Skip to main content
European Journal of Human Genetics logoLink to European Journal of Human Genetics
. 2024 Dec 19;33(5):595–606. doi: 10.1038/s41431-024-01775-9

Role of ZFHX4 in orofacial clefting based on human genetic data and zebrafish models

Nina Ishorst 1,2,✉,#, Selina Hölzel 1,3,#, Carola Greve 1,12, Öznur Yilmaz 2, Tobias Lindenberg 2, Jessica Lambertz 2, Dmitriy Drichel 4, Berina Zametica 1, Enrico Mingardo 3,13, Jeshurun C Kalanithy 1,2, Khadija Channab 3, Duygu Kibris 1,2, Sabrina Henne 1, Franziska Degenhardt 1,14, Anna Siewert 1, Michael Dixon 5, Teresa Kruse 6, Edwin Ongkosuwito 7, Katta M Girisha 8, Shruti Pande 8, Stefanie Nowak 1, Gregor Hagelueken 9, Matthias Geyer 9, Carine Carels 10, Iris A L M van Rooij 11, Kerstin U Ludwig 1, Benjamin Odermatt 2,3,, Elisabeth Mangold 1,
PMCID: PMC7617551  EMSID: EMS204109  PMID: 39702590

Abstract

Orofacial clefting (OFC) is a frequent congenital anomaly and can occur either in the context of underlying syndromes or in isolation (nonsyndromic). The two common OFC phenotypes are cleft lip with/without cleft palate (CL/P) and cleft palate only (CPO). In this study, we searched for penetrant CL/P genes, by evaluating de novo copy number variants (CNV) from an exome sequencing dataset of 50 nonsyndromic patient-parent trios. We detected a heterozygous 86 kb de novo deletion affecting exons 4–11 of ZFHX4, a gene previously associated with OFC. Genetic and phenotypic data from our in-house and the AGORA cohort (710 and 229 individuals with nonsyndromic CL/P) together with literature and database reviews demonstrate that ZFHX4 variants can lead to both nonsyndromic and syndromic forms not only of CL/P but also CPO. Expression analysis in published single-cell RNA-sequencing data (mouse embryo, zebrafish larva) at relevant time-points support an important role of Zfhx4/zfhx4 in craniofacial development. To characterize the role of zfhx4 in zebrafish craniofacial development, we knocked out/down the zebrafish orthologue. Cartilage staining of the zfhx4 CRISPR F0 knockout and morpholino knockdown at 4 days post-fertilization showed an underdeveloped and abnormally shaped ethmoid plate and cartilaginous jaw (resembling micrognathia). While there is evidence for the dominant inheritance of ZFHX4 variants in OFC, we here present a patient with a possible recessive inheritance. In conclusion, ZFHX4 has a highly heterogeneous phenotypic spectrum and variable mode of inheritance. Our data highlight that ZFHX4 should be considered in genetic testing in patients with nonsyndromic clefting.

Subject terms: Development, Disease genetics

Introduction

Orofacial clefts are among the most common congenital malformations in humans [1]. The two common phenotypes are cleft lip with or without cleft palate (CL/P) and cleft palate only (CPO). Clefting can occur in the context of complex syndromes or in an isolated nonsyndromic form, which is more common, i.e. it is assumed that 70% of CL/Ps are nonsyndromic (nsCL/P [MIM:612858]) [2]. The boundaries between mild syndromic and nonsyndromic forms can be fluid, especially when additional anomalies are continuous features, such as intellectual disability. Genetic and epidemiological data on nsCL/P indicate that high-penetrance genetic variants in “major” genes act on a multifactorial background [3]. Genome-wide association studies (GWAS) and follow-up studies have identified more than 40 common risk loci across diverse populations [47]. However, the estimated heritability is around 90% and GWAS-based estimates indicate that common variation explains only a fraction of the estimated heritability (e.g., <40% among Europeans) [6].

A fraction of the missing heritability of nsCL/P may be explained by rare, highly penetrant variants in genes potentially involved in craniofacial development. Sequencing studies have found functionally relevant penetrant variants in genes that were selected as candidate genes on the basis of evidence from syndromic forms of orofacial clefting [8, 9] or their location at previously identified linkage/GWAS loci [1012]. More systematic approaches identified novel, highly penetrant nsCL/P susceptibility genes via exome sequencing (ES) in multiple affected families [13].

