Skip to main content
Erschienen in: BMC Pediatrics 1/2023

Open Access 01.12.2023 | Research

Case–control association study of congenital heart disease from a tertiary paediatric cardiac centre from North India

verfasst von: Prachi Kukshal, Radha O Joshi, Ajay Kumar, Shadab Ahamad, Prabhatha Rashmi Murthy, Yogesh Sathe, Krishna Manohar, Soma Guhathakurta, Subramanian Chellappan

Erschienen in: BMC Pediatrics | Ausgabe 1/2023

Abstract

Background

Congenital Heart diseases (CHDs) account for 1/3rd of all congenital birth defects. Etiopathogenesis of CHDs remain elusive despite extensive investigations globally. Phenotypic heterogeneity witnessed in this developmental disorder reiterate gene-environment interactions with periconceptional factors as risk conferring; and genetic analysis of both sporadic and familial forms of CHD suggest its multigenic basis. Significant association of de novo and inherited variants have been observed. Approximately 1/5th of CHDs are documented in the ethnically distinct Indian population but genetic insights have been very limited. This pilot case–control based association study was undertaken to investigate the status of Caucasian SNPs in a north Indian cohort.

Method

A total of 306 CHD cases sub-classified into n = 198 acyanotic and n = 108 cyanotic types were recruited from a dedicated tertiary paediatric cardiac centre in Palwal, Haryana. 23 SNPs primarily prioritized from Genome-wide association studies (GWAS) on Caucasians were genotyped using Agena MassARRAY Technology and test of association was performed with adequately numbered controls.

Results

Fifty percent of the studied SNPs were substantially associated in either allelic, genotypic or sub-phenotype categories validating their strong correlation with disease manifestation. Of note, strongest allelic association was observed for rs73118372 in CRELD1 (p < 0.0001) on Chr3, rs28711516 in MYH6 (p = 0.00083) and rs735712 in MYH7 (p = 0.0009) both on Chr 14 and were also significantly associated with acyanotic, and cyanotic categories separately. rs28711516 (p = 0.003) and rs735712 (p = 0.002) also showed genotypic association. Strongest association was observed with rs735712(p = 0.003) in VSD and maximum association was observed for ASD sub-phenotypes.

Conclusions

Caucasian findings were partly replicated in the north Indian population. The findings suggest the contribution of genetic, environmental and sociodemographic factors, warranting continued investigations in this study population.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12887-023-04095-x.
Krishna Manohar is deceased.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ACTC1
Actin alpha cardiac muscle
ASD
Atrial septal defect
ATXN2-AS, BRAP
Ataxin antisense RNA, BRCA 1(Breast cancer gene1) associated protein
AVSD
Atrioventricular septal defect
CHARGE
Coloboma, Heart defects, Atresia choanae, growth Retardation, Genital abnormalities, and Ear abnormalities
CHD
Congenital heart defect
CHD7
Chromodomain helicase DNA binding protein 7
Chr.
Chromosome number
CNV
Copy number variation
CRELD1
Cysteine rich with EGF like domains 1
DCRV
Double chamber right ventricle
ELN
Elastin
ENSA
Endosulfine alpha
GAS1
Growth arrest-specific 1
GATA4
GATA binding protein 4
GOSR2
Golgi SNAP receptor complex member 2
GWAS
Genome-wide association studies
HLHS
Hypoplastic left heart syndrome
ISLET1
ISL1 transcription factor LIM/homeodomain
JAG1
Jagged canonical Notch ligand 1
LINC02252
Long intergenic non-protein coding RNA 2252
GJD2
Gap junction protein delta 2
LINC02676
Long intergenic non-protein coding RNA 2676
MEF2C
Myocyte enhancer factor 2c
MLC
Myosin light chains
MYH6
Myosin heavy chain 6
MYH7
Myosin heavy chain 7
MYH7B
Myosin heavy chain 7B
NKX2.5
Nk2 homeobox 5
NRP1
Neuropilin 1
OSR1
Odd-skipped-related transcription factor 1
PCSK6
Proprotein convertase subtilisin/kexin type 6
PKD1L2
Polycystin 1 like 2
PS
Pulmonary stenosis
PTPN11
Tyrosine-protein phosphatase non-receptor type 11
SH2B3
SH2B (Src homology 2B) adaptor protein 3
SNP
Single nucleotide polymorphism
SV
Single ventricle
SYNPR-AS1, SYNPR
Synaptoporin antisense RNA 1
TAPVC
Total anomalous pulmonary venous return
TBX
T-box transcription factors
TGA
Transposition of the great arteries
UCP2
Uncoupling protein 1; VSD: Ventricular septal defect

Background

Congenital Heart Defect (CHD) is common structural abnormality occurring at the time of foetal development. Limited information is available on the exact mechanism of CHD pathogenesis. It affects 9 in 1000 live births globally [1]. This corresponds to 17% of the world CHD load from India [2], yet meagre genetic information available for the disease in the country. In early gestation, incidence is even higher as certain CHDs are complex and have been shown to result in foetal demise [3]. Septational abnormalities account for half of the cardiac congenital defects ranging from nonpathological to lethal [4]. Cardiac development is a complex process and requires intricate coordination of several molecular events for eventual normal structure and function of the heart. Any error/s in these steps result in pathogenic remodelling of heart [5]. Chromosomal aneuploidies like Trisomy 21, 18 and 13 are commonly associated with CHDs [6, 7]. Though 80% CHDs are sporadic in origin [8], some familial cases of Atrial Septal Defect (ASD), Ventricular Septal Defect (VSD) and Hypoplastic Left Heart Syndrome (HLHS) are recorded but the inheritance patterns are complex [9, 10]. The recurrence risk in off springs of CHD patients varies from 3- 20% depending on the lesion, with slightly higher recurrence in females [11]. Almost one half of the siblings with recurrent lesions in a family have a different lesion suggesting multifactorial etiology and illusive molecular mechanisms [8, 9, 12].
Technological advances now enable study of these developmental defects, thus closing the gap of knowledge between the morphology and genetics [6]. Novel techniques identify regions in the genome on which transcription factors act, driving their target genes and provide new knowledge on CHD development. The T-box transcription factors (TBX5) gene is reported to interact with Nk2 Homeobox 5 (NKX2.5) and GATA binding protein 4 (GATA4) both transcriptional activator of Natriuretic peptide B, which positively regulates the developing heart. The involvement of several well-established cardiac transcription factors that are expressed in cardiogenic plates such as NKX2.5, GATA4, TBX5, TBX20, Myosin heavy chain 6 (MYH6), Actin alpha cardiac muscle (ACTC1) and Myocyte enhancer factor 2C (MEF2C) have been extensively studied in both human and animal experiments [13-17]. Role of Mutations in transcription factors have been studied for non-syndromic CHDs [18].
Point mutations of cardiac transcription factor genes, single nucleotide polymorphism (SNPs), aneuploidy, and chromosomal copy number variants (CNVs) are directly associated with CHDs. Association of single SNPs seldom lead to complex disease manifestation [19]. There are substantial genetic predispositions to inherited as well as de novo variants with variable effect sizes towards disease risk. Since mutations are rare it requires large numbers to be screened, also same mutations may not be present in all samples. Conventionally a multifactorial inheritance model has been proposed for CHD involving a multitude of susceptibility genes, with low-penetrant common variants or intermediate-penetrant rare variants, superposed on unfavourable environmental factors as causal [15, 20, 21]. Several ethnic or racial differences may also be observed [22]. It is important to investigate genotype–phenotype correlation to provide leads with an opportunity to predict the prognosis.
Limited genetic diagnosis is available for many of the CHDs. Therefore several commercial ventures to sequence genes have been undertaken [23, 24]. These ventures discovered several de novo mutations in the known as well as new genes [6, 12]. Genome wide association studies (GWAS) involve the comparison of genetic variants (known as well as unknown), which can be used to detect genetic risk factors of big and small effect to CHD manifestation [12, 25, 26]. So far more than 500 genes have been estimated with a potential role in the development of CHD [27]. Genetic studies on transcription factors like GATA4, NKX2.5, TBX1and TBX20 have previously shown to identify new mutations in Indian population. [28-33]. Till date limited candidate gene studies have been done in India [34] and only one using exome sequencing [35].
Next generation sequencing (NGS) technologies meet a high standard of evidence and also afford correct predictions in novel datasets. In this study we select association findings from GWAS on CHDs and evaluate in a north Indian cohort. Screening for SNPs and not mutations may reflect better in an association study. Therefore, we chose to assess association of common variants primarily from previous GWAS or meta-analysis studies [26, 36-39] in Caucasians and tested their replicability in our adequately powered study samples.

