Abstract Tetralogy of Fallot (TOF), the most common severe cyanotic congenital heart disease, has unclear genetic causes. Through next-generation sequencing in 131 patients with nonsyndromic TOF, we identified an increased burden of rare deleterious variants in ciliary genes and cilium pathway and observed a multigenic inheritance pattern, with an odds ratio (OR) of 1.672 [95% confidence interval (CI), 1.120 to 2.547; P = 0.0104] for more than two deleterious variants and a cumulative OR of 3.158 (95% CI, 1.381 to 6.371; P = 0.0038) for six variants. Functional validation in single- and double-heterozygous mouse models carrying these variants recapitulated TOF-like phenotypes and impaired normal cilia structure and function, particularly disrupting Hedgehog signaling in cardiomyocytes, and down-regulating key transcription factors Gata4 and Nkx2-5. Together, our study provides compelling evidence linking ciliary gene variants to a heightened risk of TOF in Han Chinese, offering valuable genetic insights into the etiology and pathogenesis of nonsyndromic TOF and supporting a multigenic inheritance model for the disease. __________________________________________________________________ Ciliary gene variants contribute to TOF risk, revealing a multigenic inheritance pattern and insights into TOF pathogenesis. INTRODUCTION Congenital heart disease (CHD) is the most common type of congenital malformation, accounting for 28% of all major congenital anomalies ([62]1) and affecting 8 per 1000 live births ([63]2). Tetralogy of Fallot (TOF) is the most frequent cyanotic CHD, with a global prevalence of about 1 in 3000 live births ([64]3). TOF is considered a malformation of the cardiac outflow tract (OFT) that includes structural abnormalities, i.e., overriding aorta (OA), ventricular septal defects (VSD), and pulmonary artery (PA) stenosis, and compensated right ventricular hypertrophy ([65]4). With recent advances in the diagnosis, surgical treatment, and postoperative care, almost all of those born with TOF can now be expected to survive to adulthood after successful surgical correction at a young age ([66]4). Unfortunately, as long-term sequelae, patients with TOF have high morbidity of arrhythmia and ventricular dysfunction after 40 years of age ([67]5, [68]6), which seriously affect quality of life and bring a heavy burden to the society. TOF is a complicated disease influenced by both genetic and environmental factors ([69]7). Approximately 20% of patients with TOF present with additional extracardiac anomalies and are classified as syndromic TOF ([70]8). Among them, 22q11.2 deletion syndrome (DiGeorge syndrome) and trisomy 21 (Down syndrome) account for approximately 15 and 7% of cases, respectively ([71]9). Multiple pathogenic loci, such as chromosomal copy number variations (CNVs) at 22q11.2 and mutations in genes like GATA4, have been identified in syndromic TOF and provided valuable insights into its underlying mechanisms ([72]9, [73]10). Nevertheless, nearly 80% of cases are sporadic and nonsyndromic TOF, with no clearly identified etiology or major mutated genes ([74]11, [75]12). A recent large-scale study involving nine research centers across Europe and Australia identified NOTCH1 as the most frequently mutated genes in nonsyndromic TOF, followed by FLT4 ([76]13). To date, despite the well-established function of several genes in the pathogenesis of TOF, only a small fraction of nonsyndromic TOF cases can be attributed to a detectable abnormality in any one of the known TOF genes identified: CNVs in the TBX1 gene located in the 22q11.2 region (approximately 6%) ([77]14) and NKX2.5 mutations (approximately 4%) ([78]15). The underlying causes remain unknown for most patients with nonsyndromic TOF. In addition, the lack of experimental profiling to determine the biological mechanisms involved in these genetic variants hampers our in-depth knowledge of the disease. To systematically investigate the genetic etiology of nonsyndromic TOF, we performed next-generation sequencing on 131 patients with sporadic TOF. By applying frequency-based filtering using both our internal control cohort of 6004 individuals without CHDs ([79]16) and public databases including Exome Aggregation Consortium (ExAC) and Genome Aggregation Database (gnomAD), we defined rare loss-of-function (LoF) variants as potential pathogenic candidates and found that these variants were significantly enriched in the cilia-related pathways in patients with TOF [false discovery rate (FDR)–adjusted P <0.05]. Further gene-level association analyses revealed that ciliary genes exhibited a notably higher burden of rare LoF variants in TOF cases compared to controls. The odds ratios (ORs) of ciliary gene variants for patients with TOF were calculated with different numbers of LoF variants and detrimental missense variants, and the results implied the potential oligogenic or multigenic inheritance pattern. Moreover, mouse models carrying heterozygous or homozygous knockout/mutant alleles of these ciliary genes reproduced a spectrum of cardiac phenotypes resembling TOF. Double-heterozygous mice generated through pairwise crosses of ciliary gene mutant lines exhibited an even higher incidence of TOF-like phenotypes, compared to single-heterozygous or homozygous mutants, suggesting additive or synergistic effects between distinct ciliary gene variants. In conclusion, this study demonstrates the pathogenic role of ciliary gene variants in OFT development and provides compelling genetic evidence for the etiology of nonsyndromic TOF, with both human and mouse model data supporting a multigenic inheritance mechanism underlying its pathogenesis. RESULTS Identification of pathogenic variants in known TOF-causative genes We first examined rare and deleterious variants in known causative TOF genes. To compile a list of candidate genes, we searched the OMIM and ClinVar databases and integrated pathogenic genes identified in several large-scale, multicenter studies on TOF using whole-exome sequencing (WES) and whole-genome sequencing (WGS) ([80]3, [81]10, [82]11, [83]13, [84]17, [85]18). This resulted in the inclusion of 11 genes: TBX1, NKX2-5, GATA4, ZFPM2, GATA6, JAG1, NOTCH1, FLT4, FOXC2, TBX5, and KDR for pathogenicity evaluation. We then assessed rare and deleterious variants in these known TOF-causative genes (fig. S1A). Rare variants were defined as those with a minor allele frequency (MAF) < 0.5% in our study cohorts and < 0.5% in both the ALL and East Asian populations of gnomAD and ExAC; variant pathogenicity was evaluated through manual review following the American College of Medical Genetics and Genomics (ACMG) guidelines, or annotation in the ClinVar database (table S1). Ultimately, we identified 15 pathogenic and likely pathogenic (P/LP) variants across nine known TOF genes, including 2 (13.3%) LoF variants, 12 (80.0%) missense variants, and 1 (6.7%) start lost variant (fig. S1, B to D). These P/LP variants were detected in 16 patients, yielding a 12.21% contribution to the overall TOF incidence. Identification of ciliary gene variants in nonsyndromic TOF patients Because of the relatively low explanatory power of known TOF-causative genes, we then conducted additional case-control analyses at both the gene set (pathway) and gene levels to identify enriched genes and potential genetic defects associated with nonsyndromic TOF. Since the accumulation of mild-effect variants may collectively increase disease susceptibility, gene set level analysis offers valuable insight into disease mechanisms and may aid in potential gene discovery. Following the screening procedure shown in [86]Fig. 1A, we analyzed rare and deleterious variants in 131 patients with nonsyndromic TOF. Variants were filtered on the basis of MAF_all <0.5% in our TOF and control individuals, as well as <0.5% in both of the populations (ALL and East Asians population) from gnomAD and ExAC. Deleterious variants were then defined as LoF variants, namely frameshift, stop gain, and splice site variants (splice acceptor/donor). Last, a total of 2018 rare LoF variants on 1777 genes were identified in 131 patients with TOF. Subsequently, we performed pathway enrichment analysis and found significant enrichment in the cilia-related pathways (P = 3.63 × 10^−8, FDR<0.05). A similar result was observed for high-confidence LoF variants predicted by the LoF transcript effect estimator ([87]19) package ([88]Fig. 1B). There were 108 candidate genes involved in the cilium pathway (table S2) with different localizations in cilia (fig. S2). In total, 87 of 131 cases harbored ciliary gene LoF variants, accounting for 66.4% of TOF cases. Fig. 1. Identification of ciliary gene variants in patients with nonsyndromic TOF. [89]Fig. 1. [90]Open in a new tab (A) Flowchart depicting the criteria and results for variant selection after next-generation sequencing. Rare and putatively damaging variants were filtered according to the MAFs in our reference cohort and public databases (ExAC and gnomAD), along with the predicted deleterious LoF variants. (B) Genes with rare LoF variants or variants evaluated as high-confidence (HC) using the LoF transcript effect estimator (LOFTEE) package were both significantly enriched in cilia-related pathways (FDR < 0.05). (C) Q-Q plot comparing observed versus expected P values for LoF variants in each gene between cases (n = 131) and controls (n = 6004), using two-sided Fisher’s exact test. Thirteen genes with FDR < 0.2 are highlighted, with ciliary genes marked in red. (D) Pathway enrichment analysis of significantly mutated genes that met the threshold criteria. (E to I) Immunofluorescence staining of ciliary axonemes (green, acetylated α-tubulin), basal bodies (red, γ-tubulin), and the nucleus (blue) showing the expression of cilia throughout OFT development from E10.5 to E18.5. The white arrows indicate cilia. Three levels of magnification were shown from left to right. Scale bars, 200, 20, and 10 μm. (J) Tomographic electron microscopy of the OFT at E13.5 showing a straight-cut axoneme, a primary cilium (typical 9+0 microtubule configuration), and a motile cilium (typical 9+2 microtubule configuration). Scale bars, 200, 100, 100 nm. (K) Spatial expression of all ciliary genes in cilium pathway across different cardiac structures. The centerline on the boxplot represents the median, the box limits indicate the 25th and the 75th percentiles, and the whiskers extend to the minimum and maximum values. The statistical analysis was carried out using Wilcoxon–Mann-Whitney test. *P < 0.05, **P < 0.01. To further explore the candidate pathogenic genes in TOF, we also conducted gene-based association analyses in rare deleterious variants. We aggregated the qualifying alleles into genes and tested for differences between TOF cases and controls using Fisher’s exact test. For genes with LoF variants, 13 genes showed a significantly higher burden of LoF alleles in