Next Article in Journal
Stromal Vascular Fraction Cells from Individuals Who Have Previously Undergone Radiotherapy Retain Their Pro-Wound Healing Properties
Previous Article in Journal
The Short-Term Results of Autologous Platelet-Rich Plasma as an Adjuvant to Re-Intervention in the Treatment of Refractory Full-Thickness Macular Holes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Identification of Novel Risk Variants of Non-Syndromic Cleft Palate by Targeted Gene Panel Sequencing

by
Justyna Dąbrowska
1,
Barbara Biedziak
2,
Agnieszka Bogdanowicz
2 and
Adrianna Mostowska
1,*
1
Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 6 Swiecickiego Street, 60-781 Poznan, Poland
2
Department of Orthodontics and Craniofacial Anomalies, Poznan University of Medical Sciences, 60-812 Poznan, Poland
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(5), 2051; https://doi.org/10.3390/jcm12052051
Submission received: 17 December 2022 / Revised: 1 March 2023 / Accepted: 2 March 2023 / Published: 4 March 2023
(This article belongs to the Section Dentistry, Oral Surgery and Oral Medicine)

Abstract

:
Non-syndromic cleft palate (ns-CP) has a genetically heterogeneous aetiology. Numerous studies have suggested a crucial role of rare coding variants in characterizing the unrevealed component of genetic variation in ns-CP called the “missing heritability”. Therefore, this study aimed to detect low-frequency variants that are implicated in ns-CP aetiology in the Polish population. For this purpose, coding regions of 423 genes associated with orofacial cleft anomalies and/or involved with facial development were screened in 38 ns-CP patients using the next-generation sequencing technology. After multistage selection and prioritisation, eight novel and four known rare variants that may influence an individual’s risk of ns-CP were identified. Among detected alternations, seven were located in novel candidate genes for ns-CP, including COL17A1 (c.2435-1G>A), DLG1 (c.1586G>C, p.Glu562Asp), NHS (c.568G>C, p.Val190Leu—de novo variant), NOTCH2 (c.1997A>G, p.Tyr666Cys), TBX18 (c.647A>T, p.His225Leu), VAX1 (c.400G>A, p.Ala134Thr) and WNT5B (c.716G>T, p.Arg239Leu). The remaining risk variants were identified within genes previously linked to ns-CP, confirming their contribution to this anomaly. This list included ARHGAP29 (c.1706G>A, p.Arg569Gln), FLNB (c.3605A>G, Tyr1202Cys), IRF6 (224A>G, p.Asp75Gly—de novo variant), LRP6 (c.481C>A, p.Pro161Thr) and TP63 (c.353A>T, p.Asn118Ile). In summary, this study provides further insights into the genetic components contributing to ns-CP aetiology and identifies novel susceptibility genes for this craniofacial anomaly.

1. Introduction

Orofacial clefts (OFCs) are one of the most common congenital malformations, affecting 1 in 700 live-born children worldwide [1]. Treatment of those developmental anomalies requires long-term, multi-specialist care, including craniofacial and dental corrective surgeries, speech and hearing therapy, and psychosocial support [2,3]. Moreover, the occurrence of OFCs is frequently associated with an increased risk of various cancers [2,4].
OFCs are divided into two primary forms: cleft lip with or without cleft palate (CL/P) and cleft palate (CP). Interestingly, the embryological and epidemiological data suggest a distinct developmental mechanism and genetic aetiology of these two subtypes [5,6,7,8,9]. Globally, CP occurs in 6 per 10,000 live births [10]. Approximately 50% of all CP cases are non-syndromic (ns-CP) [8], appearing in isolation with no other structural or cognitive anomalies and are aetiologically different from those occurring with cleft syndromes [9].
To date, multiple analyses have established the crucial role of genetic components in the complex aetiology of ns-CP [9,11,12], but the genetic basis of ns-CP has not been fully elucidated. The encountered issues are related to the considerable contribution of environmental factors to the ns-CP aetiology and the diversity in susceptibility loci and variants depending on the racial background, which suggests that the genetic architecture of ns-CP may vary between different populations [11,13,14,15].
Numerous studies, including genome-wide association studies (GWASs) and meta-analyses conducted in different populations, identified only several significant risk loci for ns-CP [15,16,17,18,19]. The strongest signal within detected chromosomal regions was located in the GRHL3 gene (OMIM * 608317), encoding a Grainyhead-like transcriptional factor that stimulates the migration of endothelial cells during embryonic palate development. The most significant GRHL3 variant that reached the genome-wide significance level was a deleterious transition c.1361C>T, leading to a threonine to methionine substitution at position 454 (rs41268753) [16]. The pathogenic variants of GRHL3 have also been reported to cause Van der Woude syndrome (OMIM #119300), the most common disorder associated with orofacial clefts [20]. Interestingly, the GWASs revealed that the top variants that reached the genome-wide significance threshold had a relatively low frequency in the studied populations [14,21], suggesting that rare functional nucleotide alternations might be more relevant in understanding the molecular background of ns-CP than common nucleotide changes [22,23]. Therefore, several previous studies applied next-generation sequencing (NGS) to identify rare coding variants that may be involved in the aetiology of this structural anomaly. The deleterious alternations were mainly detected within known candidate genes and genes highly expressed in mouse facial primordia [24,25,26,27,28,29].
Therefore, this study aimed to identify low-frequency functional variants that may contribute to ns-CP aetiology. For this purpose, the coding regions of 423 genes associated with orofacial clefts and/or encoding proteins involved in facial development were analysed using the NGS-based multi-gene panel in 38 ns-CP patients from the Polish population.

2. Materials and Methods

2.1. Study Population

The participants were recruited from the genetically homogenous Polish population through The Comprehensive Therapy Program for Craniofacial Anomalies at Poznan University of Medical Sciences. The study consisted of 38 ns-CP patients with no other structural or cognitive anomalies, of which, in 8 ns-CP cases (21.05%), dental anomalies were reported (Table 1). However, since these are expected consequences associated with cleft palate, they were not excluded from the analysis. A total of 35 recruited patients (92.11%) had a cleft of the hard and soft palate, while, in 3 cases (7.89%), the cleft affected only the soft palate. Only 4 participants (10.53%) had a family history of ns-CP. Peripheral blood samples from ns-CP patients and their parents, that is, 34 complete triads (89.47%) and 4 dyads (10.52%), were collected to isolate genomic DNA by salt extraction. This study was conducted according to the Declaration of Helsinki [30] and was approved by the Ethics Committee of the Poznan University of Medical Science (no 1115/18). All participants or their legal guardians provided written informed consent.

2.2. The NGS-Based Multi-Gene Panel

The whole coding sequence and flanking regions [exon-intron junction (+/−55 bp)] of 423 genes were tested to identify rare risk variants associated with ns-CP. The multi-gene panel was previously designed to detect nucleotide changes involved in the aetiology of orofacial dysmorphologies, including non-syndromic cleft lip with or without cleft palate (ns-CL/P) and non-syndromic tooth agenesis (ns-TA); please see our previous studies [31,32] for the detailed methodology and strategy for selecting the genes. In brief, the panel consisted of genes with previously documented association with ns-OFCs, those linked to cleft syndromes and/or dental anomalies, for which knockout mice models display OFCs and/or dental anomalies, are highly expressed in craniofacial regions during embryonic development, encode proteins engaged in signalling pathways during facial development and those involved in DNA repair mechanisms. The characteristics of 423 genes analysed are presented in Tables S1 and S2. Based on the provided chromosomal coordinates, the set of DNA probes complementary to the target regions (SeqCap® EZ Prime Choice, Roche Sequencing Solutions, CA, USA) was designed using the NimbleDesign® online tool (Roche, WI, USA). The cumulative length of the chosen regions was 1.59 Mb with a 98.5% coverage rate. The KAPA HyperPlus and KAPA Library Quantification kits (Roche Sequencing Solutions, CA, USA) were used to prepare the sequencing libraries and quantification of the prepared DNA samples, respectively. The targeted and enriched exonic regions were sequenced with the 150 bp paired-end reads using the HiSeq 4000 System (Ilumina Inc., San Diego, CA, USA) according to the manufacturer’s protocols.