Methodology

Study samples

Cases

The study was approved by Institutional Ethics Committee (IEC) at Sri Sathya Sai Sanjeevani Research Foundation (SSSSRF), Palwal, India. Samples were recruited from the Sai Sanjeevani biobank for Congenital Heart of SSSSRF from the period of September 2018 to September 2021. n = 306 CHD cases who underwent surgery or cath interventions at Sri Sathya Sai Sanjeevani International Centre for Child Heart Care & Research, Palwal, Haryana were recruited for this study. All methods were performed in accordance with the relevant guidelines and regulations laid down by Indian Council of Medical Research(ICMR). Samples from clinically identified known syndromes or showing distinct extracardiac features were excluded from the study. All the samples selected for the study were non-syndromic based on medical examination and the clinical phenotypes were segregated into cyanotic and acyanotic types. n = 108 cyanotic and n = 198 acyanotic CHD cases were included. Categorization of cases into 10 subphenotypes (ASD; VSD; TOF: Tetralogy of Fallot; VSD + PS:VSD + Pulmonary Stenosis; DCRV: Double Chamber Right Ventricle; TGA: Transposition of the Great Arteries; SV: Single Ventricle; AVSD: Atrioventricular Septal Defect; TAPVC: both Total and Partial Anomalous Pulmonary Venous Connection; Miscellaneous cases) was accomplished based on intervention procedure, ECHO findings and patient history. Sample distributions are given in Supplementary Table 1.

Controls groups

Age matched control group of north India origin was an ideal control for this study but was unavailable and is a limitation of the study. Therefore, we compared it to adequately numbered adults of north Indian origin available in public database [40] (http://​asia.​ensembl.​org/​Homo_​sapiens/​Variation) and published literature [41, 42]. The three groups described below were combined to get a substantial number of controls [43].
Control group1: n = 48 adults of north Indian origin unaffected for CHD based on clinical evaluation as well as an ECHO for confirmation were included in the study. Control group2: Since the inhouse control group was small, n = 489 open source 1000 Genomes South Asian data was also used as part of reference controls. Control group3: Comparable genotypes for 8 common markers from north India controls in a published study on Celiac Disease [41, 42] was available for n = 1170 adults.

Marker selection

Assuming large hypothesis-free studies would reflect true associations with disease, most of the GWAS associations were tested in this study. In the absence of contemporary Indian data, associations from Caucasians who are closer to north Indians in ancestry [44], were tested for their contribution. Top 50 markers associated in GWAS catalogue for CHD phenotypes or through maternal influence were shortlisted but only 35 could be accommodated in the assay pool. SNPs having p > 10–6 and a Minor Allele Frequency(MAF) > 0.001 were selected for inclusion. Priority was given to polymorphic SNPs having a functional implication on the gene product. The study samples had 80% power to detect associations for the SNPs having MAF > 0.1 and an Odds Ratio > 1.5.

Experimental method

Samples were isolated by the conventional phenol–chloroform phase separation method. The coded samples were blinded and genotyped through a commercial facility (Genes2Me: https://​www.​genes2me.​com) using Agena MassARRAY technology, which is based on matrix-assisted laser desorption/ionization—time of flight (MALDI-TOF) mass spectrometry. Genetic polymorphisms are distinguished by analysis of their individual mass, excluding the need for fluorescence or labelling. Control samples, duplicates, negatives and positives were used for quality control. Out of 35 SNPs, two failed in assay design and ten SNPs (rs6763159; rs11894932; rs365990; rs17189763; rs2010963; rs350916; rs436582; rs4366490; rs8061121; rs870142) gave ambiguous reads on QC and were removed from the analysis. Only 23 markers and samples which had > 80% genotype calls were retained in the study. A chi-square test was used for association measures and Fisher’s exact test was used, if the expected number was less than five. Statistical tools like SPSS version 21.0 and free online tools [45] (https://​vassarstats.​net/​) were used for analysis. Power was calculated using Quanto [46] (http://​biostats.​usc.​edu/​Quanto.​html).

Results

A total of n = 23 SNPs in Hardy Weinberg Equilibrium (stringent cut off: p > 0.001) were included in the analysis (Supplementary Table 2). Since two SNPs rs2046060 and rs12165908 were monomorphic hence they could not be utilised further for association study. No significant allelic association was seen among cyanotic and acyanotic cases groups. These were still analysed separately vs all controls.
Markers with allelic and genotypic associations are tabulated and presented in Table 1 and 2 for acyanotic, cyanotic and combined categories.
Table 1
Allelic association for all analysed SNPs
  
SNP details
All Controls Vs Acynanotic
All Controls Vs Cynotic
All Controls Vs Combined cases
Mapped gene
SNP
A1
A2
F_A
F_U
ChiSq; p
ChiSq; p
ChiSq; p
ENSA
rs12045807
C
T
0.10
0.06
0.81;0.37
0.35;0.55
0.13;0.72
CRELD1
rs73118372
C
T
0.01
0
10.89;0.001
5.71;0.02
15.7; < 0.0001
SYNPR-AS1, SYNPR
rs1975649
T
C
0.35
0.46
0.58;0.45
0.13;0.72
0.65;0.42
INTERGENIC
rs185531658
C
T
0.005
0
0.02*
1*
0.04*
ELN
rs2071307
A
G
0.25
0.24
1.4;0.24
0.75;0.39
0.19;0.66
CHD7
rs3763592
T
C
0.05
0.10
0.11;0.74
0.49;0.48
0.03;0.86
LINC02676
rs2388896
A
G
0.28
0.29
1.27;0.26
2.37;0.12
3.14;0.08
NRP1
rs2228638
T
C
0.09
0.07
0.09;0.76
1.22;0.27
0.77;0.38
UCP2
rs659366
T
C
0.39
0.44
5.74;0.02
0.01;0.92
3.57;0.06
ATXN2-AS, BRAP
rs11065987
G
A
0.05
0.02
1.18;0.27
0.02;0.89
0.77;0.38
SH2B3, PTPN11
rs11066320
A
G
0.05
0.02
0.38;0.54
0.44;0.51
0.66;0.42
INTERGENIC
rs1497062
A
T
0.33
0.34
0.17;0.68
1.23;0.27
0.09;0.76
MYH6
rs28711516
T
C
0.04
0.01
4.65;0.03
9.22;0.002
11.18;0.0008
MYH7
rs735712
A
G
0.05
0.06
7.29;0.01
4.98;0.03
10.98;0.0009
LINC02252, GJD2
rs6495706
C
G
0.05
0.06
1.64;0.2
0.38;0.54
1.67;0.2
PCSK6
rs3784481
G
A
0.49
0.5
0.07;0.79
0.89;0.35
0.5;0.48
INTERGENIC
rs6499100
C
T
0.446
0.489
0.19;0.66
0.05;0.82
0.22;0.64
PKD1L2
rs55788414
T
C
0.058
0.035
1.49;0.22
0.19;0.66
1.32;0.25
GOSR2
rs11874
A
G
0.02
0
4.31;0.04
0.48;0.49
3.81;0.05
JAG1
rs35761929
C
G
0.153
0.257
0.19;0.66
0.38;0.54
0;1
MYH7B
rs3746446
C
T
0.248
0.128
0.17;0.68
0.01;0.92
0.06;0.81
A1 Allele 1, A2 Allele 2, F_A Frequency of A1 in cases, F_U Frequency of A1 in controls, ChiSq Pearson’s correlation, p Significance
*Fishers test p values (in cell counts less than five), All Significant ChiSquare p values in bold font
Table 2
Genotypic Association for all analysed SNPs
  