TOF cases after multiple testing (FDR < 0.2 threshold; [91]Fig. 1C), with pathways related to ciliary membrane and cilium enriched in these genes (FDR < 0.05; [92]Fig. 1D). Notably, three ciliary genes, MOSMO, TAS2R46, and TTLL3, were among the significantly mutated genes. In particular, MOSMO is a well-established CHD risk gene which is active in ciliary membrane ([93]20). Loss of MOSMO results in severe developmental defects in mouse embryos, including heterotaxy, skeletal abnormalities, and complex CHDs such as double outlet right ventricle (DORV), transposition of the great arteries (TGA), and persistent truncus arteriosus, which arise due to defects in OFT development ([94]21). In parallel, we performed gene-based association tests for predicted detrimental missense variants, using multiple in silico tools (AlphaMissense, CADD, SIFT, and PolyPhen-2) to evaluate variant pathogenicity. Ciliary genes consistently ranked among the top genes with higher burdens of detrimental missense variants in TOF cases compared to in-house controls across all algorithms (fig. S3). Together, we imply the potential role of ciliary gene variants in the pathogenesis of TOF. Cilia are spatially and temporally regulated during cardiac OFT development Since TOF is characterized by abnormal development of the cardiac OFT, we initiated experiments to determine whether cilia contribute to OFT development by first performing immunofluorescence assays. As shown in [95]Fig. 1, E to I, cilia were distributed throughout all stages of cardiac OFT development from embryonic day 10.5 (E10.5) to E18.5. These findings were confirmed by transmission electron microscopy (TEM), which revealed both typical “9+0” (primary cilia) and “9+2” (motile cilia) microtubule configurations in the cardiac OFT ([96]Fig. 1J). In addition to the OFT, primary cilia were also observed in other cardiac regions, such as the atrioventricular canal (AVC), atria, ventricles, and valves, at various stages of development (fig. S4, A to M). Notably, during early heart development, primary cilia were most abundant in the OFT, where they were likely involved in the regulation of key signaling pathways, blood flow dynamics, and guiding neural crest cell migration, contributing to OFT development and the proper separation of the aorta and PA. In contrast, motile cilia exhibited a distinct spatiotemporal expression pattern and were less abundant compared to primary cilia, with a role primarily in biomechanical regulation, contributing to fluid dynamics, extracellular matrix (ECM) remodeling, and cell migration, particularly in the OFT and valve regions (fig. S4, N to Z). To further investigate the heterogeneity in the distribution of cilia within distinct anatomical regions of the heart, by intersection with spatial transcriptomics data of E10.5 hearts ([97]22), we revealed that ciliary genes showed notably higher spatial expression in the OFT [including in OFT cardiac neural crest cells (cNCCs), OFT cardiomyocytes, and mesenchymal cells] (P < 0.05), supporting the embryological hypotheses of aberrant positioning of the OFT in TOF ([98]Fig. 1K). In parallel, we performed single-cell RNA sequencing (scRNA) on anatomically dissected E10.5 hearts, dividing them into six regions: OFT, AVC, left atrium (LA), right atrium (RA), left ventricle (LV), and right ventricle (RV). Consistent with the spatial transcriptomics results, ciliary gene expression was highest in the AVC and OFT regions (fig. S5, A to B). Given the role of the AVC in endothelial-to-mesenchymal transition and cushion formation, which gives rise to both AV and OFT cushions, this expression pattern further highlights the regional specificity of ciliary gene activity during OFT development. To provide broader context for ciliary gene expression, we also analyzed transcriptomic datasets across multiple tissues ([99]23) and cardiac cell types. Bulk RNA-seq data showed that ciliary genes are broadly expressed during development, with notably high expression in the heart, brain, and testes. Genes highly expressed in the heart (clusters 5, 6, and 7) exhibited a greater LoF variant burden in TOF cases compared to controls (OR, 1.96, P = 0.002, 95% CI, 1.255 to 2.976), supporting their potential contribution to cardiac-specific phenotypes (fig. S5C). ScRNA-seq analysis further revealed that ciliary genes are expressed across diverse cardiac cell types, including cardiomyocytes, mesenchymal cells, and endocardial cells, where they participate in distinct signaling pathways critical for OFT development (fig. S5D). Together, these findings highlight that although ciliary genes are broadly expressed across multiple tissues and cardiac regions, they show particularly elevated expression and functional relevance in the OFT during early heart development, supporting their critical role in the pathogenesis of TOF. Evaluation of the impacts of ciliary gene variants in the pathogenesis of nonsyndromic TOF Increasing evidence indicates that in addition to monogenic inheritance, CHD may involve oligogenic or multigenic inheritance, which is characterized by the presence of damaging heterozygous mutations across more than one gene ([100]24, [101]25). To systematically evaluate the overall contribution of ciliary gene variants to TOF pathogenesis and assess their predominant inheritance pattern, we performed gene set–based association analyses focusing on deleterious variants in all genes involved in the cilium pathway ([102]Fig. 2A and table S3). ORs were calculated by comparing the number of patients with TOF and controls carrying different burdens of LoF variants within this predefined gene set. We observed an increased burden of ciliary gene LoF variants among patients with TOF compared to controls (OR, 1.540; 95% CI, 1.056 to 2.775; P = 0.0205). This association was more pronounced in individuals carrying two or more LoF variants (OR, 1.888; 95% CI: 1.265 to 2.778; P = 0.0017; [103]Fig. 2B). A similar enrichment pattern was observed for putatively detrimental missense variants in patients with TOF, with an OR of 1.590 (95% CI: 1.106 to 2.284; P = 0.0105) in individuals carrying more than two such variants ([104]Fig. 2B). When combining the impacts of LoF variants and detrimental missense variants, we observed that as the number of variants increased, the risk for TOF increased accordingly. Specifically, when the number of variants reached six, our TOF cohort exhibited greater than threefold enrichment (OR, 3.158; 95% CI, 1.381 to 6.371; P = 0.0038), thus supporting a multigenic etiology of ciliary genes in the pathogenesis of TOF ([105]Fig. 2B). Fig. 2. Rare LoF variants and detrimental missense variants burden analyses revealed the multigenic inheritance effect of ciliary genes in patients with nonsyndromic TOF. [106]Fig. 2. [107]Open in a new tab (A) Flowchart illustrating the selection of ciliary gene variants for gene set–based association analyses between 131 patients with nonsyndromic TOF and 6004 in-house control individuals. (B) Forest plot showing the burden test results for patients with TOF carrying different thresholds of LoF variants, detrimental missense variants, and combined deleterious variants (LoF plus detrimental missense variants) in ciliary genes, compared to control individuals. The number of participants in each category is displayed, where N represents the number of distinct heterozygous variants detected across different ciliary genes. The analysis includes biallelic heterozygous variants. To control for potential confounding effects from biallelic inheritance, we reevaluated the model after excluding individuals carrying biallelic variants. Among the 131 patients with TOF, such variants were identified in 6.87% (9 of 131), with LoF variants accounting for 2.29% (3 of 131) and detrimental missense variants for 3.05% (4 of 131) (fig. S6A). Notably, a substantial proportion (up to 66.41%) of TOF cases carried more than two pathogenic variants. After removing biallelic individuals, the multiheterozygous burden analysis in the remaining 122 TOF cases and 6004 controls yielded consistent trends, with increasing TOF risk observed as the number of pathogenic variants increased. Specifically, ORs were 2.136, 2.601, and 3.149 for individuals carrying more than four, five, and six variants, respectively (fig. S6B). These results reinforce a multigenic inheritance model in TOF and suggest that cumulative variant burden in ciliary genes may contribute to a substantial proportion of unsolved cases. Loss of ciliary genes impairs normal cardiac OFT development To explore the causative effects of ciliary genes on cardiac OFT development in vivo, we prioritized genes harboring rare LoF variants enriched in the cilium pathway ([108]Fig. 1A and fig. S7A). Among 90 previously unreported candidate genes (i.e., genes not confirmed to be associated with CHD in OMIM database or genes without CHD phenotypes in mice from MGI database), genes with recurrent variants were selected (n = 17), and after filtering out variants that were poorly conserved (<60%) in humans and mice (n = 6), failed Sanger sequencing validation (n = 1), and had technical difficulties in mouse model construction (n = 4), we ultimately opted to construct mouse models for the retained six ciliary genes, including AGBL2, MLF1, PPEF2, PKHD1L1, SPTBN5, and TEKT3. Together, we identified a heterozygous frameshift variant (c.2683_2684delTC, p.Ser895fs) and a heterozygous stop-gain variant (c.742C>T, p.Arg248*) in AGBL2; a heterozygous stop-gain variant (c.778C>T and p.Arg260*) in two individuals in MLF1; three heterozygous frameshift variants (c.1820_1821delTT, p.Phe607fs; c.2714_2717dupCAGA, p.Glu906fs; and c.4727delG, p.Gly1576fs) in PKHD1L1; a heterozygous stop-gain variant (c.1038G>A, p.Trp346*) and a heterozygous frameshift variant (c.1632_1633dupAC, p.Leu545fs) in PPEF2; three heterozygous frameshift variants (c.10970_10973delATGA, p.Asn3657fs; c.9417dupT, p.Val3140fs; c.1318_1319delAG, p.Ser440fs) in SPTBN5; a heterozygous stop-gain variant (c.544G>T, p.Glu182*) and a heterozygous frameshift variant (c.546_547delGCinsT, p.Glu182fs) in three individuals in TEKT3 (fig. S7B and table S4). Using CRISPR-Cas9 technology, we then generated six ciliary gene knockout or mutant mouse models (fig. S8). Sanger sequencing of homozygous mutant mice confirmed the presence of deletions in Agbl2, Pkhd1l1, Sptbn5, Tekt3, and anthropomorphic variants in Mlf1 and Ppef2 (table S5), which were predicted to cause premature translational termination or gene inactivation. Meanwhile, no predicted off-target sites (OTSs) were found (fig. S9 and table S6). Compared with wild-type mouse hearts, protein immunoblotting and immunofluorescence showed substantial decrease in the expression of the corresponding proteins in the heart tissues of knockout or mutant mice ([109]Fig. 3A and figs. S10 to S14A). As homozygous inactivation of mouse ciliary genes has been reported