A complementary approach to the identification of highly penetrant susceptibility genes is to search for rare, highly penetrant de novo variants [1416]. A study of nsCL/P and nonsyndromic cleft palate only (nsCPO) identified a significant enrichment of loss-of-function de novo variants in two genes, one of which was Zinc Finger Homeobox 4 (ZFHX4) [14]. A functional study in mice provided additional evidence for ZFHX4 as an OFC gene [17].

Recent studies have focused mainly on the detection of variants that affect single bases or small indels in candidate genes. In this study, we identified highly penetrant candidate genes for nsCL/P by detecting large de novo copy number variants (CNV) through reanalysis of ES data of 50 patient-parent trios. This revealed a de novo heterozygous 86 kb deletion in ZFHX4. We used a targeted-sequencing approach to evaluate the role of ZFHX4 in an independent cohort of individuals with nsCL/P. Further, we characterized the effects of zfhx4 disruption on embryonic development of the craniofacial region in zebrafish larvae (zfl). Finally, to evaluate the clinical relevance of our findings, we compiled data from an unpublished syndromic patient and from literature searches and publicly available databases.

Materials and methods

Patient-parent trio dataset

The patient-parent trio dataset included ES data of 50 patients with nsCL/P and their unaffected parents of Central European ancestry, as described by Ishorst et al. [15].

CNV calling and filtering

CNV calling from ES BAM files (Fig. 1A) was performed using three read-count based CNV detection tools: XHMM (eXome hidden Markov model) [18], CoNIFER (copy number inference from exome reads) [19], and EXCAVATOR2 [20]. For the latter, parental samples were pooled and utilized for read-count normalization and CNVs were called from patients only.

Fig. 1. Workflow of the CNV study and additional individuals/families with ZFHX4 variants.

Fig. 1

A Workflow of the CNV study. CNVs were called from an existing dataset including exome sequencing data for 50 nsCL/P trios (Ishorst et al. 2022). CNV filtering was performed as follows. (I) The CNV frequency threshold was set to ≤10% among all 50 nsCL/P trios. (II) De novo CNV calling was performed using PLINK/Seq. (III) Only CNVs that were extracted using either CoNIFER and/or EXCAVATOR2 in addition to XHMM were retained in the analysis. CNVs that (IV) did not span RefSeq Genes, (V) overlapped by >50% with regions of segmental duplication or >80% with genes in difficult to analyze regions (Segmental Dups track, UCSC Genome Browser on Human Feb. 2009 (GRCh37/hg19) Assembly), and (VI) had population frequencies ≥0.01 were excluded. B Pedigree of index trio with heterozygous de novo CNV in ZFHX4. C Exon and protein domain structure of human ZFHX4. Exons are colored in alternating white and black, and positions of the start codon (ATG) and stop codon (TAG) are marked. ZFHX4 is a transcription factor with four homeodomains (orange) and 23 zinc finger domains (blue). Protein domains are positioned in proportion to the corresponding exons. Red bar, heterozygous ZFHX4 de novo deletion in the index patient with nsCLP. P1-P3, position of primer pairs spanning the CNV for qPCR verification; black arrows, positions of additional ZFHX4 variants including loss-of-function variants from targeted-sequencing and a homozygous missense variant from a collaboration with Girisha & colleagues. D Pedigrees of four additional families (targeted-sequencing study and collaboration with Girisha & colleagues). E Tyr653 is one out of seven conserved residues (drawn bold) in the fold of a canonical zinc-finger motif. F Model of the zinc-finger structure in ZFHX4 proposes the formation of a hydrogen bond between Tyr653 and His662. Mutation of Tyr653 to histidine could change this conformational arrangement of side chains and thus impair zinc-binding. CNV copy number variation, nsCL/P nonsyndromic cleft lip with/without cleft palate, qPCR quantitative polymerase chain reaction, ES exome sequencing.

XHMM was used as the primary algorithm and, with minor changes, filtering recommendations given in basic protocol 3 of the XHMM manuscript were followed [21] (Fig. 1A, I-III). Common XHMM calls with frequencies of >10% among all nsCL/P trios were excluded. CNVs that overlapped by more than 80% were considered the same CNV. Next, de novo CNV calling on XHMM calls was performed using PLINK/Seq (quality score threshold SQ = 60). Filtering was continued for CNVs that were also detected by at least one of the secondary algorithms (Fig. 1A IV-VI). The AnnotSV web browser [22] was used for CNV population frequency annotation based on Genome Aggregation Database (gnomAD), DGV, and 1000 Genomes. For all variants, plausibility was visually inspected using the Integrative Genomics Viewer (IGV) v.2.3.98 [23].