2.2.1. Variant Identification and Annotation

The detailed methodology, including sequencing, data analysis and quality control steps, was performed as previously described [31,32]. Briefly, the sequencing reads that passed quality control were aligned using Burrows–Wheeler Aligner software (BWA; http://bio-bwa.sourceforge.net (accessed on 1 March 2022)) to the UCSC assembly GRChC38/hg38 (https://genome.ucsc.edu (accessed on 1 March 2022)) as a human genome reference sequence. The Genome Analysis Toolkit (GATK, http://www.broadinstitute.org/gatk/ (accessed on 1 March 2022)) was used to call single nucleotide variants and short insertions/deletions. Subsequently, the identified variants were annotated with the databases to provide information regarding their frequency (gnomAD, https://gnomad.broadinstitute.org (accessed on 1 March 2022)) and clinical significance (HGMD, http://www.hgmd.cf.ac.uk (accessed on 1 March 2022) and ClinVar, https://www.ncbi.nlm.nih.gov/clinvar (accessed on 1 March 2022)). Pathogenicity predictions for the variants were uploaded using the genomic variant search engine VarSome (https://varsome.com (accessed on 1 March 2022)), which provided verdicts from 17 in silico tools (SIFT, DEOGEN2, EIGEN, EIGEN PC, FATHMM, FATHMM—MKL, FATHMM—XF, LRT, Meta LR, Meta SVM, Mutation Assessor, Mutation Taster, MVP, PrimateAI, PROVEAN, REVEL, SHIT4G). Additionally, the Combined Annotation Dependent Depletion (CADD) (https://cadd.gs.washington.edu/snv (accessed on 1 March 2022)) scores and adaptive boosting (ADA) scores were provided, respectively, for the missense and splicing variants.

2.2.2. Variant Selection and Prioritisation

The analysis was limited to exonic and splicing variants with a minor allele frequency (MAF) < 0.001 in the gnomAD European (non-Finnish) population. To select variants that may substantially impact encoded protein functions or the splicing process, stop-gain, stop-loss, and frameshift variants, as well as variants with an ADA score ≥ 0.9 and missense variants with a CADD score ≥ 20 were considered. In addition, their pathogenicity must have been confirmed by at least half of the employed in silico tools. Variants detected within genes with the documented association with non-syndromic or syndromic OFCs and genes encoding proteins playing an essential role in craniofacial development were prioritised. Moreover, the selected alternations were ranked by their segregation patterns and classified into three categories: de novo, inherited from unaffected parents and unknown inheritance.

2.2.3. Variant Validation and Segregation

All selected variants were confirmed by direct Sanger sequencing, and segregation analyses were conducted in the case-parents trios when DNA samples were available. The ABI Prism™ BigDye™ Terminator Cycle Sequencing kit was employed using the ABI Prism 3730 capillary sequencer (Applied Biosystems, Foster City, CA, USA) to sequence the PCR products.

3. Results

Targeted NGS Sequencing

After filtering, prioritisation, confirmation and segregation analyses, twelve risk variants that might have the most pathogenic impact on ns-CP in the study cohort were identified (Supplementary Figure S1). The detailed characteristics, including segregation analysis and pathogenicity predictions for all nucleotide alternations detected in ns-CP patients, are presented in Table 2 and Table 3 and Supplementary Figures S2–S13.
Most of the identified alternations (n = 8) were new changes that had not been previously described, of which two were de novo, including the missense variant p.Asp75Gly (c.224A>G) in IRF6 (OMIM * 607199, pLi = 1.0, Z score = 2.7). This p.Asp75Gly substitution was predicted as pathogenic by 16 out of 17 in silico tools and obtained the CADD score of 28.5. The second de novo missense variant was detected in the NHS gene (OMIM * 300457, pLi = 1.0, Z score = 1.8), which had not been previously linked with ns-OFCs. The NHS c.568G>C transversion leading to a p.Val190Leu substitution was evaluated as disrupting by 7 out of 15 in silico tools and had a CADD score of 24.6.
A total of 5 newly identified risk variants were inherited from an unaffected parent, including the splicing variant c.2435-1G>A in COL17A1 (OMIM * 113811, pLi = 0.0, Z score = 0.7). The ADA score for this variant was 0.99, and the CADD score was 33. The other 4 novel alternations inherited from healthy parents were missense variants detected in genes with positive Z scores ranging from 0.8 to 3.5, including c.3605A>G (p.Tyr1202Cys) in FLNB (OMIM * 603381 pLi = 0.0), c.1997A>G (p.Tyr666Cys) in NOTCH2 (OMIM * 600275, pLi = 1.0), c.353A>T (p.Asn118Ile) in TP63 (OMIM * 603273, pLi = 0.9) and c.400G>A (p.Ala134Thr) in VAX1 (OMIM * 604294, pLi = 0.7). Most of the 17 in silico tools used for pathogenicity prediction (mean 15) confirmed the pathogenic status of these nucleotide changes with CADD scores ranging from 26.1 to 29.2.
The newly identified missense variants included c.647A>T (p.His225Leu) in TBX18 (OMIM * 604613, pLi = 0.7, Z score = 0.8), which has an unknown inheritance due to the lack of the father’s DNA. All 17 bioinformatics programmes applied for in silico analysis predicted this variant as pathogenic, and its CADD score was equal to 31.0.
The remaining 4 identified changes were rare missense variants with allele frequency in the gnomAD European population ranging from 0.00 to 2.21E-05, including c.1706G>A (p.Arg569Gln) in ARHGAP29 (OMIM * 610496), c.1587G>C (p.Glu562Asp) in DLG1 (OMIM * 601014), c.481C>A (p.Pro161Thr) in LRP6 (OMIM * 603507) and c.716G>T (p.Arg239Leu) in WNT5B (OMIM * 606361). Except for p.Arg569Gln in ARHGAP29, which inheritance was unknown due to the lack of the father’s DNA, all these rare variants were inherited from an unaffected parent. The selected risk alternations obtained the pathogenic status from all or most of the applied bioinformatics programmes (from 10 to 17) and had CADD scores ranging from 23.6 to 32.0.
Additional characteristics of nucleotide alternations detected in ns-CP patients are presented in Table S3. Table S4 presents characteristics of likely pathogenic variants not fulfilling the project’s strict selection criteria, which were identified in ns-CP patients.