to cause embryonic lethality ([110]26, [111]27), postnatal day one (P1) mice from heterozygous parents were examined. The genotype ratio from P1 mice was in accordance with the expected Mendelian ratio among the offspring ([112]Fig. 3B and figs. S10 to S14B), whereas homozygous P1 mice suffered from higher postnatal lethality ([113]Fig. 3, C to D, and figs. S10 to S14, C and D). Then, we examined the cardiac phenotypes of each strain. Dissected and fixed P1 hearts showed abnormal and malrotation of the aorta and PA in Pkhd1l1^−/− hearts ([114]Fig. 3E). Further experiment of hematoxylin and eosin (H&E) staining of P1 Pkhd1l1^−/− hearts revealed cardiac malformations in a subset of homozygous mice. Among them, 9 of 73 (12.33%) exhibited an OA in combination with a VSD (OA and VSD), while 7 of 73 (9.59%) showed muscular VSDs (MU-VSD), and 11 of 73 (15.07%) presented with secundum atrial septal defects (ASD), together accounting for 39.7% of the homozygotes. Notably, cardiac defects were also observed in heterozygous mice, including OA and VSD in 6 of 94 (6.38%), MU-VSD in 5 of 94 (5.32%), ASD in 5 of 94 (5.32%), and combined MU-VSD and ASD in 1 of 94 (1.06%) heterozygotes. However, no similar structural defects were observed in P1 hearts of wild-type mice ([115]Fig. 3, C and F). Mice that had died on P1 day mostly exhibited structural cardiac defects ([116]Fig. 3D). Moreover, the PA widths of the hearts in P1 Pkhd1l1^−/− mice appeared to be thinner than those of the normal controls ([117]Fig. 3G). Together, knockout of the ciliary gene Pkhd1l1 resulted in a spectrum of cardiac malformations, including OA with VSD and other defects affecting septal development and OFT formation, particularly VSD. Fig. 3. Pkhd1l1-knockout mice showed typical abnormal cardiac OFT phenotypes. [118]Fig. 3. [119]Open in a new tab (A) Immunofluorescence staining of PKHD1L1 in E10.5 OFT sections from wild-type (Pkhd1l1^+/+) and homozygous (Pkhd1l1^−/−) mice. Scale bars, 200 and 10 μm. (B) Genotype frequency analysis of offspring from Pkhd1l1^+/− heterozygote crosses, showing the expected Mendelian ratio for P1 newborns (χ2 = 0.2818, P>0.05). (C) Postnatal lethality and cardiac defects (OA and VSD, MU-VSD, ASD, and MU-VSD with ASD) in wild-type, heterozygous, and homozygous P1 offspring. (D) Proportions of normal and defective phenotypes in Pkhd1l1 knockout mice that died on P1. (E) Malrotation of the aorta and PA in Pkhd1l1^−/− hearts at P1. Scale bar, 1000 μm. (F) Representative H&E staining of abnormal hearts from Pkhd1l1^−/− homozygous offspring at P1, showing OA and VSD, MU-VSD, and ASD (asterisk). Scale bar, 500 μm. (G) Measurements of P1 hearts indicated thinner pulmonary arteries due to Pkhd1l1 deletion. The outline and diameter of the PA were indicated by black lines. Statistical analysis was performed using an unpaired two-tailed Student’s t test (**P < 0.01). n>30 mice per group. Scale bar, 1000 μm. (H) E10.5 wild-type and Pkhd1l1 deletion embryos were shown, with enlarged view of the OFT below. Homozygous mutants exhibited reduced OFT length compared to littermate controls, confirmed by quantitative measurements normalized by dorsal-ventral axis lengths. The OFT length and dorsal-ventral axes length are represented by white lines and red lines, respectively. Statistical analysis was performed using an unpaired two-tailed Student’s t test (*P < 0.05). n >15 mice per group. Scale bar, 1000 μm. (I and J) Tomographic electron microscopy of E13.5 cardiac OFT cross sections revealed typical 9+0 microtubule structures in Pkhd1l1^+/+ mice, while Pkhd1l1^−/− mice exhibited severely disorganized 9+0 axonemes. Scale bars, 200 nm. OA, overriding aorta; MU-VSD, muscular ventricular septal defect; ASD, secundum atrial septal defect. Proper folding and development of the heart tube requires recruitment of second heart field (SHF) cardiac precursors that contribute to the lengthening of the heart tube, eventually giving rise to much of the right ventricle, atria, and OFT ([120]28). Therefore, to further investigate the impact of ciliary gene deletion on early OFT development and morphogenesis, we examined E10.5 embryos and observed decreased size and inadequate looping of the OFTs in several homozygous Pkhd1l1^−/− mice ([121]Fig. 3H). To account for potential variations in overall embryonic development, we normalized OFT lengths to dorsal-ventral axis lengths. Blinded quantitative analysis revealed a significant reduction in normalized OFT length (P < 0.05) in Pkhd1l1^−/− embryos compared to littermate controls, indicating that ciliary gene loss leads to intrinsic defects in OFT elongation during early cardiac development ([122]Fig. 3H). Consistent with the phenotypes observed in Pkhd1l1^−/− mouse hearts, knockout or mutation of other ciliary genes also led to similar cardiac phenotypes, with a phenotypic incidence of approximately 30% across strains. In Agbl2^−/− mice, 31.9% (15 of 47) of homozygotes exhibited cardiac defects, including OA and VSD in one mouse (2.13%), MU-VSD in eight (17.02%), ASD in five (10.64%), and combined MU-VSD and ASD in one (2.13%). In Mlf1^KI/KI mice, the defect rate was 23.4% (15 of 64), comprising OA and VSD in one (1.56%), MU-VSD in four (6.25%), ASD in nine (14.06%), and MU-VSD with ASD in one (1.56%). In Ppef2^KI/KI mice, 28.1% (25 of 89) displayed cardiac defects, including OA and VSD in 5 (5.62%), MU-VSD in 5 (5.62%), and ASD in 15 (16.85%). In Sptbn5^−/− mice, 23.8% (15 of 63) had malformations, with OA and VSD in five (7.94%), MU-VSD in three (4.76%), ASD in six (9.52%), and combined MU-VSD and ASD in one (1.59%). In Tekt3^−/− mice, 31.3% (15 of 48) were affected, including OA and VSD in one (2.08%), MU-VSD in five (10.42%), ASD in eight (16.67%), and MU-VSD with ASD in one (2.08%) (figs. S10 to S14, C and D). In addition, we identified OFT and septal malformations in each heterozygous strain, whereas no cardiac developmental defects were observed at P1 in wild-type mouse hearts in ciliary genes above, which illuminated the supposed burden and pathogenicity of patients with heterozygous variants (figs. S10 to S14E). Similar results were observed in comparisons of PA widths and E10.5 normalized OFT lengths, with both parameters significantly reduced in all knockout and mutant mouse hearts (P < 0.05) (figs. S10 to S14, F and G). Overall, these findings implicate an important role of ciliary gene variants in OFT development. To assess whether mice with combined OA and VSD exhibit a TOF-like phenotype, we further measured PA widths in these P1 mice. Given the lack of an established reference range for PA widths in P1 mice, we compiled measurements from wild-type P1 littermates derived from heterozygous crosses of each ciliary gene knockout/mutant strain, resulting in an average PA width of 415.23 μm, which was used as the normal reference for P1 mice. PA widths in both heterozygous and homozygous ciliary gene knockout/mutant mice with combined OA and VSD were then compared against this reference. All affected mice exhibited narrower pulmonary arteries than the wild-type average, with a mean PA width of 371.52 μm (fig. S15). The simultaneous presence of OA, VSD, and PA narrowing in these P1 mice recapitulates the key features of the human TOF phenotype. Therefore, the incidence of TOF-like defects across different strains is as follows: Agbl2^−/−: 2.13%, Mlf1^KI/KI: 1.56%, Pkhd1l1^−/−: 12.33%, Ppef2^KI/KI: 5.62%, Sptbn5^−/−: 7.94%, and Tekt3^−/−: 2.08%. Building on the observed phenotypes in single ciliary gene knockout/mutant mice, we next sought to investigate the potential cumulative effects of compound heterozygosity. We performed pairwise crosses of our ciliary gene knockout/mutant mice, generating five distinct double-heterozygous lines ([123]Fig. 4A). A total of 122 P1 hearts were collected for cardiac defect phenotype analysis. The overall incidence of cardiac defects in the double-heterozygous mice was 33.6%, with the incidences for each strain as follows: Pkhd1l1+Tekt3 at 17.4%, Tekt3+Ppef2 at 50.0%, Ppef2+Mlf1 at 42.1%, Pkhd1l1+Mlf1 at 47.6%, and Tekt3+Sptbn5 at 28.6% ([124]Fig. 4A). Among these, OA was observed in 11.48% (14 of 122) of cases, while the incidences of perimembranous VSD (PM-VSD), MU-VSD, and ASD were 4.10, 11.48, and 8.20%, respectively ([125]Fig. 4B). The increased incidence of OA in double-heterozygous mice further suggests a potential cumulative or interactive genetic effect. Moreover, 21 hearts (17.21%) were collected from mice that had died on P1 day, with a markedly higher defect rate of 61.90%. In this group, 19.05% (4 of 21) exhibited OA, 9.52% had PM-VSD, 28.57% had MU-VSD, and 14.29% had ASD ([126]Fig. 4C), indicating that early postnatal mortality was closely associated with cardiac abnormalities. A comparative analysis of overall cardiac defect incidence revealed consistently higher frequencies in double-heterozygous mice compared to single-heterozygous strains ([127]Fig. 4D). In addition, the incidence of OA was higher in double-heterozygous mice compared to single-heterozygous strains and, in most cases, exceeded that observed in homozygous mice ([128]Fig. 4, E to F). In conclusion, the findings from both our human cohort and mouse models support a multigenic inheritance model in TOF. The incomplete penetrance of cardiac phenotypes in heterozygous mice further suggests that the pathogenesis of TOF may arise from the combined effects of deleterious heterozygous variants on more than one ciliary gene. Fig. 4. Cardiac phenotypic analysis of double-heterozygous ciliary gene knockout/mutant mice. [129]Fig. 4. [130]Open in a new tab (A) Breeding scheme for generating double-heterozygous mice. The numbers in colored boxes below indicate the number of cardiac defects observed in each strain, with each color representing a specific phenotype: red for OA + VSD, purple for OA + VSD + ASD, green for PM-VSD, blue for MU-VSD, and orange for ASD. (B) The number and proportion of each cardiac defect phenotype in the double-heterozygous mice. (C) The number and proportion of each cardiac defect phenotype in the double-heterozygous mice that died before heart collection. (D) Comparison of overall cardiac defect probability between single-heterozygous and double-heterozygous ciliary gene knockout/mutant mice. (E) Comparison of OA probability between single-heterozygous and double-heterozygous ciliary gene knockout/mutant mice. (F) Representative H&E staining of abnormal hearts from double-heterozygous mice at P1, showing phenotypes of OA, PM-VSD, MU-VSD, and ASD (asterisk). Scale bars, 500, 500, and 1000 μm. PM-VSD, perimembranous VSD. Loss of ciliary genes results in aberrant ciliogenesis during cardiac OFT development Given the well-established role of cilia structure in maintaining normal ciliary function ([131]29), we further investigated the effects of variants in ciliary genes on cilia phenotype and function in OFTs. We first examined the subcellular