Control**
Cases**
All Controls Vs Acyanotic cases
All Controls Vs Cyanotic cases
All Controls Vs Combined cases
Mapped gene
SNP
Control group 1
Control group 2
Control group 3#
All Control
Acyanotic
Cyanotic
Total cases
ChiSq:p
ChiSq:p
ChiSq:p
ENSA
rs12045807
0/5/35
4/83/402
19/212/930
23/300/1367
6/31/146
1/17/88
7/48/234
4.01;0.13
0.35;0.84
2.02;0.36
CRELD1 *
rs73118372
0/0/46
5/53/431
-
5/53/477
0/4/150
0/3/89
0/7/239
-
-
-
SYNPR-AS1, SYNPR
rs1975649
3/29/6
49/217/223
126/510/516
178/756/745
12/101/65
11/42/40
23/143/105
9.41;0.01
0.16;0.92
5.74;0.06
INTERGENIC *
rs185531658
0/0/45
0/0/489
-
0/0/534
0/3/180
0/0/103
0/3/283
-
-
-
ELN
rs2071307
1/15/19
19/186/284
64/436/643
84/637/946
10/74/90
3/36/59
13/110/149
1.62;0.44
0.98;0.61
0.49;0.78
CHD7 *
rs3763592
0/9/35
1/43/445
-
1/52/480
0/16/157
0/13/90
0/29/247
-
-
-
LINC02676
rs2388896
4/16/21
52/208/229
115/496/528
171/720/778
9/71/75
8/32/51
17/103/126
3.16;0.21
3.09;0.21
3.47;0.18
NRP1
rs2228638
0/7/41
10/109/370
15/186/955
25/302/1366
1/34/147
0/17/89
1/51/236
1.08;0.58
-
2.44;0.29
UCP2
rs659366
3/26/7
62/239/188
122/518/504
187/783/699
25/90/55
8/56/39
33/146/94
6.24;0.04
2.56;0.28
5.46;0.07
ATXN2-AS, BRAP
rs11065987
0/2/45
4/51/434
-
4/53/479
1/14/174
1/10/99
2/24/273
1.14;0.57
0.09;0.95
0.82;0.66
SH2B3, PTPN11
rs11066320
0/2/45
5/54/430
-
5/56/475
2/16/171
1/9/100
3/25/271
0.63;0.73
0.52;0.77
0.96;0.62
INTERGENIC
rs1497062
4/19/17
59/215/215
-
63/234/232
27/63/76
9/42/49
36/105/125
3.12;0.21
1.22;0.54
1.7;0.43
MYH6
rs28711516
0/1/46
3/79/407
-
3/80/453
1/16/174
0/5/105
1/21/279
5.28;0.07
-
11.77;0.003
MYH7
rs735712
0/5/37
4/97/388
-
4/102/425
2/15/155
0/10/86
2/25/241
10.44;0.01
-
13.03;0.002
LINC02252, GJD2
rs6495706
0/6/41
2/65/422
-
2/71/463
0/20/175
2/9/100
2/29/275
-
5.14;0.08
2.92;0.23
PCSK6
rs3784481
11/18/11
127/238/124
230/575/332
368/831/467
42/75/50
25/48/24
67/123/74
1.59;0.45
0.92;0.63
1.59;0.45
INTERGENIC
rs6499100
9/27/10
86/247/156
204/588/350
299/862/516
35/91/54
21/52/33
56/143/87
0.29;0.87
0.43;0.81
0.52;0.77
PKD1L2
rs55788414
0/3/40
1/73/415
-
1/76/455
1/18/164
1/12/95
2/30/259
2.94;0.23
2.28; 0.32
3.86;0.15
GOSR2 *
rs11874
0/0/46
1/38/450
-
1/38/496
0/6/185
0/6/102
0/12/287
-
-
-
JAG1
rs35761929
1/16/18
16/110/363
-
17/126/381
5/43/115
1/24/71
6/67/186
0.37;0.83
1.4;0.5
0.76;0.68
MYH7B
rs3746446
0/11/32
33/193/263
-
33/204/295
7/71/97
4/46/55
11/117/152
1.3;0.52
1.68;0.43
2.35;0.31
*Association could not be calculated because of very low counts
**genotype counts of variant homozygous / heterozygous/wildtype homozygous for each category are denoted
#only 8 markers data available; All associated ChiSq: p values in bold font
Allelic Association: rs73118372 on chr. 3, rs28711516 and rs735712 on chr. 14 exhibited association for all three categories (acyanotic, cyanotic and combined categories; Table 1). rs73118372 in CRELD12 = 15.7; p < 0.0001); rs28711516 in MYH62 = 11.18; p = 0.00083) and rs735712 in MYH72 = 10.98; p = 0.0009) showed strong allelic associations. Variant rs11874 in GOSR2 had nominal association (χ2 = 3.81; p = 0.051) and rs185531658 an intergenic SNP on Chr 5 [39] showed association on Fisher’s test as allele counts were low (p = 0.043) while rs659366 in UCP2 and rs2388896 intergenic SNP demonstrated a weak trend of association (Table 1).
All associations were mainly driven by similar association in the larger acyanotic group. For the SNP rs659366 the association strengthen (χ2 = 5.74; p = 0.017) in the acyanotic group. The cyanotic association was established only for rs73118372, rs28711516 and rs735712.

Genotypic association

Similar trend was observed for genotypic associations as illustrated in Table 2. rs1975649(χ2 = 5.74; p = 0.057) of SYNPR on Chr 3; rs28711516 (χ2 = 11.75; p = 0.0028) and rs735712 (χ2 = 13.03; p = 0.0015) on Chr 14 showed genotypic association. rs659366 demonstrated a trend on association (χ2 = 5.46; p = 0.065).

Associations with CHD subtypes

Seven allelic associations with ASD; three with TOF; two with VSD + PS; one each with TGA and AVSD phenotypes were observed. rs735712 of MYH7 (p = 0.0029) showed strongest association with VSD subtype amongst all associations observed. ASD, VSD and TOF categories were adequately numbered for association analysis. All subtypes except ASD, VSD and TOF were combined for analysis and showed association for rs73118372, rs28711516 and rs735712 (Table 3).
Table 3
Allelic Association of CHD subtypes for all analysed SNPs
  
ASD
VSD
TOF
VSD + PS
DCRV
TGA
SV
AVSD
TAPVC
MISC
ALL EXCEPT ASD, VSD & TOF
Mapped gene
SNP
ChiSq;p
ChiSq;p
ChiSq;p
ChiSq;p
ChiSq;p
ChiSq;p
ChiSq;p
ChiSq;p
ChiSq;p
ChiSq;p
ChiSq;p
ENSA
rs12045807
2.27;0.13
0.01;0.92
0.59;0.44
0.36;0.55
0.27;0.60
0.03;0.86
0.28;0.59
2.78;0.09
0.13;0.72
0.58;0.45
0.10;0.75
CRELD1
rs73118372
0.005*
0.07*
6.14;0.01
3.07;0.08
1*
0.62*
1*
0.86;0.35
0.39*
0.11*
6.98;0.008
SYNPR-AS1, SYNPR
rs1975649
6.99;0.01
0.09;0.76
0.19;0.66
0.01;0.92
0.51;0.47
0.04;0.84
0.03;0.87
0.27;0.60
2.55;0.11
0.26;0.61
0.17;0.67
INTERGENIC
rs185531658
1*
1*
0.03*
1*
1*
1*
1*
1*
1*
0.05*
0.20*
ELN
rs2071307
0.19;0.66
0.02;0.88
0.96;0.33
0.95;0.33
0.35*
0.19;0.66
0.25;0.62
7.22;0.007
0.11;0.74
0.09;0.76
0.15;0.69
CHD7
rs3763592
2.77;0.09
0.42;0.52
0.26;0.61
3.11;0.08
1*
0.46*
1*
1*
0.63*
0.13;0.72
0.05;0.81
LINC02676
rs2388896
0.08;0.77
0.28;0.59
2.88;0.09
1.09;0.29
0.63;0.43
0.76;0.38
0.53;0.47
1.25;0.26
0.21;0.65
0.93;0.34
2.18;0.14
NRP1
rs2228638
0.15;0.69
0.03;0.86
1.55;0.21
0.43;0.51
3.37;0.07
0.003;0.95
0.08;0.78
0.39*
0.32;0.57
0.17;0.68
0.19;0.66
UCP2
rs659366
3.85;0.05
3.17;0.07
1.1;0.29
0.48;0.49
0.005;0.95
0.08;0.78
0.06;0.81
3.45;0.06
2.72;0.09
1.28;0.26
0.83;0.36
ATXN2-AS, BRAP
rs11065987
1.54;0.21
2.10;0.15
0.36;0.55
0.23;0.63
1*
0.05;0.82
0.01;0.92
0.67;0.41
0.39*
0.68;0.41
0.08;0.77
SH2B3, PTPN11
rs11066320
1.94;0.16
1.18;0.27
0.69;0.41
0.01;0.92
1*
0.097;0.76
0.0002;0.99
0.49;0.48
0.24;0.63
0.008;0.93
0.18;0.66
INTERGENIC
rs1497062
0.02;0.88
0.29;0.59
0.57;0.45
1.4;0.24
0.001;0.97
1.36;0.24
0.09;0.77
1.10;0.29
0.85;0.35
1.02;0.31
3.56;0.06
MYH6
rs28711516
5.09;0.02
0.4;0.53
6.48;0.01
7.35;0.007
0.60;0.44
0.03;0.86
0.63*
0.10;0.75
0.16*
0.72;0.39
9.29;0.002
MYH7
rs735712
0.8;0.37
0.003*
2.96;0.09
4.34;0.04
1*
0.79;0.37
0.15;0.69
0.38*
0.04;0.85
0.72;0.39
6.48;0.011
LINC02252, GJD2
rs6495706
0.02;0.88
1.04;0.30
3.64;0.06
1.21;0.27
1*
0.15;0.71
0.62*
0.12;0.73
2.18;0.14
0.43;0.51
0.75;0.38
PCSK6
rs3784481
0.02;0.88
0.51;0.47
0.02;0.88
0.22;0.64
1.26;0.26
2.66;0.10
0.54;0.46
0.06;0.81
0.09;0.76
2.34;0.13
1.08;0.3005
INTERGENIC
rs6499100
0.53;0.46
1.13;0.29
0.1;0.75
0.51;0.48
1.76;0.19
2.15;0.14
0.98;0.32
0.27;0.60
1.71;0.19
0.94;0.33
0.38;0.54
PKD1L2
rs55788414
2.52;0.11
2.93;0.09
0.13;0.72
0.73;0.39
1*
4.51;0.03
0.61;0.43
0.39*
0.0014;0.97
0.46;0.49
0.30;0.58
GOSR2
rs11874
0.04*
0.09;0.76
2.59;0.11
0.69;0.41
1*
0.04;0.84
1*
1*
0.0008;0.98
0.69;0.40
1.72;0.19
JAG1
rs35761929
4.01;0.05
0.97;0.32
0.04;0.84
0.31;0.58
0.009;0.92
0.15 8*
0.09;0.76
0.001;0.97
0.04;0.83
0.02;0.89
0.04;0.84
MYH7B
rs3746446
6.69;0.009
0.13;0.72
0.07;0.79
0.14;0.71
1.90;0.17
0.05;0.82
0.0009;0.98
1.12;0.29
0.002;0.96
0.01;0.92
0.06;0.80
*Fishers test p values (in cell counts less than five), Misc.: Other Miscellaneous types; All associated ChiSq: p values in bold font