4. Discussion

The aetiology of non-syndromic orofacial clefts consists of multiple genetic components. Since GWASs captured only about 20% of the heritability of nsOFCs [23], many studies have been applied to identify this unrevealed portion of genetic variation and support the essential role of rare functional variants in explaining the “missing heritability” of the ns-OFCs [22,23].
Therefore, this study aimed to detect rare coding variants that are implicated in ns-CP aetiology in the Polish population. For this purpose, coding regions of 423 genes associated with orofacial cleft anomalies and/or involved with facial development were screened in 38 ns-CP patients to identify the top 12 variants that exhibit the most deleterious features. The majority of these alternations were identified in unaffected family members. However, this might provide further support for the heterogeneous nature of the ns-CP [33,34]. The incomplete penetrance of detected pathogenic alternations suggests that they require interactions with other genetics and/or environmental factors to trigger the expression of the cleft phenotype. Therefore, in many studies focusing on the genetics of complex traits and diseases, pathogenic alterations found in asymptomatic carriers are not ruled out from their contribution to disease. Thus, nucleotide variants identified in the current study may be considered one of the genetic components influencing the individual’s risk of ns-CP.
Among selected nucleotide changes, eight were novel and not previously described in the literature. A splicing variant c.2435-1G>A identified in COL17A1 is particularly noteworthy. The COL17A1 gene encodes type XVII collagen, which performs the structural function of hemidesmosomes responsible for maintaining the proper adhesion of epithelial cells to the basement membrane [35]. Interestingly, the gene ontology analysis showed that genes encoding collagen-α chain proteins are significantly enriched exclusively in cleft palate phenotypes [36]. The COL17A1 gene has not been previously linked with ns-CP. However, a GWAS conducted in the European population suggested that nucleotide variants located in COL17A1 may impact facial morphology [37]. In addition, COL17A1 expression increased during the growth and fusion of facial prominences in murine models [38]. Additionally, a rare, likely pathogenic variant of COL17A1 has been described in ns-CLP patients, suggesting that COL17A1 is a novel candidate for isolated orofacial clefts [29,31].
Other novel susceptibility genes for ns-CP detected in our study included DLG1, NHS, NOTCH2, TBX18, VAX1 and WNT5B. While common and rare variants of DLG1, NOTCH2, VAX1 and WNT5B have been previously linked with the risk of ns-CLP [29,31,39,40,41,42], the NHS deleterious variants have been implicated only in Nance–Horan syndrome (OMIM #302350) characterised by craniofacial dysmorphologies [43,44]. The NHS gene encodes three isoforms (A–C), and isoform A is highly expressed during embryonic development in the mesenchymal tissue within the craniofacial and tooth buds regions and plays an essential role in remodelling the actin filaments and maintaining cell morphology [43,44,45]. The NHS variant (p.Val190Leu) detected in the current study was de novo, thus supporting its contribution to ns-CP aetiology [46]. A total of 7 out of the 15 tools used for in silico analyses confirmed that this novel substitution may lead to disturbances in protein structure, and the gene had a positive Z score of 1.8, indicating NHS intolerance to missense variants. However, since the other bioinformatic programmes used in the study predicted the variant to be neutral, a functional study should be conducted to validate these results.
Missense variant c.1997A>G (p.Tyr666Cys) was also detected in the NOTCH2 gene, encoding one of the two primary receptors for the NOTCH conservative signalling pathway involved in craniofacial development [47]. After binding to their ligands (Jagged1, Jagged2 or Delta1), the NOTCH1 and NOTCH2 receptors form a transcription complex to regulate gene expression, including IRF6 [48,49]. The NOTCH2 gene is highly expressed in the mesenchymal cells within the junction regions of facial primordia during lip and primary palate development in murine models [50]. Moreover, mice without the Jagged2 ligand for the NOTCH2 receptor have a cleft palate [51]. Interestingly, the amino acid substitution p.Tyr666Cys identified in this present study is located within the NOTCH extracellular domain (NECD) composed of the repeating EGF-like motif, which is a ligand-binding site [47], suggesting that p.Tyr666Cys might have a deleterious impact on protein interactions. Moreover, most in silico tools used for pathogenicity predictions confirmed the damaging influence of this substitution on protein. It is worth noting that pathogenic variants within the NECD domain cause Alagille syndrome 2 (OMIM #610205), in which facial dysmorphism is a component of the phenotype [52,53]. To our knowledge, this NOTCH2 variant is the first reported damaging alternation of this gene detected in the ns-CP patient.
While rare and common variants in T-box family genes, including TBX1 (OMIM * 602054), TBX10 (OMIM * 604648) and TBX22 (OMIM * 300307), have previously been implicated in both syndromic and isolated orofacial clefts [24,26,54,55,56,57,58], the TBX18 variants have not yet been linked with the risk of these craniofacial anomalies. However, Landson et al. identified a likely damaging copy number variant (CNV) overlapping the TBX18 gene in a patient with an isolated orofacial cleft [59]. The TBX18 gene is a member of the family encoding conserved transcriptional factors highly expressed in craniofacial mesenchymal regions during embryogenesis [60]. The TBX18 variant identified in this current study might be the first reported pathogenic alternation influencing the risk of ns-CP.
Five likely pathogenic variants detected in our ns-CP patients were nucleotide alternations of already-known cleft genes. Interestingly, the de novo variant c.224A>G (p.Asp75Gly) was identified in IRF6, which is the major candidate gene engaged in the pathomechanism underlying ns-OFCs. Its crucial role in face development was previously confirmed, and a number of common and rare variants have been described in ns-OFCs patients [5,61]. The list of known candidate genes carrying likely pathogenic variants detected in this current study also included ARHGAP29, FLNB, LRP6 and TP63 [24,26,62,63,64].
Three rare nucleotide variants with strong pathogenicity predictions were detected in genes implicated in the WNT signalling pathway, which is crucial for proper craniofacial development [65]. Among them was the missense variant in DLG1 encoding essential guanylate kinase, which is involved in proliferative and adhesive processes and the regulation of cell polarity [66,67]. Decreased DLG1 expression results in impaired functioning of the WNT/β-catenin signalling pathway, leading to craniofacial defects [68,69]. DLG1 null mice display delayed embryonic development, cleft palate, mandibular hypertrophy, neural tube abnormalities, cardiovascular system disorders, and a lethal respiratory anomaly [70,71]. The DLG1 p.Glu529Asp substitution disrupts the protein, as evidenced by most in silico prediction algorithms. To our knowledge, the DLG1 variant described in this study is the first reported damaging alternation of this gene detected in the ns-CP patient. Interestingly, a GWAS conducted in the Polish population has established that common variants located in the DLG1 locus are significantly associated with the risk of ns-CL/P [41]. Additionally, Pengelly et al. described a rare heterozygous non-frameshift DLG1 deletion in the ns-CLP family [29].
Other findings supporting the critical role of the WNT signalling pathway in developing craniofacial anomalies are rare, likely pathogenic variants identified in LRP6 encoding co-receptor of the canonical WNT/β-catenin signalling pathway [72,73] and WNT5B encoding signalling ligand implicated in both β-catenin-dependent and β-catenin-independent WNT pathways. Functional studies on animal models have demonstrated that LRP6 and WNT5B knockouts lead to a cleft palate or impaired palatogenesis and short palate, respectively [74,75]. Previous studies have reported the significant association of LRP6 pathogenic variants with congenital neural tube defects [76] and congenital tooth agenesis [77]. The latter frequently occurs with craniofacial abnormalities, suggesting a common genetic background for these defects [78]. Moreover, mice with LRP6 deletion exhibit, besides a cleft palate, decreased expression of MSX1 and MSX2 [75], well-known candidate genes for both ns-OFC and tooth agenesis [78].
The recent large-scale analyses have shown that the nucleotide variants significantly associated with ns-CP were located mainly within intergenic, deep intronic regions of the genome [17,18,19]; therefore, only analysing the coding sequences of the selected genes is the main limitation of our study. Another study weakness is that the applied tools could not screen for CNVs or larger deletions/insertions, which are essential genetic components contributing to ns-CP [79,80,81]. The small number of patients recruited and the lack of parental DNA samples might also be a further limitation. In addition, rigorous variant filtering and strict prioritisation strategy might lead to missing true causative variants.