localization of these ciliary genes in both mouse embryonic fibroblasts (MEFs) and E10.5 mouse OFTs. Immunofluorescence staining analysis revealed that Mlf1 and Pkhd1l1 colocalized with acetylated α-tubulin and specifically accumulated in cilia axonemes; Sptbn5 colocalized with both acetylated α-tubulin and γ-tubulin along the entire cilia; Ppef2 and Tekt3 were localized to basal bodies of cilia; and Agbl2 was located at the base of cilia and overlapped with Nek2, a marker of centriole ([132]Fig. 5A and fig. S16). Fig. 5. Loss of ciliary genes disrupted ciliogenesis during cardiac OFT development. [133]Fig. 5. [134]Open in a new tab (A) Schematic of a cilium showing structural features and selected ciliary genes. Ciliary genes (red) were immunostained with axonemes (acetylated α-tubulin), basal bodies (γ-tubulin), and centrioles (NEK2). Nuclei were stained with DAPI (blue). Mlf1 and Pkhd1l1 localized to cilia axonemes; Sptbn5 localized along the entire cilia; Ppef2 and Tekt3 localized to basal bodies of cilia; and Agbl2 was observed both at the basal body and centriole of cilia. Boxed and color-coded numbers following each gene indicate the number of cardiac defects observed in knockout/mutant mouse hearts: red for OA + VSD, purple for OA + VSD + ASD, blue for MU-VSD, orange for ASD, and green for MU-VSD + ASD. Scale bars, 20 μm. (B) Immunofluorescence staining of cilia in the cardiac OFTs of ciliary gene knockout/mutant mice and control mice at E13.5. Axonemes (acetylated α-tubulin, green) and basal bodies (γ-tubulin, red) were shown. The right column showed higher magnification of the boxed area from the left. Scale bars, 20 and 10 μm. (C) Box plots of cilia prevalence (left) and cilia length (right) in the cardiac OFTs of ciliary gene knockout/mutant mice and control mice at E13.5. (Left): Error bars represent means ± SEM. (Right): The blue boxes showed the lower, median, and upper quartiles of measurements. Each black dot represented the cilia length from a single cell. Homozygous knockout/mutant mice exhibited shorter cilia and a lower cilia percentage compared to wild-type control mice, indicating disrupted ciliogenesis. Statistical significance was assessed by an unpaired two-tailed Student’s t test or Wilcoxon–Mann-Whitney test when appropriate. *P < 0.05, **P < 0.01, ***P < 0.001. n = 10 to 20 OFT sections from five hearts for each group were measured percentage of cilia and cilia length across all OFT sections (n > 400 cilia per group). Correct regulation of cilia length is essential for proper cilium biogenesis and function ([135]30), whereas defects in ciliary axonemes or basal proteins may lead to abnormal cilia length ([136]31). To assess the impact of ciliary gene variants on ciliogenesis during OFT development, we collected E13.5 hearts from wild-type and knockout/mutant mice. Immunofluorescence examination of E13.5 OFTs revealed significantly reduced cilia number and shorter lengths in homozygous mice compared with wild-type littermates (P < 0.05) ([137]Fig. 5, B and C). Ultrastructural analysis using TEM further revealed that Pkhd1l1^−/− mouse OFT tissues displayed severely disorganized ciliary axonemes, with microtubule loss and irregular ciliary distribution and orientation, compared to the typical 9+0 microtubule arrangement in wild-type Pkhd1l1^+/+ mice ([138]Fig. 3, I and J). Thus, these findings highlight the critical role of ciliary genes in ciliogenesis and their essential function located in cardiac OFT development. Cilia orchestrate OFT development through dysregulation of sonic Hh signaling Cilia serve as a transduction center for a series of development-related signaling pathways during cardiovascular development, including Hedgehog (Hh) signaling pathway, WNT signaling pathway, and transforming growth factor–β (TGF-β)/bone morphogenetic protein (BMP) signaling pathway ([139]32). Both abnormalities in cilia structure and changes in the number of cilia may cause alterations in signaling pathways, which in turn lead to a range of developmental disorders. To further explore the shared mechanisms underlying OFT abnormalities caused by ciliary gene knockout or mutation, we carried out whole-transcriptome sequencing (RNA-seq) of E10.5 hearts from wild-type and knockout/mutant littermates of each strain. Pathway enrichment analysis from Kyoto Encyclopedia of Genes and Genomes (KEGG) database indicated differentially expressed genes (DEGs) were associated with cellular communication, including cell adhesion molecules, tight junction, and ECM-receptor interaction, as well as pathways related to cardiac muscle contraction and Hh signaling. Intriguingly, the Hh signaling pathway was the only pathway consistently and significantly enriched across all ciliary gene knockout and mutant strains (FDR < 0.05) ([140]Fig. 6A and fig. S17), providing strong support for its critical role in mediating early cilia-dependent cardiac development. To further investigate the specific cell type responsible for the observed phenotype resulting from cilia abnormalities, we then evaluated the expression of cardiac marker genes in each group. The results showed reduced expression of several myocardial marker genes Tnnt2 and Myl2 in ciliary gene knockout/mutant groups ([141]Fig. 6B), whereas markers of other cardiac cell remained unchanged (fig. S18A). Meanwhile, as shown in [142]Fig. 6 (C and D), a reduced number of cilia was also observed in cardiomyocytes of knockout/mutant mouse OFTs from same-litter crosses of each strain. Then, by integrating scRNA-seq data ([143]Fig. 6E and fig. S18, B and C) and applying gene set variation analysis (GSVA), we further evaluated Hh pathway activity and found lower activities of Hh pathway in cardiomyocytes of ciliary gene knockout/mutant strains ([144]Fig. 6F). In addition, DEGs in these cardiomyocytes were enriched in pathways in heart development, circulatory system development, and cardiac muscle tissue development ([145]Fig. 6G), suggesting the abnormal cardiomyocyte function and cardiac development affected by cilia abnormalities. The activity of Hh signaling pathway is mainly transported downward through the activated membrane protein receptor SMO (Smoothened) and then activates the nuclear transcription factor GLI family (including GLI1, GLI2, and GLI3), which subsequently enters the nucleus and initiates the expression of target genes downstream ([146]33). Western blot assays showed that expression of GLI1 and GLI2 was notably decreased in the hearts of knockout and mutant mice (fig. S18D), confirming the disruption of Hh pathway activity. Fig. 6. Integrated analysis of bulk RNA-seq and scRNA-seq correlated cilia loss with dysregulation of Hh signaling pathway and decreased Gata4 and Nkx2-5 expression. [147]Fig. 6. [148]Open in a new tab (A) The Sankey diagram showing the top 10 enriched pathways in the hearts of homozygous ciliary gene knockout/mutant mice versus littermate controls at E10.5. (B) The expression of myocardial marker genes in E10.5 hearts of wild-type and ciliary gene knockout/mutant groups. Bars represent means ± SEM, with statistical analysis by unpaired two-tailed Student’s t test or Wilcoxon–Mann-Whitney test. *P < 0.05, **P < 0.01, ***P < 0.001. n = 3 to 4 biological replicates per group, with 10 hearts per replicate. (C) Immunofluorescence staining for cilia in cardiomyocytes of wild-type and knockout/mutant mouse OFTs from the same-litter mice, with cardiac troponin t (cTnT) in green and axonemes (acetylated α-tubulin) in red. Scale bars, 200, 20, and 10 μm. n = 3 to 4 replicates per group. (D) Quantification of cilia percentage in cardiomyocytes in (C), presented as means ± SEM. Statistical significance was assessed using unpaired two-tailed Student’s t test or Wilcoxon–Mann-Whitney test. ***P < 0.001. (E) Uniform manifold approximation and projection (UMAP) clustering of cells, showing distinct clusters. (F) The Hh pathway activity in the cardiomyocyte cluster of wild-type and each ciliary gene knockout/mutant group. Statistical analysis was performed using unpaired two-tailed Student’s t test or Wilcoxon–Mann-Whitney test. *P < 0.05, **P < 0.01, ***P < 0.001. (G) GO analyses of the scRNA-seq datasets with DEGs (FDR < 0.05) in cardiomyocytes of knockout/mutant strains versus wild-type OFTs. The top 12 enrichment pathways shown in bubble charts, with cardiac development pathways marked in red. (H) Flow chart for selecting cardiac target genes regulated by cilia-mediated Hh signaling. (I) The down-regulated genes in all cilia knockout/mutant strains expressed by SHF and/or cNCCs. (J) IGV tracks showing Gli1 and Gli2 locations at Gata4 and Nkx2-5 locus from CUT&Tag analysis in E10.5 mouse hearts. The Hh signaling pathway is critical for cardiac OFT development and septation. Several studies have reported that Hh signaling pathway is required for the survival and migration of cNCCs to the OFT cushions and plays an essential role in SHF-mediated OFT elongation and septation after cushion formation ([149]34, [150]35). On the basis of these findings, we hypothesize that ciliary gene mutations impair Hh signaling, affecting gene expression of cardiac progenitor cells in SHF and/or cNCCs, thus contributing to abnormal OFT development. Through literature review, we identified several genes co–down-regulated in all cilia knockout and mutant strains, including SHF-derived genes, namely Foxc2, Gata4, Hopx, Nkx2-5, and Srf ([151]36–[152]38), as well as cNCC-expressed genes like Cdh2, Foxc2, and Vangl1 ([153]39). Besides, we examined the Gli-target genes in heart through performing cleavage under targets and tagmentation (CUT&Tag) experiments on E10.5 mouse hearts using the GLI1 and GLI2 antibodies. Between them, we found two overlapping genes, Gata4 and Nkx2-5 ([154]Fig. 6, H and I), which have also been demonstrated to play important roles in cardiac OFT morphogenesis and development ([155]40, [156]41). Both Gli1 and Gli2 showed notable enriched at Gata4 promoter region (≤1 kb); Gli1 binding was also observed at promoter (≤1 kb) of Nkx2-5; while most Gli2 binding sites were around the intergenic region of Nkx2-5 ([157]Fig. 6J). Motif analysis of Gli1 and Gli2 revealed overrepresentation of transcription factor binding sites in Gata4 and Nkx2-5 regions respectively (P < 0.05) (fig. S18E). Moreover, immunoblotting analysis for GATA4 and NKX2-5 revealed notably reduced protein levels in ciliary gene knockout and mutant hearts (fig. S18F). Importantly, a deficiency in SHF-derived cardiomyocytes addition leads to a shorter OFT, which is consistent with the phenotype we observed in E10.5 mice with ciliary gene knockout/mutation ([158]Fig. 3H and figs. S10 to S14G). Together, our data indicate that ciliary genes knockout/mutation may result in cilia abnormalities in cardiomyocytes, down-regulating the activities of Hh signaling pathway and the expression of the Gli-target genes Gata4 and Nkx2-5, which ultimately contribute to the