of the European Society of Cardiology (ESC). Eur Heart J. 2014;35(41):2873–926.

    PubMed  Google Scholar 

  8. Hiratzka LF, Bakris GL, Beckman JA, Bersin RM, Carr VF, Casey DE Jr, Eagle KA, Hermann LK, Isselbacher EM, Kazerooni EA et al. 2010 ACCF/AHA/AATS/ACR/ASA/SCA/SCAI/SIR/STS/SVM guidelines for the diagnosis and management of patients with thoracic aortic disease: executive summary. A report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines, American Association for Thoracic Surgery, American College of Radiology, American Stroke Association, Society of Cardiovascular Anesthesiologists, Society for Cardiovascular Angiography and Interventions, Society of Interventional Radiology, Society of Thoracic Surgeons, and Society for Vascular Medicine. Catheter Cardiovasc Interv 2010, 76(2):E43-86.

  9. Pape LA, Tsai TT, Isselbacher EM, Oh JK, O’Gara PT, Evangelista A, Fattori R, Meinhardt G, Trimarchi S, Bossone E, et al. Aortic diameter > or = 5.5 cm is not a good predictor of type A aortic dissection: observations from the International Registry of Acute Aortic dissection (IRAD). Circulation. 2007;116(10):1120–7.

    PubMed  Google Scholar 

  10. Chau KH, Elefteriades JA. Natural history of thoracic aortic aneurysms: size matters, plus moving beyond size. Prog Cardiovasc Dis. 2013;56(1):74–80.

    PubMed  Google Scholar 

  11. Milewicz DM, Regalado ES. Use of genetics for personalized management of heritable thoracic aortic disease: how do we get there? J Thorac Cardiovasc Surg. 2015;149(2 Suppl):3–5.

    Google Scholar 

  12. Ostberg NP, Zafar MA, Ziganshin BA, Elefteriades JA. The Genetics of thoracic aortic aneurysms and dissection: a clinical perspective. Biomolecules 2020, 10(2):182.

  13. Andelfinger G, Loeys B, Dietz H. A decade of Discovery in the genetic understanding of thoracic aortic disease. Can J Cardiol. 2016;32(1):13–25.

    PubMed  Google Scholar 

  14. Meester JAN, De Kinderen P, Verstraeten A, Loeys B. Meester-Loeys Syndrome. 2021.

  15. Guo DC, Pannu H, Tran-Fadulu V, Papke CL, Yu RK, Avidan N, Bourgeois S, Estrera AL, Safi HJ, Sparks E, et al. Mutations in smooth muscle alpha-actin (ACTA2) lead to thoracic aortic aneurysms and dissections. Nat Genet. 2007;39(12):1488–93.

    CAS  PubMed  Google Scholar 

  16. Wang L, Guo DC, Cao J, Gong L, Kamm KE, Regalado E, Li L, Shete S, He WQ, Zhu MS, et al. Mutations in myosin light chain kinase cause familial aortic dissections. Am J Hum Genet. 2010;87(5):701–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Guo DC, Regalado E, Casteel DE, Santos-Cortez RL, Gong L, Kim JJ, Dyack S, Horne SG, Chang G, Jondeau G, et al. Recurrent gain-of-function mutation in PRKG1 causes thoracic aortic aneurysms and acute aortic dissections. Am J Hum Genet. 2013;93(2):398–404.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Lee VS, Halabi CM, Hoffman EP, Carmichael N, Leshchiner I, Lian CG, Bierhals AJ, Vuzman D, Brigham Genomic M, Mecham RP, et al. Loss of function mutation in LOX causes thoracic aortic aneurysm and dissection in humans. Proc Natl Acad Sci U S A. 2016;113(31):8759–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Cao Y, Li L, Xu M, Feng Z, Sun X, Lu J, Xu Y, Du P, Wang T, Hu R, et al. The ChinaMAP analytics of deep whole genome sequences in 10,588 individuals. Cell Res. 2020;30(9):717–31.

    PubMed  PubMed Central  Google Scholar 

  20. Ziganshin BA, Bailey AE, Coons C, Dykas D, Charilaou P, Tanriverdi LH, Liu L, Tranquilli M, Bale AE, Elefteriades JA. Routine genetic testing for thoracic aortic aneurysm and dissection in a clinical setting. Ann Thorac Surg. 2015;100(5):1604–11.