5. Conclusions

This current study provides further insights into the genetic triggers that may affect the development of the non-syndromic cleft palate and suggests new candidate genes for this craniofacial anomaly.
Moreover, most of the identified likely pathogenic variants were located in genes previously implicated in ns-OFCs; these results confirm the importance of investigating the already-known candidate genes and support the hypothesis that some genes carrying variants associated with syndromic forms of orofacial clefts may also be involved in ns-CP aetiology.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm12052051/s1, Table S1: The NGS-targeted multi-gene panel for ns-CP—characteristics of genes; Table S2: The NGS-targeted multi-gene panel for ns-CP—phenotype of knockout mouse models; Table S3: Characteristics of likely pathogenic variants fulfilling the project’s strict selection criteria, which were identified in ns-CP patients; Table S4: Characteristic of all likely pathogenic variants, including variants not fulfilling the project’s strict selection criteria, which were identified in ns-CP patients; Figure S1: Flow chart of processing next-generation sequencing data; Figure S2: Detection of a novel IRF6 variant in a patient with non-syndromic cleft of the hard and soft palate; Figure S3: Detection of a novel NHS variant in a patient with non-syndromic cleft of the hard and soft palate; Figure S4: Detection of a novel COL17A1 variant in a patient with non-syndromic cleft of the hard and soft palate; Figure S5: Detection of a known DLG1 variant in a patient with non-syndromic cleft of the hard and soft palate; Figure S6: Detection of a novel FLNB variant in a patient with non-syndromic cleft of the hard and soft palate; Figure S7: Detection of a known LRP6 variant in a patient with non-syndromic cleft of the hard and soft palate; Figure S8: Detection of a novel NOTCH2 variant in a patient with non-syndromic cleft of the hard and soft palate; Figure S9: Detection of a novel TP63 variant in a patient with non-syndromic cleft of the hard and soft palate; Figure S10: Detection of a novel VAX1 variant in a patient with non-syndromic cleft of the hard and soft palate; Figure S11: Detection of a known WNT5B variant in a patient with non-syndromic cleft of the hard and soft palate; Figure S12: Detection of a known ARHGAP29 variant in a patient with non-syndromic cleft of the hard and soft palate; Figure S13: Detection of a novel TBX18 variant in a patient with cleft of the soft palate.

Author Contributions

J.D. contributed to the study design, the molecular analyses, the interpretation of the results and the drafting of the manuscript. B.B. and A.B. contributed to the study design, patient and control recruitment and collection and analysis of the clinical data. A.M. was responsible for the study design, the clinical and NGS data analysis, interpreting the results, revising the manuscript, and preparing tables. All authors have read and agreed to the published version of the manuscript.

Funding

The Comprehensive Therapy Program for Craniofacial Anomalies (RPWP.07.02.02-30-0037/16) was supported by European Social Fund and Wielkopolska Voivodeship (Regional Operational Programme for Wielkopolskie Voivodeship 2014-2020).

Institutional Review Board Statement

The study was conducted following the Declaration of Helsinki [30] and approved by the Institutional Review Board of Poznan University of Medical Sciences, Poland (approval number 1115/18).

Informed Consent Statement

Written informed consent was obtained from all study participants or their legal guardians.