abnormal OFT development and TOF phenotype. DISCUSSION In this study, we provide a detailed characterization of genetic landscape for nonsyndromic TOF. For known causative TOF genes, we adopted Clinvar and ACMG standards to classify variant pathogenicity and identified 15 P/LP variants in these genes, yielding a 12.21% contribution to TOF incidence (16 of 131 cases). Subsequent association analyses comparing TOF cases with our in-house controls (6004 individuals without TOF) identified cilia-related pathways and ciliary genes enriched with rare LoF variants both at gene set (pathway) level and gene level. Multihit ciliary genes were notably enriched among patients with TOF with LoF variants and putatively detrimental missense variants, with an OR greater than threefold when the number of combined deleterious variants reached six. Through construction of knockout/mutant mouse models, including double-heterozygous models, we further recapitulated the TOF-like phenotypes observed in patients with TOF and explored the potential molecular mechanism. The LoF mouse models showed TOF-like phenotypes, a severe reduction in the number and length of cilia, and disorganized axoneme arrangements in the cardiac OFTs. The incidence of combined OA and VSD was substantially higher in double-heterozygous mice, suggesting a cumulative genetic effect. By integrating RNA-seq and single-cell sequencing data from cardiac OFTs, followed by CUT&Tag analysis in hearts, we speculated that ciliary genes knockout/mutation might down-regulate Gli expression by disrupting Hh signaling pathway mainly in cardiomyocytes. The target genes of Gli (Gata4 and Nkx2-5) in the heart were experimentally verified, which in turn lead to reduced OFT length and ultimately TOF phenotype ([159]Fig. 7). Fig. 7. Proposed mechanism of ciliary gene knockout/mutations leading to tetralogy of Fallot. [160]Fig. 7. [161]Open in a new tab Population-based genetic analyses and mouse models collectively highlight the pathogenic role of ciliary genes in cardiac OFT development. Knockout or mutation of ciliary genes resulted in reduced cilia number and length, as well as disorganized axonemal structure in the cardiac OFT. Mechanistically, ciliary dysfunction inactivated Hh signaling in cardiomyocytes, leading to transcriptional down-regulation of its target genes, Gata4 and Nkx2-5, ultimately disrupting OFT morphogenesis and contributing to the development of TOF. The ethnic differences between European and Asian populations may lead to certain variances in TOF-associated genetic susceptibility. To elucidate the discrepancies between our findings and those of the European TOF cohort, we examined ultrarare variants in NOTCH1 and FLT4, two top TOF-associated mutated genes reported with increased mutational burden in TOF cases of European ancestry from prior WES studies ([162]13). We then examined the overrepresentation of variants in these two genes in our TOF cohort. Among our 131 TOF cases, we detected four patients with ultrarare damaging variants: three in NOTCH1 (2.29%) and one in FLT4 (0.76%), accounting for 3.05% of the TOF cases. In contrast, our control group (n = 6004) harbored 68 variants across 80 individuals, a rate of 1.33%. Thus, our Chinese patients with TOF had a twofold increase in mutational burden compared to controls, although no statistically significant association was detected (OR, 2.332; 95% CI, 0.611 to 6.354; P = 0.1042). Of note, consistent with findings from another Chinese TOF cohort by Tang et al. ([163]42), the proportion of carriers in the Chinese population was considerably lower than that of the European populations (i.e., 4.46% for NOTCH1, 2.53% for FLT4) (fig. S19). Hence, ethnic differences of the population may partly explain the diversity of pathogenic mechanisms of TOF. Previous studies have reported that cilia are closely associated with cardiac development ([164]43). Mouse studies revealed a wide distribution of primary cilia throughout the embryonic mouse heart, among the ventricular surface of trabeculations in both ventricles and were more prominent in the atrial endocardial layer, endocardial cushion mesenchyme, and epicardium ([165]44). A large-scale forward genetic screening study mutagenized with ethylnitrosourea (ENU) constructed a number of mutant mouse models and identified approximately 100 variants in 61 genes induced cardiovascular phenotypes. This included 34 cilia-related genes, 16 genes involved in cilia-transduced cell signaling, and 10 genes regulating vesicular trafficking, a pathway important for ciliogenesis and cell signaling ([166]32). Although this mutagenesis screen provided compelling evidence that cilia may play a central role in CHD pathogenesis, the relevance of this finding to human CHD has yet to be fully clarified. Moreover, the roles of ciliary gene variants contributing to the pathogenesis of TOF have not been studied in a systematic manner, either through population-based studies or through mouse models. In the present study, we systematically screened rare LoF variants as potential pathogenic variants in nonsyndromic TOF. Through gene set–based pathway analysis and gene-based association analysis, we identified notable enrichment of cilia-related pathways and ciliary genes and verified the overall impacts of deleterious ciliary gene variants on TOF. To our knowledge, this study represents comprehensive investigation of ciliary gene involvement in the pathogenesis of nonsyndromic TOF. By applying stringent filtering criteria, we further identified six ciliary genes (AGBL2, MLF1, PKHD1L1, PPEF2, SPTBN5, and TEKT3) with recurrent rare LoF variants enriched in patients with TOF, and conducted corresponding mouse models for subsequent functional validation. Increasing evidence suggests that a large proportion of CHD might be oligogenic or polygenic rather than single genetic inheritance or caused by a regulatory network and shared signaling pathways constructed by interrelated disease-related genes ([167]45), which could account for the reduced penetrance and variable expressivity commonly observed in CHD ([168]46). In support of this, an exome-based case-control analysis of 249 patients supported a complex and heterogeneous genetic architecture of TGA, with 33% of the TGA probands most likely affected by oligogenic or polygenic inheritance ([169]24). Gifford et al. ([170]47) further demonstrated an oligogenic basis for CHD, showing that combined heterozygous mutations in three genes (MKL2, MYH7, and NKX2-5) resulted in left ventricular noncompaction in both humans and mice. In our study, we observed that heterozygous ciliary gene knockout/mutant mice developed mild but discernible cardiac phenotypes, with some strains presenting OA and associated defects such as ASD and MU-VSD, supporting the pathogenic potential of heterozygous variants in patients, albeit with limited penetrance. The relatively low phenotypic incidence implies that these variants may act with moderate effect sizes and are insufficient to drive disease alone in a dominant manner. To further investigate the potential additive effects of multiple deleterious variants, our analysis of the human cohort revealed an elevated TOF risk among individuals with more than two deleterious variants in ciliary genes (OR, 1.672; P = 0.0104). This risk increased further with variant burden, exceeding a threefold rise when six deleterious variants were present, suggesting a cumulative genetic effect ([171]Fig. 2). Complementing our human data findings, we then generated five double-heterozygous mouse lines by pairwise crossing of ciliary gene knockout/mutant strains. Cardiac phenotyping showed a 33.6% overall incidence of cardiac defects, including OA in 11.48% (14 of 122) of cases. Notably, the frequency of OA in double-heterozygous mice was consistently higher than in single-heterozygous strains, and in most cases, also exceeded that observed in homozygous mutants, indicating possible synergistic or cumulative effects between different ciliary gene variants ([172]Fig. 4). Together, our findings from both human genetic analysis and mouse models provide strong evidence for a multigenic inheritance mechanism in TOF, in which combinations of heterozygous variants across multiple ciliary genes contribute to disease risk. The incomplete penetrance observed in heterozygous mice further underscores the importance of genetic interactions and cumulative variant burden in modulating phenotypic expression. Furthermore, beyond the influence of multigenic inheritance pattern, the observation of additional cardiac phenotypes such as MU-VSD and ASD in ciliary gene knockout/mutant mice may reflect the broader role of cilia in cardiac development. Given the widespread distribution of cilia across multiple cardiac regions (fig. S4), their dysfunction may affect not only OFT-specific processes, resulting in features like OA and PM-VSD but also affect other regions involved in septation, contributing to MU-VSD and ASD. These phenotypic variations may also be influenced by shared molecular pathways among different CHD subtypes ([173]13, [174]48), effects of genetic background differences between humans and mice ([175]49, [176]50), or environmental factors during intrauterine and postnatal development. During heart development, cilia serve as transduction centers for various development-related signaling pathways, such as the Hh signaling pathway, WNT/β-catenin signaling pathway, and TGF-β/BMP signaling pathway ([177]51). Previous studies revealed that abnormal expression or localization of ciliary genes affects structural stability and function of cilia ([178]31). To investigate the common mechanisms of cilia in OFT development, we performed RNA-seq of E10.5 cardiac tissues from wild-type and littermate knockout/mutant mice. The DEGs were enriched in pathways related to cell-cell communication (including cell adhesion molecules, tight junctions, and ECM-receptor interactions), cardiac muscle contraction pathway and Hh signaling pathway. Altered activity of cardiomyocyte contraction pathway may indicate abnormal cardiac function in knockout and mutant mice. Cell adhesion molecules are known to mediate adhesion between cells (mostly based on E-cadherin and N-cadherin) and between cells and the ECM (integrins). Among them, E-cadherin modulates OFT development by activating the Wnt/β-catenin signaling pathway and influencing ISL1 expression ([179]52, [180]53), whereas integrins regulate OFT separation mainly by influencing neural crest cell migration, proliferation, and BMP expression ([181]54). Mechanistically, we observed shortened cilia length in all six ciliary gene knockout and mutant mouse models. Complementary RNA-seq and single-cell transcriptomic analyses revealed that ciliary dysfunction specifically disrupts Hh pathway activation in cardiomyocytes, accompanied by reduced expression of the key transcription factors GLI1 and GLI2. Previous