Discussion

50% of tested SNPs were substantially associated in either allelic, genotypic or sub-phenotypes of north Indian CHD cohort validating their strong correlation with disease manifestation. Burden of CHD is overall heavy in India [2] and is prominent in north India. Several genetic determinants of this complex developmental disorder have been reported based on conventional candidate genes and contemporary GWAS but mostly in Caucasian populations. However it is very poorly investigated in the ethnically distinct Indian population. This study was an attempt to test the association of Caucasian findings prior to performing a hypothesis-free approach in the study cohort. Of the 23 SNPs which were successfully genotyped in the modest sized study cohort, 11 SNPs showing allelic or genotypic or association with the CHD sub-phenotypes (Table 1, 23) in a trans ethnic population was noteworthy and reiterates the functional relevance of the associated genes/pathways in CHD pathogenesis.
Of the seven associations observed, four SNPs namely rs73118372 (missense variant) Chr 3; rs659366 (promoter region) on Chr 11; rs735712 (synonymous variant) and rs28711516 (missense variant) on Chr 14; are of functional relevance. Strongest allelic association was observed for rs73118372, rs28711516 and rs735712.
The variant rs73118372(c.1136 T > C) in Exon 9 of CRELD1 is associated with Downs syndrome [47]. CRELD1 is involved in the formation of atrioventricular cushion [48] and disrupts existing exon splicing, thus altering the protein configuration and making it unstable. It was associated for all cyanotic, acyanotic and combined categories, and with ASD and TOF sub-phenotypes. Intronic SNP rs1975649 (SYNPR; Intron2) also on Chr3 exhibited strong association both in allelic and genotypic categories and was also associated with ASD. Cardiac myosin is the molecular motor that powers heart contraction, a property essential for heart function. It also plays a pivotal role in muscle regulation, development, and mechanotransduction [49]. The α-MYHC (MYH6) is expressed in atrial muscle and the β-MYHC (MYH7) in skeletal slow-twitch muscle and have arisen through a tandem gene duplication event [50] on Chr. 14. The duplication event is not evident in the genomes of other vertebrates (e.g. birds, fish, amphibia) [51]. MYH6, MYH7 and MYH7B are associated with R amplitude [52]. Heterozygous pathogenic variants in MHY7 have been associated with septal defects or Ebstein anomaly [48] and of MYH6 with HLHS and cardiac conduction [53, 54]. MYH6 is associated with non-syndromic coarctation of the aorta [38, 50] and also presented in families of Shone complex [54]. Previous reports on MYH6 rs28711516 (c.166G > A; p.G56R) associations with atrial fibrillation [53] and sporadic dilated cardiomyopathy [55] and a GWAS study from south India [35] warrant further investigation of this gene. MYH7 mutations have been reported for Indian families [56, 57]. Exon 12 variant rs735712 (c.1062C > T; p.G354G) has previously been reported in dilated cardiomyopathies in Indian population [58]. Previous linkage study using microarray identified rs1055061in HOMEZ, a ubiquitously expressed transcription factor on the same locus, in 83 consanguineous CHD families from India [59]. In present study, synonymous variant rs3746446 in MYH7B was associated only with ASD phenotype. This SNP showed strong association with congenital cardiovascular left-sided lesions [38]along with rs12045807, a SNP not associated in our study.
Allelic and genotypic association for combined as well as the acyanotic group for promoter variant rs659366 in UCP2, having a role in reactive oxygen species (ROS) pathway [60, 61] is already been reported to be associated with maternal diabetes in CHD offsprings [62] and with dietary factors in Asian populations [60]. It was also associated with ASD subtype. rs185531658, a SNP with the strongest association in 4034 patients of CHD [39], was nominally associated both with our combined data and in acyanotic group.
ELN rs2071307 showed mild association with AVSD and PKD1L2 rs55788414 with TGA subtype. GOSR2 rs11874, a promoter SNP, was detected in patients with anomalies of thoracic arteries and veins, and may affect the expression of GOSR2 [39]. It showed a marginal association in our population for combined and acyanotic categories. Missense variant rs35761929 in JAG1, involved in Notch cell signalling was associated with ASD subtype in present study and was previously found in ten exome sequenced families from India [38]. During development, the notch pathway regulates embryonic cells destiny to be part of the heart, liver, eyes, ears, and spinal column. The Jagged-1 protein continues to play a role throughout life in the development of new blood cells. These four markers reported are too small in number to make a conclusive statement on its role based on the present results and warrant replication in larger sample set.
Findings in the study were predominantly from the coding region and a few from the intergenic region. The CRELD1, MYH6 and MYH7 interact with each other during development of the myosin filament, an active and essential component of the heart tissue. Only one of the strongly associated common variants per gene was tested in this study. There may be more rare and common variants associated from these genes and additional association studies are essential to estimate polygenic risk score to make a significant genotype- phenotype risk prediction for CHD.

Conclusion

This is a first study testing association of Caucasian GWAS SNPs in a north Indian population. All the SNPs studied have been previously shown to have a strong role in the development of CHD. 11 out of 21 SNPs were associated in the study cohort and highlighted the role of CRELD1, MYH6 and MYH7 in non-syndromic CHD. Strong association of markers from Chr3 and Chr14, and with ASD, VSD and TOF sub-phenotypes were notable in this study and warrant replication in independent CHD cohorts of north Indian origin. This may help uncover the mechanism of disease manifestation by a complex landscape of events influenced by variations in genetic, environmental and demographic patterns.

Acknowledgements

The authors acknowledge the contribution of Cardiology and Surgery teams of Sri Sathya Sai Health and Education Trust for sharing the clinical findings and Sri Sathya Sai Sanjeevani Biobank for providing samples of the study respectively. The Authors thank ILS and Genes2Me for genotyping the samples. Authors acknowledge Dr. Thelma for sharing genotype information of samples used as part of controls in the study and Dr. Upasana Bhattacharya for technical suggestions.

Declarations

Ethics approval from Institutional Ethics Committee (IEC), Sri Sathya Sai Sanjeevani Research Foundation (SSSSRF) registered with National Ethics Committee Registry for Biomedical and Health Research, Department of Health Research (File no.: EC/NEW/INST/2022/2673) was granted under the number PSR00007/1/IEC/10/2019. Written informed consent was obtained for all participants and parents/ guardians and assent was provided by children above 8 years of age as per IEC, SSSSRF and ICMR guidelines. Involvement in the study was voluntary and there were no repercussions for non-participation. This study was performed in accordance with the declaration of Helsinki.
Not Applicable.