Data Availability Statement

The de-identified datasets generated through this study can be provided by the corresponding author upon request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mossey, P.A.; Little, J.; Munger, R.G.; Dixon, M.J.; Shaw, W.C. Cleft Lip and Palate. Lancet 2009, 374, 1773–1785. [Google Scholar] [CrossRef]
  2. Christensen, K.; Juel, K.; Herskind, A.M.; Murray, J.C. Long term follow up study of survival associated with cleft lip and palate at birth. BMJ 2004, 328, 1405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Wehby, G.; Cassell, C.H. The impact of orofacial clefts on quality of life and healthcare use and costs. Oral Dis. 2010, 16, 3–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Zhu, J.L.; Basso, O.; Hasle, H.; Winther, J.F.; Olsen, J.H.; Olsen, J. Do parents of children with congenital malformations have a higher cancer risk? A nationwide study in Denmark. Br. J. Cancer 2002, 87, 524–528. [Google Scholar] [CrossRef] [Green Version]
  5. Mangold, E.; Ludwig, K.U.; Nöthen, M.M. Breakthroughs in the genetics of orofacial clefting. Trends Mol. Med. 2011, 17, 725–733. [Google Scholar] [CrossRef]
  6. Yu, Y.; Zuo, X.; He, M.; Gao, J.; Fu, Y.; Qin, C.; Meng, L.; Wang, W.; Song, Y.; Cheng, Y.; et al. Genome-wide analyses of non-syndromic cleft lip with palate identify 14 novel loci and genetic heterogeneity. Nat. Commun. 2017, 8, 14364. [Google Scholar] [CrossRef] [Green Version]
  7. Ferguson, M.W. Palate Development. Development 1988, 103, 41–60. [Google Scholar] [CrossRef] [PubMed]
  8. Jugessur, A.; Farlie, P.G.; Kilpatrick, N. The genetics of isolated orofacial clefts: From genotypes to subphenotypes. Oral Dis. 2009, 15, 437–453. [Google Scholar] [CrossRef] [PubMed]
  9. Leslie, E.J.; Marazita, M.L. Genetics of cleft lip and cleft palate. Am. J. Med. Genet. Part C Semin. Med. Genet. 2013, 163, 246–258. [Google Scholar] [CrossRef] [Green Version]
  10. Watkins, S.E.; Meyer, R.E.; Strauss, R.P.; Aylsworth, A.S. Classification, Epidemiology, and Genetics of Orofacial Clefts. Clin. Plast. Surg. 2014, 41, 149–163. [Google Scholar] [CrossRef]
  11. Dixon, M.J.; Marazita, M.L.; Beaty, T.H.; Murray, J.C. Cleft lip and palate: Understanding genetic and environmental influences. Nat. Rev. Genet. 2011, 12, 167–178. [Google Scholar] [CrossRef] [Green Version]
  12. Murray, J. Gene/environment causes of cleft lip and/or palate. Clin. Genet. 2002, 61, 248–256. [Google Scholar] [CrossRef]
  13. Gowans, L.J.J.; Adeyemo, W.; Eshete, M.; Mossey, P.; Busch, T.; Aregbesola, B.; Donkor, P.; Arthur, F.K.N.; Bello, S.; Martinez, A.; et al. Association Studies and Direct DNA Sequencing Implicate Genetic Susceptibility Loci in the Etiology of Nonsyndromic Orofacial Clefts in Sub-Saharan African Populations. J. Dent. Res. 2016, 95, 1245–1256. [Google Scholar] [CrossRef] [PubMed]
  14. Ishorst, N.; Francheschelli, P.; Böhmer, A.C.; Khan, M.F.J.; Heilmann-Heimbach, S.; Fricker, N.; Little, J.; Steegers-Theunissen, R.P.M.; Peterlin, B.; Nowak, S.; et al. Nonsyndromic Cleft Palate: An Association Study at GWAS Candidate Loci in a Multiethnic Sample. Birth Defects Res. 2018, 110, 871–882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Beaty, T.H.; Ruczinski, I.; Murray, J.C.; Marazita, M.L.; Munger, R.G.; Hetmanski, J.B.; Murray, T.; Redett, R.J.; Fallin, M.D.; Liang, K.Y.; et al. Evidence for gene-environment interaction in a genome wide study of nonsyndromic cleft palate. Genet. Epidemiol. 2011, 35, 469–478. [Google Scholar] [CrossRef] [PubMed]
  16. Leslie, E.J.; Liu, H.; Carlson, J.C.; Shaffer, J.R.; Feingold, E.; Wehby, G.; Laurie, C.A.; Jain, D.; Laurie, C.C.; Doheny, K.F.; et al. A Genome-Wide Association Study of Nonsyndromic Cleft Palate Identifies an Etiologic Missense Variant in GRHL3. Am. J. Hum. Genet. 2016, 98, 744–754. [Google Scholar] [CrossRef] [Green Version]
  17. Butali, A.; Mossey, P.A.; Adeyemo, W.L.; Eshete, M.A.; Gowans, L.J.J.; Busch, T.D.; Jain, D.; Yu, W.; Huan, L.; Laurie, C.A.; et al. Genomic analyses in African populations identify novel risk loci for cleft palate. Hum. Mol. Genet. 2019, 28, 1038–1051. [Google Scholar] [CrossRef] [Green Version]
  18. Huang, L.; Jia, Z.; Shi, Y.; Du, Q.; Shi, J.; Wang, Z.; Mou, Y.; Wang, Q.; Zhang, B.; Wang, Q.; et al. Genetic factors define CPO and CLO subtypes of nonsyndromicorofacial cleft. PLoS Genet. 2019, 15, e1008357. [Google Scholar] [CrossRef] [PubMed]
  19. He, M.; Zuo, X.; Liu, H.; Wang, W.; Zhang, Y.; Fu, Y.; Zhen, Q.; Yu, Y.; Pan, Y.; Qin, C.; et al. Genome-wide Analyses Identify a Novel Risk Locus for Nonsyndromic Cleft Palate. J. Dent. Res. 2020, 99, 1461–1468. [Google Scholar] [CrossRef]
  20. Mangold, E.; Böhmer, A.C.; Ishorst, N.; Hoebel, A.-K.; Gültepe, P.; Schuenke, H.; Klamt, J.; Hofmann, A.; Gölz, L.; Raff, R.; et al. Sequencing the GRHL3 Coding Region Reveals Rare Truncating Mutations and a Common Susceptibility Variant for Nonsyndromic Cleft Palate. Am. J. Hum. Genet. 2016, 98, 755–762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Martinelli, M.; Palmieri, A.; Carinci, F.; Scapoli, L. Non-syndromic Cleft Palate: An Overview on Human Genetic and Environmental Risk Factors. Front. Cell Dev. Biol. 2020, 8, 592271. [Google Scholar] [CrossRef] [PubMed]
  22. Marini, N.J.; Asrani, K.; Yang, W.; Rine, J.; Shaw, G.M. Accumulation of rare coding variants in genes implicated in risk of human cleft lip with or without cleft palate. Am. J. Med. Genet. Part A 2019, 179, 1260–1269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Leslie, E.J.; Carlson, J.C.; Shaffer, J.R.; Buxó, C.J.; Castilla, E.E.; Christensen, K.; Deleyiannis, F.W.B.; Field, L.L.; Hecht, J.T.; Moreno, L.; et al. Association studies of low-frequency coding variants in nonsyndromic cleft lip with or without cleft palate. Am. J. Med. Genet. Part A 2017, 173, 1531–1538. [Google Scholar] [CrossRef] [PubMed]
  24. Machado, R.A.; Martelli-Junior, H.; Reis, S.R.D.A.; Küchler, E.C.; Scariot, R.; das Neves, L.T.; Coletta, R.D. Identification of Novel Variants in Cleft Palate-Associated Genes in Brazilian Patients With Non-syndromic Cleft Palate Only. Front. Cell Dev. Biol. 2021, 9, 638522. [Google Scholar] [CrossRef]
  25. Bureau, A.; Parker, M.; Ruczinski, I.; Taub, M.A.; Marazita, M.L.; Murray, J.C.; Mangold, E.; Noethen, M.M.; Ludwig, K.U.; Hetmanski, J.B.; et al. Whole Exome Sequencing of Distant Relatives in Multiplex Families Implicates Rare Variants in Candidate Genes for Oral Clefts. Genetics 2014, 197, 1039–1044. [Google Scholar] [CrossRef] [Green Version]
  26. Basha, M.; Demeer, B.; Revencu, N.; Helaers, R.; Theys, S.; Saba, S.B.; Boute, O.; Devauchelle, B.; Francois, G.; Bayet, B.; et al. Whole exome sequencing identifies mutations in 10% of patients with familial non-syndromic cleft lip and/or palate in genes mutated in well-known syndromes. J. Med. Genet. 2018, 55, 449–458. [Google Scholar] [CrossRef] [Green Version]
  27. Hoebel, A.; Drichel, D.; Van De Vorst, M.; Böhmer, A.; Sivalingam, S.; Ishorst, N.; Klamt, J.; Gölz, L.; Alblas, M.; Maaser-Hecker, A.; et al. Candidate Genes for Nonsyndromic Cleft Palate Detected by Exome Sequencing. J. Dent. Res. 2017, 96, 1314–1321. [Google Scholar] [CrossRef]