studies have shown that mutations in cilia-related proteins might impair Hh signaling and/or Gli processing, leading to severe developmental disorders, while Hh signaling pathway is critical for cardiac OFT development and septation ([182]34, [183]35). Notably, Gata4 and Nkx2.5 emerged as the only two genes both consistently down-regulated across all cilia knockout/mutant strains and identified as direct Gli-target genes in the heart by CUT&Tag profiling. As expected, the protein expression levels of GATA4 and NKX2-5 were prominently decreased in knockout and mutant hearts. GATA4 and Nkx2.5 act as essential cardiogenic transcriptional regulators implicated in many aspects of cardiac development and function. Human genetic studies have implicated haploinsufficiency of GATA4 ([184]55, [185]56) and Nkx2.5 ([186]57, [187]58) in human TOF. Zhou et al. also reported that Gata4 and Gli1 interact genetically, with SHF-specific constitutive Hh signaling remarkably rescued CHDs in Gata4 mutant embryos ([188]59). Sonic Hh (Shh) signaling has previously been shown to regulate OFT alignment by promoting the proliferation of anterior SHF (aSHF) progenitors, facilitating myocardial differentiation, and coordinating endocardial cushion remodeling ([189]34, [190]40, [191]60, [192]61). Shh also maintains Nkx2-5 expression in SHF-derived cardiomyocytes and promotes myocardial migration and ECM remodeling within the OFT cushions, processes essential for OFT elongation and septation. In line with these roles, our data demonstrate that ciliary gene disruption not only leads to impaired Hh-Gli signaling and reduced Gata4/Nkx2-5 expression but also results in shortened OFTs due to reduced SHF-derived cardiomyocyte recruitment at E10.5, closely resembling the phenotype of Shh/Smo mutant embryos. Our findings extend the classical Shh-OFT paradigm by revealing that cilia act as hubs for Hh signal amplification in cardiomyocytes, ensuring Gli-dependent transcriptional activation of Gata4 and Nkx2-5. This unique mechanism explains how subtle perturbations in ciliary function (e.g., mutations) could disrupt OFT alignment independent of canonical Shh ligand availability, a concept not previously emphasized in developmental studies. While prior research has established the essential role of Shh signaling in OFT development, our study uncovers an additional regulatory layer in which ciliary genes modulate Hh signaling efficiency in cardiomyocytes, offering a potential explanation for OFT defects in ciliopathies. Moreover, we provide evidence linking Gli1/2 with SHF-derived Gata4/Nkx2-5 in heart, thus bridging cilia biology with cardiac morphogenesis in a previously unrecognized manner. These insights not only enhance our understanding of OFT development but also underscore the broader importance of ciliary dysfunction in TOF pathogenesis. Although our bioinformatic analyses and mouse models supported the pathogenic role of ciliary gene variants in nonsyndromic TOF, this study was restricted by several shortcomings. Through our screening procedure and enrichment analysis, we found 108 ciliary genes with rare LoF variants in our patients with TOF. However, because of the polygenic genetic architecture and low penetrance of nonsyndromic TOF, it was impossible to construct mouse models for each candidate ciliary gene to verify its pathogenic role in TOF. We therefore selected six previously unidentified genes (namely AGBL2, MLF1, PKHD1L1, PPEF2, SPTBN5, and TEKT3) with recurrent mutations for mouse models construction and phenotype validation, both to illustrate the reliability of our screening strategy and to provide in vivo evidence supporting the contribution of ciliary gene variants to TOF pathogenesis. Besides, because of the incomplete penetrance of cardiac phenotypes in mouse models, we used homozygous mice for subsequent mechanistic studies, as they offer an advantage over heterozygous mice in exploring the potential downstream mechanisms of ciliary gene mutations affecting TOF pathogenesis. In addition, while ciliary dysfunction is known to contribute to a spectrum of systemic disorders, this study focused primarily on cardiac phenotypes and left-right patterning anomalies (no lateral defects or visceral heterotaxy were observed in our models). Whether ciliary genes deficiency caused additional abnormal phenotypes in other tissues needs to be further studied. Last, this study may still suffer from its relatively small sample size due to the challenges in obtaining TOF cases, and the absence of parental DNA samples precluded the determination of whether the variants were de novo or inherited from the unaffected parents. Furthermore, because of ethical and practical constraints and given the difficulty of obtaining cardiac tissue from living individuals, genetic analyses were based on blood samples rather than cardiac tissues. However, as blood may not fully reflect the genetic landscape of native cardiomyocytes, future studies incorporating cardiac tissue from patients with TOF and matched healthy controls will be valuable for uncovering cardiac-specific variants and further elucidating the molecular mechanisms underlying TOF. In summary, nonsyndromic TOF is a heterogeneous disease with complex and diverse etiologies. Our data from next-generation sequencing in 131 patients systematically identified a higher burden of cilium pathway and ciliary gene deleterious variants among individuals with TOF and also suggested a multigenic inheritance pattern of ciliary gene variants on TOF. Functional studies using mouse models harboring these variants recapitulated TOF-like phenotypes as well as demonstrated impaired cilia structure and function. Notably, double-heterozygous mice exhibited a higher incidence of OA than single-heterozygous mutants, further supporting the additive effects of multiple ciliary gene variants. Together, our findings highlight the essential role of cilia in cardiac OFT development and provide valuable genetic and mechanistic insights into the etiology and pathogenesis of nonsyndromic TOF. Expanded genetic screening of ciliary genes in larger TOF cohorts will be valuable for advancing both diagnosis and precision medicine in this population. MATERIALS AND METHODS Human sample collection Patients This study enrolled 131 patients who received a confirmed diagnosis of nonsyndromic TOF from the First Affiliated Hospital of Nanjing Medical University (Nanjing, China) and the Affiliated Children’s Hospital of Nanjing Medical University (Nanjing, China) from 2008 to 2014. The nonsyndromic TOF samples were diagnosed consistently by pediatric cardiologists through comprehensive physical examination, echocardiography, and in some cases, further confirmed by surgery. Patients were excluded if they showed (i) any extracardiac malformation or other types of developmental abnormalities; (ii) chromosomal abnormalities; (iii) a positive family history of CHD in their first-degree relatives (parent or sibling); (iv) a history of maternal diabetes mellitus, phenylketonuria, teratogen exposure, or therapeutic drug exposure during pregnancy. For each participant, 5 ml of the whole blood were collected for DNA extraction and genomic analyses. Controls For association tests, we applied WGS data of 6004 unrelated individuals with noncardiac defects from our Nanjing Lung Cancer Cohort project ([193]16) as population genome controls in this study, for comparison of gene-based and gene set–based variant frequencies. This project was approved by the Human Ethics Committee of Nanjing Medical University. Notably, the control group consisted of adults rather than children, which was a deliberate choice to mitigate the potential underdiagnosis of CHD in younger populations and to reduce selection bias in the sample. The sequencing data were processed using the same data processing pipeline, and variant annotation were performed in a unified manner across case and control samples to minimize potential systematic biases due to technical factors or analytical methods. Methods and experimental protocols involving human subjects were approved by the institutional review boards of Nanjing Medical University (2022SCJ2325). All participants completed written informed consent form before taking part in this research, and all investigations conformed to the principles outlined in the Helsinki Declaration. Animal studies All animal experiments were performed under Guidelines for the Care and Use of Laboratory Animals and were approved by the Institutional Animal Care and Ethical Committee of Nanjing Medical University (IACUC-1912049). Mice (C57BL/6J) were housed at a constant temperature of 23° ± 2°C, humidity of 55 ± 5%, and under a regular 12-hour light-dark cycle in the Animal Research Center of Nanjing Medical University. A vaginal plug was checked on the morning following mating with an experienced male, and gestational ages were determined by visualizing the presence of a vaginal plug (E0.5 = vaginal plug day). After mating, pregnant females were individually housed. Before cardiac resection, the mice were anesthetized through intraperitoneal administration of tribromoethanol (Nanjing Aibei Biotechnology Co. LTD, M2920, 0.02 ml/g per mouse), a noncontrolled fast-acting anesthetic specifically used for mice, which also allowed for precise dosage control. Afterward, the mice were euthanized by cervical dislocation and the hearts or embryonic hearts were harvested using a stereomicroscope for subsequent experiments. All measurements, including PA widths, OFT lengths, cilia lengths and proportions, along with histological analysis and CHD diagnosis, were conducted by two independent individuals blinded to the mouse genotype. In cases of any discrepancies, a third reviewer reevaluated and finalized the measurements. Next-generation sequencing WES was performed on 104 patients with nonsyndromic TOF and WGS was performed on 27 patients with nonsyndromic TOF. Genomic DNA was isolated from blood samples of the subjects using a whole-blood DNA purification kit (Qiagen, Hilden, Germany). For WES, exome capture was carried out using the Agilent SureSelect Human All Exon V6 Library Prep Kit (Agilent, CA, USA), followed by SureSelectXT Target Enrichment System for library preparation (Agilent, CA, USA). For WGS, libraries were constructed using the standard protocols provided by the manufacturer for NEBNext DNA Sample Prep Master Mix Set (NEB, MA, USA). All libraries were sequenced with a fragment of 2 × 150–base pair (bp) (PE 150) using Illumina HiSeq X Ten sequencers to a depth of 100× (WES) and 30× (WGS) base coverage. We used CASAVA (v1.8.2) to convert Illumina BCL files to FASTQ files. Then, adapters and low-quality bases were trimmed with Trimmomatic (v0.32) ([194]62). After that, sequence reads were mapped to the GRCh37 human reference sequence using the Burrows-Wheeler Aligner (BWA-MEM v0.7.15-r1140) ([195]63) with the default