Competing interests

The Authors have no potential competing interests to declare.
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.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Hoffman JI, Kaplan S. The incidence of congenital heart disease. J Am Coll Cardiol. 2002;39(12):1890–900. Hoffman JI, Kaplan S. The incidence of congenital heart disease. J Am Coll Cardiol. 2002;39(12):1890–900.
2.
Zurück zum Zitat Saxena A. Congenital Heart Disease in India: A Status Report. Indian Pediatr. 2018;55:1075–82.PubMedCrossRef Saxena A. Congenital Heart Disease in India: A Status Report. Indian Pediatr. 2018;55:1075–82.PubMedCrossRef
3.
Zurück zum Zitat Bravo-valenzuela NJ, Peixoto AB, Araujo JE. Prenatal diagnosis of congenital heart disease: A review of current knowledge. Indian Heart J. 2018;70:150–64.PubMedCrossRef Bravo-valenzuela NJ, Peixoto AB, Araujo JE. Prenatal diagnosis of congenital heart disease: A review of current knowledge. Indian Heart J. 2018;70:150–64.PubMedCrossRef
4.
Zurück zum Zitat Buijtendijk MFJ, Barnett P, van den Hoff MJB. Development of the human heart. Am J Med Genet Part C Semin Med Genet. 2020;184:7–22.PubMedCrossRef Buijtendijk MFJ, Barnett P, van den Hoff MJB. Development of the human heart. Am J Med Genet Part C Semin Med Genet. 2020;184:7–22.PubMedCrossRef
5.
Zurück zum Zitat Cohn JN, Ferrari R, Sharpe N. Cardiac remodeling-concepts and clinical implications: A consensus paper from an International Forum on Cardiac Remodeling. J Am Coll Cardiol. 2000;35:569–82.PubMedCrossRef Cohn JN, Ferrari R, Sharpe N. Cardiac remodeling-concepts and clinical implications: A consensus paper from an International Forum on Cardiac Remodeling. J Am Coll Cardiol. 2000;35:569–82.PubMedCrossRef
6.
Zurück zum Zitat Pierpont ME, Brueckner M, Chung WK, Garg V, Lacro RV, McGuire AL, et al. Genetic Basis for Congenital Heart Disease: Revisited: A Scientific Statement from the American Heart Association. Circulation. 2018;138:e653-711.PubMedCentralCrossRef Pierpont ME, Brueckner M, Chung WK, Garg V, Lacro RV, McGuire AL, et al. Genetic Basis for Congenital Heart Disease: Revisited: A Scientific Statement from the American Heart Association. Circulation. 2018;138:e653-711.PubMedCentralCrossRef
7.
Zurück zum Zitat Santoro SL, Steffensen EH. Congenital heart disease in Down syndrome – A review of temporal changes. J Congenit Cardiol. 2021;5:1–14.CrossRef Santoro SL, Steffensen EH. Congenital heart disease in Down syndrome – A review of temporal changes. J Congenit Cardiol. 2021;5:1–14.CrossRef
8.
Zurück zum Zitat Nees SN, Chung WK. Genetic basis of human congenital heart disease. Cold Spring Harb Perspect Biol. 2020;12(9):a036749.CrossRef Nees SN, Chung WK. Genetic basis of human congenital heart disease. Cold Spring Harb Perspect Biol. 2020;12(9):a036749.CrossRef
9.
Zurück zum Zitat Øyen N, Poulsen G, Boyd HA, Wohlfahrt J, Jensen PKA, Melbye M. Recurrence of congenital heart defects in families. Circulation. 2009;120:295–301.PubMedCrossRef Øyen N, Poulsen G, Boyd HA, Wohlfahrt J, Jensen PKA, Melbye M. Recurrence of congenital heart defects in families. Circulation. 2009;120:295–301.PubMedCrossRef
10.
Zurück zum Zitat Ellesøe SG, Workman CT, Bouvagnet P, Loffredo CA, McBride KL, Hinton RB, et al. Familial co-occurrence of congenital heart defects follows distinct patterns. Eur Heart J. 2018;39:1015–22.PubMedCrossRef Ellesøe SG, Workman CT, Bouvagnet P, Loffredo CA, McBride KL, Hinton RB, et al. Familial co-occurrence of congenital heart defects follows distinct patterns. Eur Heart J. 2018;39:1015–22.PubMedCrossRef
11.
Zurück zum Zitat Burn J, Brennan P, Little J, Holloway S, Coffey R, Somerville J, et al. Recurrence risks in offspring of adults with major heart defects: results from first cohort of British collaborative study. Lancet. 1998;351:311–6.PubMedCrossRef Burn J, Brennan P, Little J, Holloway S, Coffey R, Somerville J, et al. Recurrence risks in offspring of adults with major heart defects: results from first cohort of British collaborative study. Lancet. 1998;351:311–6.PubMedCrossRef
13.
Zurück zum Zitat Andersen TA, Troelsen KDLL, Larsen LA. Of mice and men: Molecular genetics of congenital heart disease. Cell Mol Life Sci. 2014;71:1327–52.PubMedCrossRef Andersen TA, Troelsen KDLL, Larsen LA. Of mice and men: Molecular genetics of congenital heart disease. Cell Mol Life Sci. 2014;71:1327–52.PubMedCrossRef
14.
Zurück zum Zitat Bruneau BG. Signaling and transcriptional networks in heart development and regeneration. Cold Spring Harb Perspect Biol. 2013;5(3):a008292. Bruneau BG. Signaling and transcriptional networks in heart development and regeneration. Cold Spring Harb Perspect Biol. 2013;5(3):a008292.
15.
Zurück zum Zitat Wessels MW, Willems PJ. Genetic factors in non-syndromic congenital heart malformations. Clin Genet. 2010;78:103–23.PubMedCrossRef Wessels MW, Willems PJ. Genetic factors in non-syndromic congenital heart malformations. Clin Genet. 2010;78:103–23.PubMedCrossRef
16.
Zurück zum Zitat Xu H, Baldini A. Genetic pathways to mammalian heart development: Recent progress from manipulation of the mouse genome. Semin Cell Dev Biol. 2007;18(1):77–83. Xu H, Baldini A. Genetic pathways to mammalian heart development: Recent progress from manipulation of the mouse genome. Semin Cell Dev Biol. 2007;18(1):77–83.
17.
Zurück zum Zitat Suluba E, Shuwei L, Xia Q, Mwanga A. Congenital heart diseases: genetics, non-inherited risk factors, and signaling pathways. Egypt J Med Hum Genet. 2020;21:1–12.CrossRef Suluba E, Shuwei L, Xia Q, Mwanga A. Congenital heart diseases: genetics, non-inherited risk factors, and signaling pathways. Egypt J Med Hum Genet. 2020;21:1–12.CrossRef
18.
Zurück zum Zitat Kodo K, Nishizawa T, Furutani M, Arai S, Ishihara K, Oda M, et al. Genetic analysis of essential cardiac transcription factors in 256 patients with non-syndromic congenital heart defects. Circ J. 2012;76:1703–11.PubMedCrossRef Kodo K, Nishizawa T, Furutani M, Arai S, Ishihara K, Oda M, et al. Genetic analysis of essential cardiac transcription factors in 256 patients with non-syndromic congenital heart defects. Circ J. 2012;76:1703–11.PubMedCrossRef
19.
Zurück zum Zitat Cerrone M, Remme CA, Tadros R, Bezzina CR, Delmar M. Beyond the one gene-one disease paradigm complex genetics and pleiotropy in inheritable cardiac disorders. Circulation. 2019;140:595–610.PubMedPubMedCentralCrossRef Cerrone M, Remme CA, Tadros R, Bezzina CR, Delmar M. Beyond the one gene-one disease paradigm complex genetics and pleiotropy in inheritable cardiac disorders. Circulation. 2019;140:595–610.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Zhang TN, Wu QJ, Liu YS, Lv J Le, Sun H, Chang Q, et al. Environmental risk factors and congenital heart disease: an umbrella review of 165 Systematic reviews and meta-analyses with more than 120 million participants. Front Cardiovasc Med. 2021;8:640729. Zhang TN, Wu QJ, Liu YS, Lv J Le, Sun H, Chang Q, et al. Environmental risk factors and congenital heart disease: an umbrella review of 165 Systematic reviews and meta-analyses with more than 120 million participants. Front Cardiovasc Med. 2021;8:640729.
21.
Zurück zum Zitat Spendlove SJ, Bondhus L, Lluri G, Sul JH, Arboleda VA. Polygenic risk scores of endo-phenotypes identify the effect of genetic background in congenital heart disease. Hum Genet Genomics Adv. 2022;3: 100112.CrossRef Spendlove SJ, Bondhus L, Lluri G, Sul JH, Arboleda VA. Polygenic risk scores of endo-phenotypes identify the effect of genetic background in congenital heart disease. Hum Genet Genomics Adv. 2022;3: 100112.CrossRef
22.
Zurück zum Zitat Knowles RL, Ridout D, Crowe S, Bull C, Wray J, Tregay J, et al. Ethnic and socioeconomic variation in incidence of congenital heart defects. Arch Dis Child. 2017;102:496–502.PubMedCrossRef Knowles RL, Ridout D, Crowe S, Bull C, Wray J, Tregay J, et al. Ethnic and socioeconomic variation in incidence of congenital heart defects. Arch Dis Child. 2017;102:496–502.PubMedCrossRef
23.
Zurück zum Zitat Jerves T, Beaton A, Kruszka P. The genetic workup for structural congenital heart disease. Am J Med Genet Part C Semin Med Genet. 2020;184:178–86.PubMedCrossRef Jerves T, Beaton A, Kruszka P. The genetic workup for structural congenital heart disease. Am J Med Genet Part C Semin Med Genet. 2020;184:178–86.PubMedCrossRef
24.
Zurück zum Zitat Paige SL, Saha P, Priest JR. Beyond Gene Panels: Whole Exome Sequencing for Diagnosis of Congenital Heart Disease. Circ Genom Precis Med. 2018;11:e002097.PubMedPubMedCentralCrossRef Paige SL, Saha P, Priest JR. Beyond Gene Panels: Whole Exome Sequencing for Diagnosis of Congenital Heart Disease. Circ Genom Precis Med. 2018;11:e002097.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Blue GM, Kirk EP, Giannoulatou E, Sholler GF, Dunwoodie SL, Harvey RP, et al. Advances in the Genetics of Congenital Heart Disease: A Clinician’s Guide. J Am Coll Cardiol. 2017;69:859–70.PubMedCrossRef Blue GM, Kirk EP, Giannoulatou E, Sholler GF, Dunwoodie SL, Harvey RP, et al. Advances in the Genetics of Congenital Heart Disease: A Clinician’s Guide. J Am Coll Cardiol. 2017;69:859–70.PubMedCrossRef
26.