  28. Liu, H.; Busch, T.; Eliason, S.; Anand, D.; Bullard, S.; Gowans, L.J.; Nidey, N.; Petrin, A.; Augustine-Akpan, E.-A.; Saadi, I.; et al. Exome sequencing provides additional evidence for the involvement ofARHGAP29in Mendelian orofacial clefting and extends the phenotypic spectrum to isolated cleft palate. Birth Defects Res. 2017, 109, 27–37. [Google Scholar] [CrossRef] [Green Version]
  29. Pengelly, R.J.; Arias, L.; Martínez, J.; Upstill-Goddard, R.; Seaby, E.G.; Gibson, J.; Ennis, S.; Collins, A.; Briceño, I. Deleterious coding variants in multi-case families with non-syndromic cleft lip and/or palate phenotypes. Sci. Rep. 2016, 6, 30457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Sawicka-Gutaj, N.; Gruszczyński, D.; Guzik, P.; Mostowska, A.; Walkowiak, J. Publication Ethics of Human Studies in the Light of the Declaration of Helsinki—A Mini-Review. J. Med. Sci. 2022, 91, e700. [Google Scholar] [CrossRef]
  31. Dąbrowska, J.; Biedziak, B.; Szponar-Żurowska, A.; Budner, M.; Jagodziński, P.P.; Płoski, R.; Mostowska, A. Identification of Novel Susceptibility Genes for Non-Syndromic Cleft Lip with or without Cleft Palate Using NGS-Based Multigene Panel Testing. Mol. Genet. Genomics 2022, 297, 1315–1327. [Google Scholar] [CrossRef]
  32. Biedziak, B.; Firlej, E.; Dąbrowska, J.; Bogdanowicz, A.; Zadurska, M.; Mostowska, A. Novel Candidate Genes for Non-Syndromic Tooth Agenesis Identified Using Targeted Next-Generation Sequencing. J. Clin. Med. 2022, 11, 6089. [Google Scholar] [CrossRef] [PubMed]
  33. Carlson, J.C.; Taub, M.A.; Feingold, E.; Beaty, T.H.; Murray, J.C.; Marazita, M.L.; Leslie, E.J.; Anand, D.; Butali, A.; Buxo, C.J.; et al. Identifying Genetic Sources of Phenotypic Heterogeneity in Orofacial Clefts by Targeted Sequencing. Genet. Epidemiol. 2017, 43, 704–716. [Google Scholar] [CrossRef] [PubMed]
  34. Carlson, J.C.; Anand, D.; Butali, A.; Buxo, C.J.; Christensen, K.; Deleyiannis, F.; Hecht, J.T.; Moreno, L.M.; Orioli, I.M.; Padilla, C.; et al. A Systematic Genetic Analysis and Visualization of Phenotypic Heterogeneity among Orofacial Cleft GWAS Signals. Genet. Epidemiol. 2019, 43, 704–716. [Google Scholar] [CrossRef]
  35. Kroeger, J.; Hoppe, E.; Galiger, C.; Has, C.; Franzke, C.W. Amino Acid Substitution in the C-Terminal Domain of Collagen XVII Reduces Laminin-332 Interaction Causing Mild Skin Fragility with Atrophic Scarring. Matrix Biol. 2019, 80, 72–84. [Google Scholar] [CrossRef]
  36. Funato, N.; Nakamura, M. Identification of shared and unique gene families associated with oral clefts. Int. J. Oral Sci. 2017, 9, 104–109. [Google Scholar] [CrossRef]
  37. Liu, F.; van der Lijn, F.; Schurmann, C.; Zhu, G.; Chakravarty, M.M.; Hysi, P.G.; Wollstein, A.; Lao, O.; de Bruijne, M.; Ikram, M.A.; et al. A Genome-Wide Association Study Identifies Five Loci Influencing Facial Morphology in Europeans. PLoS Genet. 2012, 8, e1002932. [Google Scholar] [CrossRef] [Green Version]
  38. Feng, W.; Leach, S.M.; Tipney, H.; Phang, T.; Geraci, M.; Spritz, R.A.; Hunter, L.E.; Williams, T. Spatial and Temporal Analysis of Gene Expression during Growth and Fusion of the Mouse Facial Prominences. PLoS ONE 2009, 4, e8066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Mangold, E.; Ludwig, K.U.; Birnbaum, S.; Baluardo, C.; Ferrian, M.; Herms, S.; Reutter, H.; de Assis, N.A.; Al Chawa, T.; Mattheisen, M.; et al. Genome-wide association study identifies two susceptibility loci for nonsyndromic cleft lip with or without cleft palate. Nat. Genet. 2010, 42, 24–26. [Google Scholar] [CrossRef] [Green Version]
  40. Butali, A.; Suzuki, S.; Cooper, M.E.; Mansilla, A.M.; Cuenco, K.; Leslie, E.J.; Suzuki, Y.; Niimi, T.; Yamamoto, M.; Ayanga, G.; et al. Replication of Genome Wide Association Identified Candidate Genes Confirm the Role of Common and Rare Variants in PAX7 and VAX1 in the Etiology of Nonsyndromic CL(P). Am. J. Med. Genet. A 2013, 161, 965–972. [Google Scholar] [CrossRef] [Green Version]
  41. Mostowska, A.; Gaczkowska, A.; Żukowski, K.; Ludwig, K.U.; Hozyasz, K.K.; Wójcicki, P.; Mangold, E.; Böhmer, A.C.; Heilmann-Heimbach, S.; Knapp, M.; et al. Common Variants in DLG1 Locus Are Associated with Non-Syndromic Cleft Lip with or without Cleft Palate. Clin. Genet. 2018, 93, 784–793. [Google Scholar] [CrossRef]
  42. Li, Q.; Kim, Y.; Suktitipat, B.; Hetmanski, J.B.; Marazita, M.L.; Duggal, P.; Beaty, T.H.; Bailey-Wilson, J.E. Gene-Gene Interaction Among WNT Genes for Oral Cleft in Trios. Genet. Epidemiol. 2015, 39, 385–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Brooks, S.P.; Coccia, M.; Tang, H.R.; Kanuga, N.; Machesky, L.M.; Bailly, M.; Cheetham, M.E.; Hardcastle, A.J. The Nance–Horan syndrome protein encodes a functional WAVE homology domain (WHD) and is important for co-ordinating actin remodelling and maintaining cell morphology. Hum. Mol. Genet. 2010, 19, 2421–2432. [Google Scholar] [CrossRef] [Green Version]
  44. Burdon, K.P.; McKay, J.D.; Sale, M.M.; Russell-Eggitt, I.M.; Mackey, D.A.; Wirth, M.G.; Elder, J.E.; Nicoll, A.; Clarke, M.; FitzGerald, L.; et al. Mutations in a Novel Gene, NHS, Cause the Pleiotropic Effects of Nance-Horan Syndrome, Including Severe Congenital Cataract, Dental Anomalies, and Mental Retardation. Am. J. Hum. Genet. 2003, 73, 1120–1130. [Google Scholar] [CrossRef] [Green Version]
  45. Khan, A.O.; Aldahmesh, M.A.; Mohamed, J.Y.; Alkuraya, F.S. Phenotype-genotype Correlation in Potential Female Carriers of X-linked Developmental Cataract (Nance-Horan Syndrome). Ophthalmic Genet. 2012, 33, 89–95. [Google Scholar] [CrossRef] [PubMed]
  46. Bishop, M.R.; Diaz Perez, K.K.; Sun, M.; Ho, S.; Chopra, P.; Mukhopadhyay, N.; Hetmanski, J.B.; Taub, M.A.; Moreno-Uribe, L.M.; Valencia-Ramirez, L.C.; et al. Genome-Wide Enrichment of De Novo Coding Mutations in Orofacial Cleft Trios. Am. J. Hum. Genet. 2020, 107, 124–136. [Google Scholar] [CrossRef] [PubMed]
  47. Pakvasa, M.; Haravu, P.; Boachie-Mensah, M.; Jones, A.; Coalson, E.; Liao, J.; Zeng, Z.; Wu, D.; Qin, K.; Wu, X.; et al. Notch signaling: Its essential roles in bone and craniofacial development. Genes Dis. 2021, 8, 8–24. [Google Scholar] [CrossRef] [PubMed]
  48. Restivo, G.; Nguyen, B.-C.; Dziunycz, P.; Ristorcelli, E.; Ryan, R.J.H.; Özuysal, Ö.Y.; Di Piazza, M.; Radtke, F.; Dixon, M.J.; Hofbauer, G.F.L.; et al. IRF6 is a mediator of Notch pro-differentiation and tumour suppressive function in keratinocytes. EMBO J. 2011, 30, 4571–4585. [Google Scholar] [CrossRef] [PubMed]
  49. Reynolds, K.; Zhang, S.; Sun, B.; Garland, M.A.; Ji, Y.; Zhou, C.J. Genetics and signaling mechanisms of orofacial clefts. Birth Defects Res. 2020, 112, 1588–1634. [Google Scholar] [CrossRef]
  50. Li, H.; Jones, K.L.; Hooper, J.E.; Williams, T. The Molecular Anatomy of Mammalian Upper Lip and Primary Palate Fusion at Single Cell Resolution. Development 2019, 146, dev174888. [Google Scholar] [CrossRef] [Green Version]