parameters, and the duplicates were marked and discarded using SAMBLASTER (v0.1.24) ([196]64). The quality metrics (including coverage, median insert size, and percentage of chimeric reads) and contamination estimation of “Bam” files were obtained by Picard (v1.70) and VerifyBamID2 (v1.04), respectively. Haplotype calling was performed using HaplotypeCaller in GATK (v3.8) in GVCF mode according to the best practice ([197]65). Individual gVCF files were jointly genotyped for high-confidence alleles using Genotype GVCFs for single-nucleotide polymorphisms (SNPs) and small insertions and deletions (INDELs) in the coding sequences. We performed relatedness and sex checking based on a temporary list of high-confidence variants: biallelic, high-call rate (>0.99), and common SNPs (allele frequency > 1%). We estimated pairwise relatedness by PLINK (v1.9) ([198]66) and inferred the chromosomal sex of the samples based on the inbreeding coefficient (F) for common variants on chromosome X, excluding pseudo-autosomal regions. Quality control of sequencing data Sample quality control We excluded individuals with (i) median insert size <250 bp, (ii) excess chimeric reads: pct_chimera >0.05, (iii) high contamination: freemix >0.05, (iv) unmatched relatedness, and (v) inconsistent chromosomal sex and questionnaire information. None of the patients with TOF were excluded according to the above criteria, and we finally included all of the 131 nonsyndromic TOF cases for further analyses. Variant quality control GATK variant quality score recalibration (VQSR) was used to filter variants. The SNP VQSR model was trained using HapMap 3.3 and 1000 Genomes Omni 2.5 SNP sites, and a 99.6% sensitivity threshold was applied to filter variants. We excluded variants if they (i) were on sex chromosomes, (ii) failed to pass VQSR filtering criteria, (iii) had inbreeding coefficients <−0.3, (vi) had proportions of samples with high genotype quality (≥20) ≤0.8, (v) had proportions of samples with enough sequencing depth (≥10) ≤0.8, (vi) had call rates ≤0.95, or (vii) indels in segment duplication or simple sequence repeat regions. Variants annotation and identification of potential pathogenic variants in nonsyndromic TOF Frequency annotation and the definition of rare variants Variant allele frequencies were obtained from the gnomAD (v2.1.1) and the ExAC (v0.3.1), both downloaded from [199]https://gnomad.broadinstitute.org/downloads, as well as from our in-house reference dataset comprising 6004 individuals without CHDs. These frequency data were annotated to the variants identified in this study using vcfanno (v0.3.2) ([200]67). Variants with estimated MAF_all <0.5% in our study cohorts (mutation rate of patients with TOF plus control group) and <0.5% in both of the populations [ALL and East Asian (EAS)] of gnomAD and ExAC were used for the following analyses. Definition of potential pathogenic variants of TOF: LoF variants (frameshift, stop gain, and splice acceptor/donor) All coding variants were annotated with SnpEff version 4.3-3 and then loaded into the GEMINI software version 0.19.1. For gene set–based analyses, all genes annotated to the potential pathogenic variants of TOF were used for the following pathway enrichment analysis based on GO databases by the ToppGene Suite ([201]https://toppgene.cchmc.org/). Generation of gene-targeted mouse models Ciliary gene knockout and mutant mice were generated by the CRISPR-Cas9 genome editing system. The single guide RNA (sgRNA) sequence derived from each gene (GRCm38/mm10) was designed using CRISPR Design ([202]https://crispor.gi.ucsc.edu/), and a DNA oligo corresponding to this sequence was cloned into PGL3 for guide RNA in vitro transcription. Two single-stranded DNA (ssDNA) homology-directed repair (HDR) templates were designed for anthropomorphic variants in Mlf1 and Ppef2. A system pool of Cas9 mRNA (25 ng/μl), sgRNA1 (15 ng/μl), and sgRNA2 (15 ng/μl) were mixed and microinjected into single-cell mouse embryos of the C57BL/6 strain (Agbl2, Pkhd1l1, Sptbn5, and Tekt3); a pooled system of Cas9 mRNA (20 ng/μl), sgRNA (20 ng/μl), and ssDNA (1 μM) conducted the similar experiment as above (Mlf1 and Ppef2). The edited sites were amplified by polymerase chain reaction (PCR) and Sanger sequencing in the founders, and the founders were followed by at least three consecutive backcrosses with congenic mice (C57BL/6) to obtain a pure background. All sgRNAs and primer sequences for genotyping for each knockout/mutant mouse line are listed in table S4. Off-target analysis The potential OTSs of the sgRNA were predicted with the online CRISPR Design Tool ([203]https://cm.jefferson.edu/Off-Spotter/). The top five OTSs for each sgRNA were selected, and then the genomic regions covering the OTSs were PCR amplified using the primers listed in table S6. Immunofluorescence staining To visualize OFT cilia, wild-type and knockout/mutant embryonic hearts were harvested at E10.5, E12.5, E14.5, E16.5, and E18.5. Hearts were fixed in 4% buffered paraformaldehyde (PFA) (Biosharp, BL539A) on ice for 2 hours, rinsed in phosphate-buffered saline (PBS) and incubated in 20 and 30% sucrose overnight at 4°C until the hearts sunk completely. Then, the tissues were embedded in optimal cutting temperature compound and stored at −80°C. Cryosections of 15-μm thickness were collected on positively charged slides and stored at −20°C. For immunofluorescence staining, the hearts were washed in PBS and blocked with donkey serum for 1 hour at room temperature before being incubated overnight at 4°C with primary antibodies. After three washes with PBS for 5 min, the samples were stained for 1 hour at room temperature with secondary antibodies followed by 4′,6-diamidino-2-phenylindole (DAPI) staining for nuclei visualization. After a second series of washes, the slides were mounted in Fluoromount-G (Southern Biotech, 0100-01). Images were acquired by a confocal laser scanning microscope (Zeiss, LSM800). The antibodies and dilutions were as follows: acetylated tubulin (Sigma-Aldrich, T-6793, RRID:AB_477585, 1:1000 dilution), acetyl–α-tubulin (Cell Signaling Technology, D20G3, RRID:AB_10544694, 1:1000 dilution), γ-tubulin (Abcam, ab11317, RRID:AB_297921, 1:1000 dilution), γ-tubulin (Sigma-Aldrich, T6557, RRID:AB_477584, 1:1000 dilution), RSPH9 (Proteintech, 23253-1-AP, RRID:AB_2879240, 1:200 dilution), cardiac troponin t (Abcam, ab8295, RRID:AB_306445, 1:200 dilution), donkey anti-mouse immunoglobulin G (IgG) Alexa Fluor 488 (Thermo Fisher Scientific, A21202, RRID:AB_141607, 1:1000), and donkey anti-rabbit IgG Alexa Fluor 546 (Thermo Fisher Scientific, A10040, RRID:AB_2534016, 1:1000). Transmission electron tomography For TEM, embryonic mouse hearts (E13.5) were excised after cervical dislocation and cardiac OFTs were dissected and isolated under a stereomicroscope. Then, OFTs were fixed at 4°C overnight with 2.5% glutaraldehyde in 0.2 M cacodylate buffer. After washing in cacodylate buffer, the OFTs were postfixed in 1% OsO[4] in 0.2 M cacodylate buffer for 2 hours at 4°C. Next, the OFTs were washed and poststained with 2% aqueous uranyl acetate for 2 hours, dehydrated in a graded ethanol series, immersed in acetone and embedded in resin (EPON, Low Viscosity Embedding Media Spurr’s Kit #14300). Ultrathin sections (60 to 80 nm) were cut on an ultramicrotome (Leica, EM UC7) and mounted on copper grids. The sections were then stained in a 2% uranyl acetate saturated alcohol solution and lead citrate for 10 min. Images were acquired using an FEI transmission electron microscope (FEI, Tecnai G2 Spirit Bio TWIN). Protein extraction and Western blot analysis Embryonic mouse heart (E10.5) tissues were collected and lysed using universal protein extraction lysis buffer radioimmunoprecipitation assay (Beyotime, P0013B) containing a protease inhibitor cocktail (MedChemExpress, HY-K0010). The protein lysates were centrifuged and the supernatants were quantified using the BCA protein assay kit (Beyotime, P0012). Then, the lysates were denatured at 100°C for 10 min with 5× SDS–polyacrylamide gel electrophoresis sample buffer (Beyotime, P0015L). For Western blotting, 40 μg of protein was separated on 8% or 10% SurePAGE according to the molecular weight of the protein (GenScript, M00661/M00666) and transferred onto a polyvinylidene difluoride membrane (Millipore, ISEQ00010). The antibodies used in Western blotting are as follows: AGBL2 (Invitrogen, PA5-101547, RRID:AB_2850981, 1:1000 dilution), MLF1 (Santa Cruz Biotechnology, sc-514294, 1:500 dilution), PPEF2 (Abcam, ab183667, 1:1000 dilution), TEKT3 (Invitrogen, PA5-70344, RRID:AB_2690235, 1:1000 dilution), GLI1 (Proteintech, 66905-1-Ig, RRID:AB_2882232, 1:1000 dilution), GLI2 (R&D Systems, AF3635, RRID:AB_2111902, 1:1000 dilution), GATA4 (Santa Cruz Biotechnology, sc-25310, RRID:AB_627667, 1:500 dilution), NKX2-5 (Abcam, ab97355, RRID:AB_10680260, 1:1000 dilution), and GAPDH (glyceraldehyde-3-phosphate dehydrogenase; Cell Signaling Technology, 5174, RRID:AB_10622025, 1:2000 dilution). Knockout/mutation of AGBL2, MLF1, PPEF2 and TEKT3 were verified by Western blotting, whereas validation of PKHD1L1 or SPTBN5 knockout in mice was performed using immunofluorescence with a PKHD1L1 (Abnova, K7241-1F5, 1:1000 dilution) or SPTBN5 antibody (Abcam, ab111223, RRID:AB_10864996, 1:1000 dilution) respectively, as described above. Histological analysis of mouse hearts For histological examination, the hearts of wild-type and knockout/mutant newborns at P1 were harvested and fixed overnight in 4% PFA (Biosharp, BL539A) and then dehydrated in a graded ethanol series, cleared, and paraffin wax embedded. Transverse sections (6 μm) were stained with H&E using standard histology techniques. The stained hearts were photographed with a stereomicroscope (Nikon, SMZ1270). MEF isolation and location analysis Primary MEFs were isolated from E13.5 embryos. Briefly, embryos were minced using a sterile razor blade. Then, 4 ml of 0.25% trypsin-EDTA (Gibco, 25200-056) was added, and the samples were incubated at 37°C for 15 min. After incubation for 15 min in 10-cm tissue culture dishes, the remaining fragments were pipetted multiple times to dissociate the cells and incubated overnight in Dulbecco’s modified Eagle’s medium/Nutrient mixture F-12 (DMEM/F-12; Gibco, C11330500BT) supplemented with 10% fetal calf serum (Gibco, 10099141C). For primary cilia analysis, MEFs were serum starved in DMEM/F-12 supplemented with 0.1% serum for 24 hours to induce cilia growth. Cells were fixed with 4% PFA in PBS and immunostained with antibodies against AGBL2 (Invitrogen, PA5-101547, RRID:AB_2850981, 1:200 dilution), MLF1 (Santa Cruz Biotechnology, sc-514294, 1:200 dilution), PPEF2 (Abcam, ab183667, 1:200 dilution), PKHD1L1 (Abnova, K7241-1F5, 1:200 dilution), SPTBN5 (Abcam, ab111223, RRID:AB_10864996, 1:200 dilution), and TEKT3 (Invitrogen, PA5-70344, RRID:AB_2690235, 1:200 dilution), together with structural markers acetylated