Zurück zum Zitat Agopian AJ, Goldmuntz E, Hakonarson H, Sewda A, Taylor D, Mitchell LE. Genome-Wide Association Studies and Meta-Analyses for Congenital Heart Defects. Circ Cardiovasc Genet. 2017;10:1–9.CrossRef Agopian AJ, Goldmuntz E, Hakonarson H, Sewda A, Taylor D, Mitchell LE. Genome-Wide Association Studies and Meta-Analyses for Congenital Heart Defects. Circ Cardiovasc Genet. 2017;10:1–9.CrossRef
27.
Zurück zum Zitat Jia Y, Louw JJ, Breckpot J, Callewaert B, Barrea C, Sznajer Y, et al. The diagnostic value of next generation sequencing in familial nonsyndromic congenital heart defects. Am J Med Genet Part A. 2015;167:1822–29.CrossRef Jia Y, Louw JJ, Breckpot J, Callewaert B, Barrea C, Sznajer Y, et al. The diagnostic value of next generation sequencing in familial nonsyndromic congenital heart defects. Am J Med Genet Part A. 2015;167:1822–29.CrossRef
28.
Zurück zum Zitat Dixit R, Kumar A, Mohapatra B. Implication of GATA4 synonymous variants in congenital heart disease: A comprehensive in-silico approach. Mutat Res Mol Mech Mutagen. 2019;813:31–8.CrossRef Dixit R, Kumar A, Mohapatra B. Implication of GATA4 synonymous variants in congenital heart disease: A comprehensive in-silico approach. Mutat Res Mol Mech Mutagen. 2019;813:31–8.CrossRef
29.
Zurück zum Zitat Dixit R, Narasimhan C, Balekundri VI, Agrawal D, Kumar A, Mohapatra B. Functional analysis of novel genetic variants of NKX2-5 associated with nonsyndromic congenital heart disease. Am J Med Genet Part A. 2021;185:3644–63.PubMedCrossRef Dixit R, Narasimhan C, Balekundri VI, Agrawal D, Kumar A, Mohapatra B. Functional analysis of novel genetic variants of NKX2-5 associated with nonsyndromic congenital heart disease. Am J Med Genet Part A. 2021;185:3644–63.PubMedCrossRef
30.
Zurück zum Zitat Bose D, Vaigundan D, Shetty M, Krishnappa J, Kutty AVM. Identification of intronic-splice site mutations in GATA4 gene in Indian patients with congenital heart disease. Mutat Res - Fundam Mol Mech Mutagen. 2017;803–8050:26–34. Bose D, Vaigundan D, Shetty M, Krishnappa J, Kutty AVM. Identification of intronic-splice site mutations in GATA4 gene in Indian patients with congenital heart disease. Mutat Res - Fundam Mol Mech Mutagen. 2017;803–8050:26–34.
31.
Zurück zum Zitat Mattapally S, Singh M, Murthy KS, Asthana S, Banerjee SK. Computational modeling suggests impaired interactions between NKX2.5 and GATA4 in individuals carrying a novel pathogenic D16N NKX2.5 mutation. Oncotarget. 2018;9(17):13713–32.PubMedPubMedCentralCrossRef Mattapally S, Singh M, Murthy KS, Asthana S, Banerjee SK. Computational modeling suggests impaired interactions between NKX2.5 and GATA4 in individuals carrying a novel pathogenic D16N NKX2.5 mutation. Oncotarget. 2018;9(17):13713–32.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Dinesh SM, Lingaiah K, Savitha MR, Krishnamurthy B, Narayanappa D, Ramachandra NB. GATA4 specific nonsynonymous single-nucleotide polymorphisms in congenital heart disease patients of Mysore. India Genet Test Mol Biomarkers. 2011;15:715–20.PubMedCrossRef Dinesh SM, Lingaiah K, Savitha MR, Krishnamurthy B, Narayanappa D, Ramachandra NB. GATA4 specific nonsynonymous single-nucleotide polymorphisms in congenital heart disease patients of Mysore. India Genet Test Mol Biomarkers. 2011;15:715–20.PubMedCrossRef
33.
Zurück zum Zitat Kusuma L, Dinesh SM, Savitha MR, Krishnamurthy B, Narayanappa D, Ramachandra NB. Mutations of TFAP2B in congenital heart disease patients in Mysore, south India. Indian J Med Res. 2011;134:621–26.CrossRef Kusuma L, Dinesh SM, Savitha MR, Krishnamurthy B, Narayanappa D, Ramachandra NB. Mutations of TFAP2B in congenital heart disease patients in Mysore, south India. Indian J Med Res. 2011;134:621–26.CrossRef
34.
Zurück zum Zitat Raina JK, Panjaliya RK, Dogra V, Sharma S, Anupriya, Kumar P. “Association of MTHFR and MS/MTR gene polymorphisms with congenital heart defects in North Indian population (Jammu and Kashmir): a case–control study encompassing meta-analysis and trial sequential analysis”. BMC Pediatr. 2022;22(1):223. Raina JK, Panjaliya RK, Dogra V, Sharma S, Anupriya, Kumar P. “Association of MTHFR and MS/MTR gene polymorphisms with congenital heart defects in North Indian population (Jammu and Kashmir): a case–control study encompassing meta-analysis and trial sequential analysis”. BMC Pediatr. 2022;22(1):223.
35.
Zurück zum Zitat Ramachandra NB. Nuclear co-repressor 1: a potential candidate gene in the manifestation of congenital heart diseases. Int J Hum Genet. 2020;20:55–65. Ramachandra NB. Nuclear co-repressor 1: a potential candidate gene in the manifestation of congenital heart diseases. Int J Hum Genet. 2020;20:55–65.
36.
Zurück zum Zitat Cordell HJ, Töpf A, Mamasoula C, Postma AV, Bentham J, Zelenika D, et al. Genome-wide association study identifies loci on 12q24 and 13q32 associated with Tetralogy of Fallot. Hum Mol Genet. 2013;22:1473–81.PubMedPubMedCentralCrossRef Cordell HJ, Töpf A, Mamasoula C, Postma AV, Bentham J, Zelenika D, et al. Genome-wide association study identifies loci on 12q24 and 13q32 associated with Tetralogy of Fallot. Hum Mol Genet. 2013;22:1473–81.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Mitchell LE, Agopian AJ, Bhalla A, Glessner JT, Kim CE, Swartz MD, et al. Genome-wide association study of maternal and inherited effects on left-sided cardiac malformations. Hum Mol Genet. 2015;24:265–73.PubMedCrossRef Mitchell LE, Agopian AJ, Bhalla A, Glessner JT, Kim CE, Swartz MD, et al. Genome-wide association study of maternal and inherited effects on left-sided cardiac malformations. Hum Mol Genet. 2015;24:265–73.PubMedCrossRef
38.
Zurück zum Zitat Hanchard NA, Swaminathan S, Bucasas K, Furthner D, Fernbach S, Azamian MS, et al. A genome-wide association study of congenital cardiovascular left-sided lesions shows association with a locus on chromosome 20. Hum Mol Genet. 2016;25:2331–41.PubMedPubMedCentralCrossRef Hanchard NA, Swaminathan S, Bucasas K, Furthner D, Fernbach S, Azamian MS, et al. A genome-wide association study of congenital cardiovascular left-sided lesions shows association with a locus on chromosome 20. Hum Mol Genet. 2016;25:2331–41.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Lahm H, Jia M, Dreßen M, Wirth F, Puluca N, Gilsbach R, et al. Congenital heart disease risk loci identified by genome-wide association study in European patients. J Clin Invest. 2021;131(2):e141837. Lahm H, Jia M, Dreßen M, Wirth F, Puluca N, Gilsbach R, et al. Congenital heart disease risk loci identified by genome-wide association study in European patients. J Clin Invest. 2021;131(2):e141837.
40.
Zurück zum Zitat Cunningham F, Allen JE, Allen J, et al. Ensembl 2022. Nucleic Acids Res. 2022;50(D1):D988–95. Cunningham F, Allen JE, Allen J, et al. Ensembl 2022. Nucleic Acids Res. 2022;50(D1):D988–95.
41.
Zurück zum Zitat Juyal RC, Negi S, Wakhode P, Bhat S, Bhat B, Thelma BK. Potential of ayurgenomics approach in complex trait research: leads from a pilot study on rheumatoid arthritis. PLoS One. 2012;7(9):e45752. Juyal RC, Negi S, Wakhode P, Bhat S, Bhat B, Thelma BK. Potential of ayurgenomics approach in complex trait research: leads from a pilot study on rheumatoid arthritis. PLoS One. 2012;7(9):e45752.
42.
Zurück zum Zitat Negi S, Juyal G, Senapati S, Prasad P, Gupta A, Singh S, et al. A genome-wide association study reveals ARL15, a novel non-HLA susceptibility gene for rheumatoid arthritis in North Indians. Arthritis Rheum. 2013;65:3026–35.PubMedCrossRef Negi S, Juyal G, Senapati S, Prasad P, Gupta A, Singh S, et al. A genome-wide association study reveals ARL15, a novel non-HLA susceptibility gene for rheumatoid arthritis in North Indians. Arthritis Rheum. 2013;65:3026–35.PubMedCrossRef
43.
Zurück zum Zitat Kang M, Choi S, Koh I. The effect of increasing control-to-case ratio on statistical power in a simulated case-control SNP association study. Genomics Inform. 2009;7(3):148–51. Kang M, Choi S, Koh I. The effect of increasing control-to-case ratio on statistical power in a simulated case-control SNP association study. Genomics Inform. 2009;7(3):148–51.
46.
Zurück zum Zitat Gauderman WJ. Sample size requirements for association studies of gene-gene interaction. 2002;155(5):478–84. Gauderman WJ. Sample size requirements for association studies of gene-gene interaction. 2002;155(5):478–84.
47.
Zurück zum Zitat Ghosh P, Bhaumik P, Ghosh S, Ozbek U, Feingold E, Maslen C, et al. Polymorphic haplotypes of CRELD1 differentially predispose down syndrome and euploids individuals to atrioventricular septal defect. Am J Med Genet Part A. 2012;158 A:2843–48. Ghosh P, Bhaumik P, Ghosh S, Ozbek U, Feingold E, Maslen C, et al. Polymorphic haplotypes of CRELD1 differentially predispose down syndrome and euploids individuals to atrioventricular septal defect. Am J Med Genet Part A. 2012;158 A:2843–48.
48.
Zurück zum Zitat Ritter A, Leonard J, Gray C, Izumi K, Levinson K, Nair DR, et al. MYH7 variants cause complex congenital heart disease. Am J Med Genet Part A. 2022;188:2772–6.PubMedCrossRef Ritter A, Leonard J, Gray C, Izumi K, Levinson K, Nair DR, et al. MYH7 variants cause complex congenital heart disease. Am J Med Genet Part A. 2022;188:2772–6.PubMedCrossRef
49.