  51. Richardson, R.J.; Dixon, J.; Jiang, R.; Dixon, M.J. Integration of IRF6 and Jagged2 signalling is essential for controlling palatal adhesion and fusion competence. Hum. Mol. Genet. 2009, 18, 2632–2642. [Google Scholar] [CrossRef] [Green Version]
  52. Gilbert, M.A.; Bauer, R.C.; Rajagopalan, R.; Grochowski, C.M.; Chao, G.; McEldrew, D.; Nassur, J.A.; Rand, E.B.; Krock, B.L.; Kamath, B.M.; et al. Alagille syndrome mutation update: Comprehensive overview of JAG1 and NOTCH2 mutation frequencies and insight into missense variant classification. Hum. Mutat. 2019, 40, 2197–2220. [Google Scholar] [CrossRef] [Green Version]
  53. Kamath, B.M.; Bauer, R.C.; Loomes, K.M.; Chao, G.; Gerfen, J.; Hutchinson, A.; Hardikar, W.; Hirschfield, G.; Jara, P.; Krantz, I.D.; et al. NOTCH2mutations in Alagille syndrome. J. Med. Genet. 2012, 49, 138–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Herman, S.B.; Guo, T.; McGinn, D.M.M.; Blonska, A.; Shanske, A.L.; Bassett, A.S.; Chow, E.W.C.; Bowser, M.; Sheridan, M.; Beemer, F.; et al. Overt Cleft Palate Phenotype and TBX1 Genotype Correlations in Velo-Cardio-Facial/DiGeorge/22q11. 2 Deletion Syndrome Patients. Am. J. Med. Genet. A 2012, 158A, 2781–2787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Vieira, A.R.; Avila, J.R.; Daack-Hirsch, S.; Dragan, E.; Félix, T.M.; Rahimov, F.; Harrington, J.; Schultz, R.R.; Watanabe, Y.; Johnson, M.; et al. Medical Sequencing of Candidate Genes for Nonsyndromic Cleft Lip and Palate. PLoS Genet. 2005, 1, e64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Mostowska, A.; Hozyasz, K.K.; Wojcicki, P.; Biedziak, B.; Paradowska, P.; Jagodzinski, P.P. Association between genetic variants of reported candidate genes or regions and risk of cleft lip with or without cleft palate in the polish population. Birth Defects Res. Part A Clin. Mol. Teratol. 2010, 88, 538–545. [Google Scholar] [CrossRef] [PubMed]
  57. Marcano, A.C.B.; Doudney, K.; Braybrook, C.; Squires, R.; Patton, M.A.; Lees, M.M.; Richieri-Costa, A.; Lidral, A.C.; Murray, J.C.; Moore, G.E.; et al. TBX22 mutations are a frequent cause of cleft palate. J. Med. Genet. 2004, 41, 68–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Stanier, P.; Moore, G.E. Genetics of Cleft Lip and Palate: Syndromic Genes Contribute to the Incidence of Non-Syndromic Clefts. Hum. Mol. Genet. 2004, 13, 73–81. [Google Scholar] [CrossRef] [PubMed]
  59. Lansdon, L.A.; Darbro, B.W.; Petrin, A.L.; Hulstrand, A.M.; Standley, J.M.; Brouillette, R.B.; Long, A.; Mansilla, M.A.; Cornell, R.A.; Murray, J.C.; et al. Identification of Isthmin 1 as a Novel Clefting and Craniofacial Patterning Gene in Humans. Genetics 2018, 208, 283–296. [Google Scholar] [CrossRef] [Green Version]
  60. Kraus, F.; Haenig, B.; Kispert, A. Cloning and expression analysis of the mouse T-box gene Tbx18. Mech. Dev. 2001, 100, 83–86. [Google Scholar] [CrossRef]
  61. Richardson, B.; Dixon, J.; Malhotra, S.; Hardman, M.; Knowles, L.; Boot-Handford, R.; Shore, P.; Whitmarsh, A.; Dixon, M.J. Irf6 is a key determinant of the keratinocyte proliferation-differentiation switch. Nat. Genet. 2006, 38, 1329–1334. [Google Scholar] [CrossRef]
  62. Letra, A.; Maili, L.; Mulliken, J.B.; Buchanan, E.; Blanton, S.H.; Hecht, J.T. Further evidence suggesting a role for variation in ARHGAP29 variants in nonsyndromic cleft lip/palate. Birth Defects Res. Part A Clin. Mol. Teratol. 2014, 100, 679–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Khandelwal, K.D.; van den Boogaard, M.-J.H.; Mehrem, S.L.; Gebel, J.; Fagerberg, C.; Van Beusekom, E.; Van Binsbergen, E.; Topaloglu, O.; Steehouwer, M.; Gilissen, C.; et al. Deletions and loss-of-function variants in TP63 associated with orofacial clefting. Eur. J. Hum. Genet. 2019, 27, 1101–1112. [Google Scholar] [CrossRef] [PubMed]
  64. Jugessur, A.; Shi, M.; Gjessing, H.K.; Lie, R.T.; Wilcox, A.J.; Weinberg, C.; Christensen, K.; Boyles, A.L.; Daack-Hirsch, S.; Nguyen, T.T.; et al. Maternal Genes and Facial Clefts in Offspring: A Comprehensive Search for Genetic Associations in Two Population-Based Cleft Studies from Scandinavia. PLoS ONE 2010, 5, e11493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. He, F.; Chen, Y. Wnt Signaling in Lip and Palate Development. Front. Oral Biol. 2012, 16, 81–90. [Google Scholar] [CrossRef] [PubMed]
  66. Woods, D.F.; Hough, C.; Peel, D.; Callaini, G.; Bryant, P.J. Dlg Protein Is Required for Junction Structure, Cell Polarity, and Proliferation Control in Drosophila Epithelia. J. Cell Biol. 1996, 134, 1469–1482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Yamanaka, T.; Ohno, S. Role of Lgl/Dlg/Scribble in the Regulation of Epithelial Junction, Polarity and Growth. Front. Biosci. 2008, 13, 6693–6707. [Google Scholar] [CrossRef] [Green Version]
  68. Iizuka-Kogo, A.; Senda, T.; Akiyama, T.; Shimomura, A.; Nomura, R.; Hasegawa, Y.; Yamamura, K.-I.; Kogo, H.; Sawai, N.; Matsuzaki, T. Requirement of DLG1 for Cardiovascular Development and Tissue Elongation during Cochlear, Enteric, and Skeletal Development: Possible Role in Convergent Extension. PLoS ONE 2015, 10, e0123965. [Google Scholar] [CrossRef] [Green Version]
  69. Seelan, R.S.; Mukhopadhyay, P.; Warner, D.R.; Webb, C.L.; Pisano, M.; Greene, R.M.; Hong, C.-S.; Saint-Jeannet, J.-P.; Lefebvre, V.; Bhattaram, P.; et al. Requirement for Dlgh-1 in Planar Cell Polarity and Skeletogenesis during Vertebrate Development. Development 2013, 134, 6693–6707. [Google Scholar]
  70. Caruana, G.; Bernstein, A. Craniofacial Dysmorphogenesis Including Cleft Palate in Mice with an Insertional Mutation in the discs large Gene. Mol. Cell. Biol. 2001, 21, 1475–1483. [Google Scholar] [CrossRef] [Green Version]
  71. Iizuka-Kogo, A.; Ishidao, T.; Akiyama, T.; Senda, T. Abnormal development of urogenital organs in Dlgh1-deficient mice. Development 2007, 134, 1799–1807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Menezes, R.; Letra, A.; Kim, A.H.; Küchler, E.C.; Day, A.; Tannure, P.N.; Gomes da Motta, L.; Paiva, K.B.S.; Granjeiro, J.M.; Vieira, A.R. Studies with Wnt Genes and Nonsyndromic Cleft Lip and Palate. Birth Defects Res. A Clin. Mol. Teratol. 2010, 88, 995–1000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Chiquet, B.T.; Blanton, S.H.; Burt, A.; Ma, D.; Stal, S.; Mulliken, J.B.; Hecht, J.T. Variation in WNT genes is associated with non-syndromic cleft lip with or without cleft palate. Hum. Mol. Genet. 2008, 17, 2212–2218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Rochard, L.; Monica, S.D.; Ling, I.T.C.; Kong, Y.; Roberson, S.; Harland, R.; Halpern, M.; Liao, E.C. Roles of Wnt Pathway Genes Wls, Wnt9a, Wnt5b, Frzb and Gpc4 in Regulating Convergent-Extension during Zebrafish Palate Morphogenesis. Development 2016, 143, 2541–2547. [Google Scholar]
  75. Song, L.; Li, Y.; Wang, K.; Wang, Y.-Z.; Molotkov, A.; Gao, L.; Zhao, T.; Yamagami, T.; Wang, Y.; Gan, Q.; et al. Lrp6-mediated canonical Wnt signaling is required for lip formation and fusion. Development 2009, 136, 3161–3171. [Google Scholar] [CrossRef] [Green Version]