tubulin (Sigma-Aldrich, T-6793, RRID:AB_477585, 1:1000 dilution), acetyl–α-tubulin (Cell Signaling Technology, D20G3, RRID:AB_10544694, 1:1000 dilution), γ-tubulin (Abcam, ab11317, RRID:AB_297921, 1:1000 dilution), γ-tubulin (Sigma-Aldrich, T6557, RRID:AB_477584, 1:1000 dilution), and Nek2 (Santa Cruz Biotechnology, sc-55601, RRID:AB_1126558, 1:200 dilution). The cells were then imaged using a confocal laser scanning microscope (Zeiss, LSM800). Quantification of cilia The percentage of ciliated cells and the length of cilia were quantified and measured on 15-μm-thick heart sections using ZEN software. Z-stacks were set by finding the highest and lowest depths (with 63× objectives) with visible fluorescence and using the system optimized setting to determine steps. Z-stacks were then compiled to form maximum projection images. Three-dimensional reconstructions of these images were performed by importing Z-stack confocal images into Imaris 9.0 (Bitplane Inc.) and creating surface renderings based on stain intensities. Cilia length was measured from the base of the axoneme (acetylated tubulin–positive staining) to the tip. The percentage of ciliated cells was calculated as the number of cilia (acetylated tubulin and γ-tubulin–positive staining)/number of DAPI-labeled nuclei for each field. RNA-seq and data analysis Total RNA from the heart tissues of ciliary gene knockout/mutant mice and wild-type littermate mice at E10.5 was extracted with TRIzol X-100 (Thermo Fisher Scientific, 15596018) and a total amount of 2 μg of RNA per sample was used for RNA library construction. RNA integrity and concentrations were assessed using the RNA Nano 6000 Assay Kit of the Bioanalyzer 2100 system (Agilent Technologies). Sequencing libraries were then generated using the NEBNext Ultra RNA Library Prep Kit (Illumina, E7530). mRNA was purified from total RNA using poly-T oligo–attached magnetic beads and cut into small fragments for cDNA synthesis. After adenylation of the 3′ ends of DNA fragments, adaptors with hairpin loop structures were ligated, and then PCR was performed. High-throughput RNA sequencing was carried out using Illumina NovaSeq6000 platform and 150-bp paired-end reads were generated. Raw data were filtered and processed to obtain clean data with high quality and aligned to the mouse reference genome GRCm38/mm10 with STAR software (v2.5.3a). Then, assembly and quantification of the transcripts were accomplished with RESM software (v1.3.3) guided by the Ensembl gtf gene annotation file ([204]https://ftp.ensembl.org/pub/release-93/gtf/mus_musculus/). Read counts were used for the measurements of the relative abundance of the transcripts. Fold change and P values were calculated on the basis of read counts using edgeR package (v3.36.0), and DEGs were identified (FDR < 0.05). KEGG pathway enrichment analysis was performed on the basis of the DEGs by the ToppGene Suite ([205]https://toppgene.cchmc.org/). scRNA-seq and data analysis E10.5 wild-type and homozygous ciliary gene knockout/mutant mouse embryos were isolated and dissected for cardiac OFTs. The obtained tissues were washed for two to three times with Dulbecco’s phosphate-buffered saline (DPBS) precooled at 4°C and then was transferred to a 2-ml centrifuge tube with tissue storage buffer (Miltenyi Biotec, #130-100-008). The tube was centrifuged at 50g for 1 min at 4°C, after which the tissue storage buffer was discarded, and 1 ml of prewarmed digestion solution (Miltenyi Biotec, #130-110-203; 37°C) was added to the OFT tissue. The tissues were incubated in a 37°C water bath and gently pipetted every 5 min, for a total digestion time of approximately 15 min. Enzymatic digestion was terminated by adding 10% fetal bovine serum (Pricella, #164210-50), and the cell suspension was filtered through a 40-μm cell strainer (Corning, #CLS431750). The filtered suspension was centrifuged at 300g for 3 min at 4°C, and the pellets were resuspended in PBS containing 0.01% bovine serum albumin (Sigma-Aldrich, #A1933). After treating with AO/PI double fluorescent dye, cell quality was conducted on Countstar Rigel S2 to determine cell concentration, cell viability, and cell clump. At the same time, Trypan blue staining was performed to observe the cell morphology, cell concentration, cell fragments and impurities. Single-cell encapsulation was performed using the Chip A Single Cell Kit v2.0 (MobiDrop, #S050100201) on the MobiNova-100 platform (MobiDrop, #A1A40001). Each droplet contained one cell and a gel bead carrying millions of oligonucleotides with unique cell barcodes. Droplets were then processed with MobiNovaSP-100 (MobiDrop, #A2A40001), which initiated oligo release and hybridization with cellular mRNAs. Reverse transcription and cDNA amplification were performed within droplets. Libraries were constructed using the High-Throughput Single Cell 3′RNA-Seq Kit v2.0 (MobiDrop, #S050200201) and indexed using the 3′ Single Index Kit (MobiDrop, #S050300201). Sequencing was carried out on an Illumina NovaSeq 6000 platform using 150-bp paired-end reads. Raw sequencing data (FASTQ files) were demultiplexed and aligned to the mouse reference genome (GRCm38/mm10) using Cell Ranger (v6.1.1) pipeline with default parameters, generating a feature-barcode expression matrix. Downstream analysis was performed in R using the Seurat package (v4.1.3). Genes expressed in fewer than three cells, as well as cells with <200 genes or >10% mitochondrial gene content, were excluded. Doublets were identified and removed using the DoubletFinder package (v2.0.3). Batch correction was conducted using the Harmony package (v0.1.1) to minimize potential sample-specific effects. Eleven significant principal components were selected on the basis of ElbowPlot and JackStrawPlot outputs in Seurat. Cell clustering and dimensionality reduction were performed using the Seurat FindClusters and RunUMAP functions, respectively. Cluster-specific marker genes were identified using the FindAllMarkers function. Cell types were annotated on the basis of known marker gene expression. Gene set variation analysis GSVA was performed to evaluate Hh signaling pathway activity in cardiomyocytes from wild-type and ciliary gene knockout/mutant mice ([206]68). A curated mouse gene set for Hh signaling ([207]HALLMARK_HEDGEHOG_SIGNALING.v2023.1.Mm) was downloaded from the GSEA database. This gene set includes representative components of the Hh pathway such as GLI family genes, Shh, Gli1, Ptch1, and others directly related to Hh signaling. GSVA was conducted using the “GSVA” R package (version 1.40.0) with default parameters. Enrichment scores were calculated for each cardiomyocyte to represent the relative activity level of the Hh signaling pathway based on gene expression. Cleavage under targets and tagmentation CUT&Tag assays and subsequent high-throughput sequencing were performed by Seqhealth Technology Co. Ltd. (Wuhan, China). For each group, 10 fresh E10.5 hearts from wild-type mouse embryos were collected and homogenized to isolate cells, which were then incubated with concanavalin A–coated magnetic beads. The cells were subsequently incubated with primary antibodies against GLI1 and GLI2 (GLI1: R&D Systems, AF3324, RRID:AB_663902; GLI2: R&D Systems, AF3526, RRID:AB_2279108) for 2 hours, followed by incubation with a secondary antibody for 1 hour at room temperature. Hyperactive pG-Tn5 transposase was added to initiate tagmentation. After terminating the reaction, DNA fragments were purified using PCI extraction and amplified via PCR with indexed P5 and P7 primers. Final libraries were enriched, quantified, and sequenced on an Illumina NovaSeq 6000 platform using a PE150 configuration. Raw sequencing data were processed using Fastp (v0.23.1) for quality filtering and adapter trimming. Clean reads were aligned to the GRCm38/mm10 mouse reference genome using Bowtie2 (v2.2.6) with default settings. Sambamba (v0.7.1) was used to convert file formats and remove PCR duplicates. DeepTools (v2.4.1) was used to visualize read distributions around transcription start sites. Peak calling was conducted using MACS2 (v2.2.7.1), and peak annotation and distribution analysis were performed using bedtools (v2.30.0). Motif analysis was carried out using HOMER (v4.10). Last, transcription factor binding sites were predicted on the basis of enriched peak sequences and motif data from the JASPAR database. Statistical analysis Gene-based association analyses were performed by aggregating the qualifying rare deleterious variants into genes and comparing allele counts between TOF cases and controls using two-sided Fisher’s exact tests. Benjamini-Hochberg correction was applied, with an FDR threshold of <0.2. Input data consisted of rare, predicted deleterious variants (e.g., LoF and detrimental missense variants) and filtered on the basis of MAF thresholds (<0.005) and functional annotations. The LoF model focused on LoF variants, defined as frameshift, stop-gain, and canonical splice site (splice acceptor/donor) variants. The detrimental missense variant model applied four prediction criteria: (i) predicted as pathogenic by AlphaMissense (score, > 0.564) ([208]69); (ii) CADD score, >20 ([209]70); (iii) predicted as deleterious by SIFT (score, ≤ 0.05) ([210]71); and (iv) predicted as probably_damaging (score, 0.447 to 0.909) or possibly_damaging (score, 0.909 to 1) by PolyPhen2 ([211]72). Each criterion was applied independently to identify genes enriched for detrimental missense variants in TOF cases. Gene set–based association analyses were conducted for LoF variants and detrimental missense variants in all ciliary genes involved in cilium pathway between TOF cases and controls using two-sided Fisher’s exact test. Missense variants were considered deleterious only if all four criteria were simultaneously met to ensure high-confidence classification in oligogenic burden assessment. ORs were calculated to evaluate the burden of varying numbers of deleterious variants in ciliary genes among patients with TOF compared to control individuals, with statistical significance defined as P < 0.05 and OR > 1. For multigenic burden analysis, variant counts per individual were based on the number of unique affected genes. Multiple variants within the same gene were consolidated and counted once, whereas variants in different genes were counted individually on the basis of their actual occurrence. This approach ensured that the per-sample mutation burden reflected the number of distinct genes affected. For in vitro and in vivo experiments, the results were presented as the means ± SEM. Statistical calculations were performed using unpaired two-tailed Student’s t test or Wilcoxon–Mann-Whitney test for continuous variables data and Pearson’s Chi-squared test or Fisher’s exact test for categorical data, as appropriate. A value of P < 0.05 (*) was considered statistically significant, with **P < 0.01 and ***P < 0.001. Acknowledgments