Zurück zum Zitat Barrick SK, Greenberg MJ. Cardiac myosin contraction and mechanotransduction in health and disease. J Biol Chem. 2021;297(5):101297. Barrick SK, Greenberg MJ. Cardiac myosin contraction and mechanotransduction in health and disease. J Biol Chem. 2021;297(5):101297.
50.
Zurück zum Zitat Bjornsson T, Thorolfsdottir RB, Sveinbjornsson G, Sulem P, Norddahl GL, Helgadottir A, et al. A rare missense mutation in MYH6 associates with non-syndromic coarctation of the aorta. Eur Heart J. 2018;39:3243–9.PubMedPubMedCentralCrossRef Bjornsson T, Thorolfsdottir RB, Sveinbjornsson G, Sulem P, Norddahl GL, Helgadottir A, et al. A rare missense mutation in MYH6 associates with non-syndromic coarctation of the aorta. Eur Heart J. 2018;39:3243–9.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Desjardins CA, Naya FJ. The function of the mef2 family of transcription factors in cardiac development, cardiogenomics, and direct reprogramming. Journal of Cardiovascular Development and Disease. 2016;3(3):26. Desjardins CA, Naya FJ. The function of the mef2 family of transcription factors in cardiac development, cardiogenomics, and direct reprogramming. Journal of Cardiovascular Development and Disease. 2016;3(3):26.
52.
Zurück zum Zitat Norland K, Sveinbjornsson G, Thorolfsdottir RB, Davidsson OB, Tragante V, Rajamani S, et al. Sequence variants with large effects on cardiac electrophysiology and disease. Nat Commun. 2019;10:1–10.CrossRef Norland K, Sveinbjornsson G, Thorolfsdottir RB, Davidsson OB, Tragante V, Rajamani S, et al. Sequence variants with large effects on cardiac electrophysiology and disease. Nat Commun. 2019;10:1–10.CrossRef
53.
Zurück zum Zitat Ntalla I, Weng LC, Cartwright JH, Hall AW, Sveinbjornsson G, Tucker NR, et al. Multi-ancestry GWAS of the electrocardiographic PR interval identifies 202 loci underlying cardiac conduction. Nat Commun. 2020;11(1):2542. Ntalla I, Weng LC, Cartwright JH, Hall AW, Sveinbjornsson G, Tucker NR, et al. Multi-ancestry GWAS of the electrocardiographic PR interval identifies 202 loci underlying cardiac conduction. Nat Commun. 2020;11(1):2542.
54.
Zurück zum Zitat Anfinson M, Fitts RH, Lough JW, James JM, Simpson PM, Handler SS, et al. Significance of α-Myosin Heavy Chain (MYH6) variants in hypoplastic left heart syndrome and related cardiovascular diseases. J Cardiovasc Dev Dis. 2022;9(5):144. Anfinson M, Fitts RH, Lough JW, James JM, Simpson PM, Handler SS, et al. Significance of α-Myosin Heavy Chain (MYH6) variants in hypoplastic left heart syndrome and related cardiovascular diseases. J Cardiovasc Dev Dis. 2022;9(5):144.
55.
Zurück zum Zitat Shen C, Xu L, Sun X, Sun A, Ge J. Genetic variants in Chinese patients with sporadic dilated cardiomyopathy: a cross-sectional study. Ann Transl Med. 2022;10:129–129.PubMedPubMedCentralCrossRef Shen C, Xu L, Sun X, Sun A, Ge J. Genetic variants in Chinese patients with sporadic dilated cardiomyopathy: a cross-sectional study. Ann Transl Med. 2022;10:129–129.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Basu R, Hazra S, Shanks M, Paterson DI, Oudit GY. Novel mutation in exon 14 of the sarcomere gene MYH7 in familial left ventricular noncompaction with bicuspid aortic valve. Circ Hear Fail. 2014;7:1059–62.CrossRef Basu R, Hazra S, Shanks M, Paterson DI, Oudit GY. Novel mutation in exon 14 of the sarcomere gene MYH7 in familial left ventricular noncompaction with bicuspid aortic valve. Circ Hear Fail. 2014;7:1059–62.CrossRef
57.
Zurück zum Zitat Alawani S, Paul A, Krishna M, Ahamed H. Familial left ventricular noncompaction cardiomyopathy due to a novel mutation in the MYH 7 gene. Ann Pediatr Cardiol. 2021;14:544–46.PubMedCrossRef Alawani S, Paul A, Krishna M, Ahamed H. Familial left ventricular noncompaction cardiomyopathy due to a novel mutation in the MYH 7 gene. Ann Pediatr Cardiol. 2021;14:544–46.PubMedCrossRef
58.
Zurück zum Zitat Rani S, Masood S. Predicting congenital heart disease using machine learning techniques. J Discret Math Sci Cryptogr. 2020;23:293–303.CrossRef Rani S, Masood S. Predicting congenital heart disease using machine learning techniques. J Discret Math Sci Cryptogr. 2020;23:293–303.CrossRef
59.
Zurück zum Zitat McGregor TL, Misri A, Bartlett J, Orabona G, Friedman RD, Sexton D, et al. Consanguinity mapping of congenital heart disease in a South Indian population. PLoS One. 2010;5(4):e10286. McGregor TL, Misri A, Bartlett J, Orabona G, Friedman RD, Sexton D, et al. Consanguinity mapping of congenital heart disease in a South Indian population. PLoS One. 2010;5(4):e10286.
60.
Zurück zum Zitat Zhang S, Liu X, Wang T, Chen L, Yang T, Huang P, et al. Association and interaction effect of UCP2 gene polymorphisms and dietary factors with congenital heart diseases in Chinese Han population. Sci Rep. 2021;11:1–10. Zhang S, Liu X, Wang T, Chen L, Yang T, Huang P, et al. Association and interaction effect of UCP2 gene polymorphisms and dietary factors with congenital heart diseases in Chinese Han population. Sci Rep. 2021;11:1–10.
61.
Zurück zum Zitat Tian XY, Ma S, Tse G, Wong WT, Huang Y. Uncoupling protein 2 in cardiovascular health and disease. Front Physiol. 2018;9:1060. Tian XY, Ma S, Tse G, Wong WT, Huang Y. Uncoupling protein 2 in cardiovascular health and disease. Front Physiol. 2018;9:1060.
62.
Zurück zum Zitat Luo L, Huang P, Wang TT, et al. Association of maternal diabetes mellitus and UCP2 gene polymorphisms with congenital heart disease in offspring: a case-control study. Chinese J Contemp Pediatr. 2020;22:1092–99. Luo L, Huang P, Wang TT, et al. Association of maternal diabetes mellitus and UCP2 gene polymorphisms with congenital heart disease in offspring: a case-control study. Chinese J Contemp Pediatr. 2020;22:1092–99.
63.
Zurück zum Zitat Junco-Vicente A, Río-García Á Del, Martín M, Rodríguez I. Update in biomolecular and genetic bases of bicuspid aortopathy. Int J Mol Sci. 2021;22(11):5694. Junco-Vicente A, Río-García Á Del, Martín M, Rodríguez I. Update in biomolecular and genetic bases of bicuspid aortopathy. Int J Mol Sci. 2021;22(11):5694.
64.
Zurück zum Zitat Bilan F, Legendre M, Charraud V, Manière B, Couet D, Gilbert-Dussardier B, et al. Complete screening of 50 patients with CHARGE syndrome for anomalies in the CHD7 gene using a denaturing high-performance liquid chromatography-based protocol: New guidelines and a proposal for routine diagnosis. J Mol Diagnostics. 2012;14:46–55.CrossRef Bilan F, Legendre M, Charraud V, Manière B, Couet D, Gilbert-Dussardier B, et al. Complete screening of 50 patients with CHARGE syndrome for anomalies in the CHD7 gene using a denaturing high-performance liquid chromatography-based protocol: New guidelines and a proposal for routine diagnosis. J Mol Diagnostics. 2012;14:46–55.CrossRef
65.
Zurück zum Zitat Bagheri M, Wang C, Shi M, Manouchehri A, Murray KT, Murphy MB, et al. The genetic architecture of plasma kynurenine includes cardiometabolic disease mechanisms associated with the SH2B3 gene. Sci Rep. 2021;11:1–10.CrossRef Bagheri M, Wang C, Shi M, Manouchehri A, Murray KT, Murphy MB, et al. The genetic architecture of plasma kynurenine includes cardiometabolic disease mechanisms associated with the SH2B3 gene. Sci Rep. 2021;11:1–10.CrossRef
66.
Zurück zum Zitat Ramachandran D, Zeng Z, Locke AE, Mulle JG, Bean LJH, Rosser TC, et al. Genome-wide association study of down syndrome-associated atrioventricular septal defects. G3 (Bethesda). 2015;5:1961–71. Ramachandran D, Zeng Z, Locke AE, Mulle JG, Bean LJH, Rosser TC, et al. Genome-wide association study of down syndrome-associated atrioventricular septal defects. G3 (Bethesda). 2015;5:1961–71.
67.
Zurück zum Zitat Rani DS, Vijaya Kumar A, Nallari P, Sampathkumar K, Dhandapany PS, Narasimhan C, et al. Novel Mutations in β-MYH7 Gene in Indian Patients With Dilated Cardiomyopathy. CJC Open. 2022;4:1–11.PubMedCrossRef Rani DS, Vijaya Kumar A, Nallari P, Sampathkumar K, Dhandapany PS, Narasimhan C, et al. Novel Mutations in β-MYH7 Gene in Indian Patients With Dilated Cardiomyopathy. CJC Open. 2022;4:1–11.PubMedCrossRef
68.
Zurück zum Zitat Gaspar HA, Breen G. Probabilistic ancestry maps: A method to assess and visualize population substructures in genetics. BMC Bioinformatics. 2019;20(1):116.CrossRef Gaspar HA, Breen G. Probabilistic ancestry maps: A method to assess and visualize population substructures in genetics. BMC Bioinformatics. 2019;20(1):116.CrossRef
69.
Zurück zum Zitat Li L, Dong J, Wang X, Guo H, Wang H, Zhao J, et al. JAG1 mutation spectrum and origin in Chinese children with clinical features of alagille syndrome. PLoS ONE. 2015;10:1–11. Li L, Dong J, Wang X, Guo H, Wang H, Zhao J, et al. JAG1 mutation spectrum and origin in Chinese children with clinical features of alagille syndrome. PLoS ONE. 2015;10:1–11.
Metadaten
Titel
Case–control association study of congenital heart disease from a tertiary paediatric cardiac centre from North India
verfasst von
Prachi Kukshal
Radha O Joshi
Ajay Kumar
Shadab Ahamad
Prabhatha Rashmi Murthy
Yogesh Sathe
Krishna Manohar
Soma Guhathakurta
Subramanian Chellappan
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Pediatrics / Ausgabe 1/2023
Elektronische ISSN: 1471-2431
DOI
https://doi.org/10.1186/s12887-023-04095-x