    PubMed  Google Scholar 

  21. Weerakkody R, Ross D, Parry DA, Ziganshin B, Vandrovcova J, Gampawar P, Abdullah A, Biggs J, Dumfarth J, Ibrahim Y, et al. Targeted genetic analysis in a large cohort of familial and sporadic cases of aneurysm or dissection of the thoracic aorta. Genet Med. 2018;20(11):1414–22.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Renard M, Francis C, Ghosh R, Scott AF, Witmer PD, Ades LC, Andelfinger GU, Arnaud P, Boileau C, Callewaert BL, et al. Clinical validity of genes for heritable thoracic aortic aneurysm and dissection. J Am Coll Cardiol. 2018;72(6):605–15.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Li Y, Fang M, Yang J, Yu C, Kuang J, Sun T, Fan R. Analysis of the contribution of 129 candidate genes to thoracic aortic aneurysm or dissection of a mixed cohort of sporadic and familial cases in South China. Am J Transl Res. 2021;13(5):4281–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Loeys BL, Dietz HC, Braverman AC, Callewaert BL, De Backer J, Devereux RB, Hilhorst-Hofstee Y, Jondeau G, Faivre L, Milewicz DM, et al. The revised Ghent nosology for the Marfan syndrome. J Med Genet. 2010;47(7):476–85.

    CAS  PubMed  Google Scholar 

  25. Duan Y, Li P, Ding T, Wang Y, Liao Y, Du Z, Ling J, Liu S, Li W, Liu Z. Combining clinical examination with exome sequencing for the diagnosis and treatment of Marfan syndrome: a case series of 6 families from China. Ann Palliat Med. 2021;10(9):9953–62.

    PubMed  Google Scholar 

  26. Tan L, Li Z, Zhou C, Cao Y, Zhang L, Li X, Cianflone K, Wang Y, Wang DW. FBN1 mutations largely contribute to sporadic non-syndromic aortic dissection. Hum Mol Genet. 2017;26(24):4814–22.

    CAS  PubMed  Google Scholar 

  27. Takeda N, Inuzuka R, Maemura S, Morita H, Nawata K, Fujita D, Taniguchi Y, Yamauchi H, Yagi H, Kato M, et al. Impact of pathogenic FBN1 variant types on the progression of aortic disease in patients with Marfan Syndrome. Circ Genom Precis Med. 2018;11(6):e002058.

    CAS  PubMed  Google Scholar 

  28. Regalado ES, Guo DC, Santos-Cortez RL, Hostetler E, Bensend TA, Pannu H, Estrera A, Safi H, Mitchell AL, Evans JP, et al. Pathogenic FBN1 variants in familial thoracic aortic aneurysms and dissections. Clin Genet. 2016;89(6):719–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Buoni S, Zannolli R, Macucci F, Ansaldi S, Grasso M, Arbustini E, Fois A. The FBN1 (R2726W) mutation is not fully penetrant. Ann Hum Genet. 2004;68(Pt 6):633–8.

    CAS  PubMed  Google Scholar 

  30. Landenhed M, Engstrom G, Gottsater A, Caulfield MP, Hedblad B, Newton-Cheh C, Melander O, Smith JG. Risk profiles for aortic dissection and ruptured or surgically treated aneurysms: a prospective cohort study. J Am Heart Assoc. 2015;4(1):e001513.

    PubMed  PubMed Central  Google Scholar 

  31. Mule G, Nardi E, Morreale M, Castiglia A, Geraci G, Altieri D, Cacciatore V, Schillaci M, Vaccaro F, Cottone S. The relationship between aortic Root size and hypertension: an Unsolved Conundrum. Adv Exp Med Biol. 2017;956:427–45.

    PubMed  Google Scholar 

  32. Hannuksela M, Stattin EL, Klar J, Ameur A, Johansson B, Sorensen K, Carlberg B. A novel variant in MYLK causes thoracic aortic dissections: genotypic and phenotypic description. BMC Med Genet. 2016;17(1):61.

    PubMed  PubMed Central  Google Scholar 

  33. Majithia AR, Tsuda B, Agostini M, Gnanapradeepan K, Rice R, Peloso G, Patel KA, Zhang X, Broekema MF, Patterson N, et al. Prospective functional classification of all possible missense variants in PPARG. Nat Genet. 2016;48(12):1570–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Fraile-Bethencourt E, Diez-Gomez B, Velasquez-Zapata V, Acedo A, Sanz DJ, Velasco EA. Functional classification of DNA variants by hybrid minigenes: identification of 30 spliceogenic variants of BRCA2 exons 17 and 18. PLoS Genet. 2017;13(3):e1006691.

    PubMed  PubMed Central  Google Scholar 

  35. Flicek P, Amode MR, Barrell D, Beal K, Brent S, Carvalho-Silva D, Clapham P, Coates G, Fairley S, Fitzgerald S, et al. Ensembl 2012. Nucleic Acids Res. 2012;40(Database issue):D84–90.