  76. Lei, Y.; Fathe, K.; McCartney, D.; Zhu, H.; Yang, W.; Ross, M.E.; Shaw, G.M.; Finnell, R.H. Rare LRP6 Variants Identified in Spina Bifida P atients. Hum. Mutat. 2015, 36, 342–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Ockeloen, C.W.; Khandelwal, K.D.; Dreesen, K.; Ludwig, K.U.; Sullivan, R.; Van Rooij, I.A.L.M.; Thonissen, M.; Swinnen, S.; Phan, M.; Conte, F.; et al. Novel mutations in LRP6 highlight the role of WNT signaling in tooth agenesis. Genet. Med. 2016, 18, 1158–1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Phan, M.; Conte, F.; Khandelwal, K.D.; Ockeloen, C.W.; Bartzela, T.; Kleefstra, T.; van Bokhoven, H.; Rubini, M.; Zhou, H.; Carels, C.E.L. Tooth Agenesis and Orofacial Clefting: Genetic Brothers in Arms? Hum. Genet. 2016, 135, 1299–1327. [Google Scholar] [CrossRef] [Green Version]
  79. Conte, F.; Oti, M.; Dixon, J.; Carels, C.E.L.; Rubini, M.; Zhou, H. Systematic analysis of copy number variants of a large cohort of orofacial cleft patients identifies candidate genes for orofacial clefts. Hum. Genet. 2016, 135, 41–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Shi, M.; Mostowska, A.; Jugessur, A.; Johnson, M.K.; Mansilla, M.A.; Christensen, K.; Lie, R.T.; Wilcox, A.J.; Murray, J.C. Identification of Microdeletions in Candidate Genes for Cleft Lip and/or Palate. Birth Defects Res. A Clin. Mol. Teratol. 2009, 85, 42–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Osoegawa, K.; Vessere, G.M.; Utami, K.H.; Mansilla, M.A.; Johnson, M.K.; Riley, B.M.; L’Heureux, J.; Pfundt, R.; Staaf, J.; Van Der Vliet, W.A.; et al. Identification of novel candidate genes associated with cleft lip and palate using array comparative genomic hybridisation. J. Med. Genet. 2008, 45, 81–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Characteristics of the patient group.
Table 1. Characteristics of the patient group.
All Patients (n = 38)
N%
Gender distribution
  Males1744.74
  Females2155.26
Cleft type
  Cleft of the soft palate37.89
  Cleft of the hard and soft palate3592.11
Associated anomalies
  Dental anomalies 1821.05
Positive family history
  Extended family410.53
1 For 21 patients, panoramic radiographs were not available for analysis.
Table 2. Nucleotide variants identified in patients with ns-CP.
Table 2. Nucleotide variants identified in patients with ns-CP.
Variant EffectgnomADGenotyping Results c
PatientGenderType of CleftFamily History aGeneRs NumberDNA ChangeProtein ChangeFrequency bPatientFatherMother
De novo Variants
CP_1MCleft of the hard and soft palateNOIRF6novelc.224A>Gp.Asp75Glynot foundHETwtwt
CP_2FCleft of the hard and soft palateYES—ex. fam.NHSnovelc.568G>Cp.Val190Leunot foundHETwtwt
Variants Inherited from Unaffected Parent
CP_3FCleft of the hard and soft palateNOCOL17A1novelc.2435-1G>A not foundHETwtHET
CP_4FCleft of the hard and soft palateYES—ex. fam.DLG1rs139789027c.1586G>Cp.Glu562Asp2.95 × 10−5HETwtHET
CP_5FCleft of the hard and soft palateNOFLNBnovelc.3605A>Gp.Tyr1202Cysnot foundHETno DNAHET
CP_6FCleft of the hard and soft palateNOLRP6rs769446193c.481C>Ap.Pro161Thrnot foundHETno DNAHET
CP_7FCleft of the hard and soft palateNONOTCH2novelc.1997A>Gp.Tyr666Cysnot foundHETHETwt
CP_8MCleft of the hard and soft palateNOTP63novelc.353A>Tp.Asn118Ilenot foundHETwtHET
CP_9FCleft of the hard and soft palateNOVAX1novelc.400G>Ap.Ala134Thrnot foundHETwtHET
CP_10MCleft of the hard and soft palateNOWNT5Brs529807731c.716G>Tp.Arg239Leu0.000HETHETwt
Variants with Unknown Inheritance
CP_11MCleft of the hard and soft palateNOARHGAP29rs867470445c.1706G>Ap.Arg569Gln1.47 × 10−5HETno DNAwt
CP_12FCleft of the soft palateNOTBX18novelc.674A>Tp.His225Leunot foundHETno DNAwt
a ex. fam., extended family. b The Genome Aggregation Database v.3.1.1 [gnomAD, genome sequencing data, population: European (non-Finish)] https://gnomad.broadinstitute.org/ (accessed on 1 March 2022). c Sanger sequencing; HET, heterozygote; wt, wild-type.
Table 3. Pathogenicity scores of variants detected in ns-CP patients.
Table 3. Pathogenicity scores of variants detected in ns-CP patients.
GENEVARIANTIN SILICO PATHOGENICITY PREDICTION cADACADD
NamepLi aZ score bAlternationType1234567891011121314151617Score dScore e
De novo Variants
IRF61.0002.774c.224A>G; p.Asp75GlyMissenseDcMDDDDDPPDDDPPDDD 28.5
NHS1.0001.822c.568G>C; p.Val190LeuMissenseDcNTDDDT T, DT, DNBBDTT 24.6
Variants Inherited from Unaffected Parent
COL17A10.000−0.748c.2435-1G>ASplicingD D P P 0.9933.0
DLG10.9941.496c.1586G>C; p.Glu562AspMissenseDcMDDNDTBPDDNBBTDD 23.6
FLNB0.0002.141c.3605A>G; p.Tyr1202CysMissenseDcMTDDDDPPDDDPPDDD 29.2
LRP60.6972.754c.481C>A; p.Pro161ThrMissenseDcHDDDDDPPDDDPPDDD 26.0
NOTCH21.0003.502c.1997A>G; p.Tyr666CysMissenseDcHDDDUDPPDDDPPTDD 26.1
TP630.9972.208c.353A>T; p.Asn118IleMissenseDcLT, DDDDDPPDDDPPDDD 27.6
VAX10.7710.869c.400G>A; p.Ala134ThrMissenseDcHDDDDDPPDDDPPDDD 27.6
WNT5B0.5891.788c.716G>T; p.Arg239LeuMissenseDcHTDDDDPPDDDBPDDD 32.0
Variants with Unknown Inheritance
ARHGAP291.0001.211c.1706G>A; p.Arg569GlnMissenseDcMTDDNTPPDDDBBDTT 28.4
TBX180.9990.099c.674A>T; p.His225LeuMissenseDcHDDDDDPPDDDPPDDD 31.0
a pLi, the probability of being loss-of-function (LoF) intolerant; pLi ≥ 0.9 indicates that gene is LoF intolerant—The Genome Aggregation Database v.2.1.1. https://gnomad.broadinstitute.org/ (accessed on1 March 2022 ). b Z score indicates a gene’s intolerance to missense variants; positive Z scores indicate increased constraint (intolerance to variation)—The Genome Aggregation Database v.2.1.1. c Pathogenicity of missense variants was predicted using 17 in silico tools (1—MutationTaster, 2—MutationAssessor, 3—FATHMM, 4—FATHMM-MKL, 5—FATHMM-XF, 6—LRT, 7—DEOGEN2, 8—EIGEN, 9—EIGEN PC, 10—SIFT, 11—SIFT4G, 12—PROVEAN, 13—MVP, 14—REVEL, 15—PrimateAI, 16—MetaSVM and 17—MetaLR). B, Benign; D, Damaging/Deleterious; Dc, Disease causing; H, High; L, Low; M, Medium; N, Neutral; P, Pathogenic; T, Tolerated; U, Uncertain. d Adaptive boosting (ADA) score for variants predicted to affect splicing; the ADA score close to 1 indicates a high probability of being splice-affecting variant—dbNSFP database. e CADD—Combined Annotation Dependent Depletion version 1.6. https://cadd.gs.washington.edu/ (accessed on 1 March 2022).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dąbrowska, J.; Biedziak, B.; Bogdanowicz, A.; Mostowska, A. Identification of Novel Risk Variants of Non-Syndromic Cleft Palate by Targeted Gene Panel Sequencing. J. Clin. Med. 2023, 12, 2051. https://doi.org/10.3390/jcm12052051

AMA Style

Dąbrowska J, Biedziak B, Bogdanowicz A, Mostowska A. Identification of Novel Risk Variants of Non-Syndromic Cleft Palate by Targeted Gene Panel Sequencing. Journal of Clinical Medicine. 2023; 12(5):2051. https://doi.org/10.3390/jcm12052051

Chicago/Turabian Style

Dąbrowska, Justyna, Barbara Biedziak, Agnieszka Bogdanowicz, and Adrianna Mostowska. 2023. "Identification of Novel Risk Variants of Non-Syndromic Cleft Palate by Targeted Gene Panel Sequencing" Journal of Clinical Medicine 12, no. 5: 2051. https://doi.org/10.3390/jcm12052051

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

Article Metrics

Back to TopTop