Weitere Artikel der Ausgabe 1/2023

BMC Pediatrics 1/2023 Zur Ausgabe

Mit dem Seitenschneider gegen das Reißverschluss-Malheur

03.06.2024 Urologische Notfallmedizin Nachrichten

Wer ihn je erlebt hat, wird ihn nicht vergessen: den Schmerz, den die beim Öffnen oder Schließen des Reißverschlusses am Hosenschlitz eingeklemmte Haut am Penis oder Skrotum verursacht. Eine neue Methode für rasche Abhilfe hat ein US-Team getestet.

Reanimation bei Kindern – besser vor Ort oder während Transport?

29.05.2024 Reanimation im Kindesalter Nachrichten

Zwar scheint es laut einer Studie aus den USA und Kanada bei der Reanimation von Kindern außerhalb einer Klinik keinen Unterschied für das Überleben zu machen, ob die Wiederbelebungsmaßnahmen während des Transports in die Klinik stattfinden oder vor Ort ausgeführt werden. Jedoch gibt es dabei einige Einschränkungen und eine wichtige Ausnahme.

Alter der Mutter beeinflusst Risiko für kongenitale Anomalie

28.05.2024 Kinder- und Jugendgynäkologie Nachrichten

Welchen Einfluss das Alter ihrer Mutter auf das Risiko hat, dass Kinder mit nicht chromosomal bedingter Malformation zur Welt kommen, hat eine ungarische Studie untersucht. Sie zeigt: Nicht nur fortgeschrittenes Alter ist riskant.

Begünstigt Bettruhe der Mutter doch das fetale Wachstum?

Ob ungeborene Kinder, die kleiner als die meisten Gleichaltrigen sind, schneller wachsen, wenn die Mutter sich mehr ausruht, wird diskutiert. Die Ergebnisse einer US-Studie sprechen dafür.

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.