    CAS  PubMed  Google Scholar 

  36. Kechin A, Boyarskikh U, Kel A, Filipenko M. cutPrimers: a New Tool for Accurate cutting of primers from reads of targeted next generation sequencing. J Comput Biol. 2017;24(11):1138–43.

    CAS  PubMed  Google Scholar 

  37. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25(14):1754–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, Garimella K, Altshuler D, Gabriel S, Daly M, et al. The genome analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20(9):1297–303.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R. Genome Project Data Processing S: the sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25(16):2078–9.

    PubMed  PubMed Central  Google Scholar 

  40. Ye K, Guo L, Yang X, Lamijer EW, Raine K, Ning Z. Split-Read Indel and structural variant calling using PINDEL. Methods Mol Biol. 2018;1833:95–105.

    CAS  PubMed  Google Scholar 

  41. Fan X, Abbott TE, Larson D, Chen K. BreakDancer: identification of genomic structural variation from paired-end read Mapping. Curr Protoc Bioinformatics. 2014;45:151611–11.

    Google Scholar 

  42. Abyzov A, Urban AE, Snyder M, Gerstein M. CNVnator: an approach to discover, genotype, and characterize typical and atypical CNVs from family and population genome sequencing. Genome Res. 2011;21(6):974–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Genomes Project C, Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, Korbel JO, Marchini JL, McCarthy S, McVean GA, et al. A global reference for human genetic variation. Nature. 2015;526(7571):68–74.

    Google Scholar 

  44. Karczewski KJ, Weisburd B, Thomas B, Solomonson M, Ruderfer DM, Kavanagh D, Hamamsy T, Lek M, Samocha KE, Cummings BB, et al. The ExAC browser: displaying reference data information from over 60 000 exomes. Nucleic Acids Res. 2017;45(D1):D840–5.

    CAS  PubMed  Google Scholar 

  45. Landrum MJ, Lee JM, Benson M, Brown G, Chao C, Chitipiralla S, Gu B, Hart J, Hoffman D, Hoover J, et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 2016;44(D1):D862–868.

    CAS  PubMed  Google Scholar 

  46. Amberger JS, Bocchini CA, Schiettecatte F, Scott AF, Hamosh A. OMIM.org: online mendelian inheritance in man (OMIM(R)), an online catalog of human genes and genetic disorders. Nucleic Acids Res. 2015;43(Database issue):D789–798.

    PubMed  Google Scholar 

  47. Stenson PD, Mort M, Ball EV, Shaw K, Phillips A, Cooper DN. The human gene mutation database: building a comprehensive mutation repository for clinical and molecular genetics, diagnostic testing and personalized genomic medicine. Hum Genet. 2014;133(1):1–9.

    CAS  PubMed  Google Scholar 

  48. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, 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(5):405–24.

    PubMed  PubMed Central  Google Scholar 

  49. Baudhuin LM, Kotzer KE, Lagerstedt SA. Increased frequency of FBN1 truncating and splicing variants in Marfan syndrome patients with aortic events. Genet Med. 2015;17(3):177–87.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We gratefully thank all participants for their supports to this research.

Funding

The work was financially supported by the Department of Science and Technology of Jiangxi Province (No. 20213BCJ22001), the National Natural Science Foundation of China (No. 82260332; 81960326), the Science and Technology Project of Jiangxi Health Committee (No. 202130676), and the Innovation Team of Gannan Medical University (No. TD201902).

Author information

Authors and Affiliations

Authors

Contributions

ZYL and YYD developed the study concept and the design. All patients were interviewed and examined by JXX, ZHL, YMZ, CNT, ZMD, ZFL, JJY, WTL, and WCX. Data and sample collection was conducted by YBW, TD, XHZ, MMP, YQ, XML, THC, PJL, KL, MG and YQH. YYD wrote the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Ziyou Liu.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the ethics committee at the First Affiliated Hospital of Gannan Medical University (No. 201713104150) and informed consent was taken from all the patients.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

13023_2023_2855_MOESM1_ESM.xlsx

Supplementary Material 1. Additional file 1. Table S1. List of 23 targeted genes for multiplex PCR-targeted amplicon enrichment. Additional file 2. Table S2. Primer sequences for Sanger validation of 9 pathogenic or likely pathogenic and 4 shared genetic variants. Additional file 3. Table S3. Four variants of uncertain significance were shared in 17 patients. Additional file 4. Table S4. Six patients had more than 1 variant of uncertain significance

Supplementary Material 2

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Duan, Y., Xiong, J., Lai, Z. et al. Analysis of the genetic contribution to thoracic aortic aneurysm or dissection in a prospective cohort of patients with familial and sporadic cases in East China. Orphanet J Rare Dis 18, 251 (2023). https://doi.org/10.1186/s13023-023-02855-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13023-023-02855-7

Keywords