CNV validation with quantitative PCR

The identified CNV was experimentally validated using quantitative polymerase chain reaction (qPCR). Utilizing Primer3 version 4.1.0, three primer pairs were designed with product sizes of 120 to 150 bp covering the 5′- and 3′-end and the center of the CNV. Two control primer pairs spanning the human housekeeping genes basonuclin 1 (BNC1) and cystic fibrosis transmembrane conductance factor (CFTR) were included (for primer sequences, refer to Table S1). Light Cycler® SYBR Green I Master was used for relative quantification in triplicates on a Light Cycler® 480 (both: Roche, Basel, Switzerland). Cycle threshold values (CT) had to be < 35 to be included. If a triplicate CT standard deviation exceeded 0.2, the most divergent value was excluded from analyses. For copy number determination, we applied the ∆∆CT method [24]. BNC1 and CFTR were used for normalization. The DNAs of two randomly allocated unaffected parents from the patient-parent ES cohort not carrying the CNV of interest according to the three CNV calling algorithms were used as references.

Targeted-sequencing

The targeted-sequencing cohort included 710 individuals with nsCL/P from a Central European cohort from Bonn (Germany), 229 individuals with nsCL/P from the AGORA cohort (Netherlands), and 845 population-matched control individuals [25, 26]. All individuals were phenotypically well characterized. ZFHX4 was part of a larger targeted-sequencing panel of 14 genes evaluated using single-molecule molecular inversion probes (smMIPs) (unpublished). In short, the smMIP assay design was performed using an in-house version of MIPgen, as described previously [15]. Library preparation followed standard procedures with minor modifications [15]. Final libraries were sequenced on the Illumina NovaSeq6000 platform (S2, 2 × 150 bp). Base call files were demultiplexed and converted into FASTQ files using bcl2fastq. Reads were aligned to GRCh37/hg19 using BWA-MEM. Trimming and collapsing of reads were performed using MIPgen scripts as described elsewhere [15]. Collapsing reads that originate from the same starting molecule (identified by a molecular tag) into a single strand consensus sequence allows correction for errors introduced by PCR amplification. The coverages per sample and per exon were calculated using samtools 0.1.19. Samples with an average collapsed coverage of <50× and exons with an average coverage of <20× were excluded. Variant calling was performed using UnifiedGenotyper and annotations were done using Ensembl Variant Predictor (VEP). The gnomAD was used to assign the population frequencies of variants. When DNA was available, the de novo status was determined by Sanger sequencing.

ZFHX4 protein structure modeling

The protein structure of human ZFHX4 (UniProt accession code Q86UP3; 3567 aa) was modeled with AlphaFold3, adding 20 zinc atoms as optional ligands [27]. Protein graphics were generated using the PyMOL Molecular Graphics System (Version 2.5.5 Schrödinger, LLC).

Rare variant association testing for ZFHX4

Rare variant association for ZFHX4 was performed using SKAT-O (SKAT package v2.0.0) suitable for gene-based multiple variant tests. For the analysis a genotype matrix, and vectors of binary phenotypes were prepared.

Assessment of ZFHX4 conservation and Zfhx4/zfhx4 expression

Conservation of ZFHX4 at the amino acid level between human, mouse, and zebrafish (zf) was assessed using Clustal Omega from EMBL-EBI [28]. Zfhx4 expression was inspected in two mouse RNA-Seq datasets representing the period of craniofacial development. A bulk RNA-Seq dataset from murine secondary palatal shelves (E10.5 to E14.5) (BioStudies accession number E-MTAB-3157) and single-cell RNA-Seq (scRNA-Seq) datasets from whole mouse embryos from E9.5 to E13.5 and mouse embryonic facial tissues at E11.5 were analyzed [29]. Further, zfhx4 expression was evaluated using a publicly available in situ hybridization dataset and a scRNA-Seq Atlas of zfl development (Zebrahub) at 30 to 48 h post-fertilization and 1 to 3 days post-fertilization (dpf), representing the zfl period of craniofacial development [30, 31]. Finally, we compared the expression of Zfhx4/zfhx4 with mouse/zebrafish (zf) orthologues of three established OFC genes, CDH1, IRF6, and GRHL3 [13, 32, 33].

Zebrafish husbandry and embryo maintenance

Danio rerio (zebrafish, zf) were kept according to national laws and recommendations by Westerfield [34]. Zfl of the wild-type AB/TL strain acquired by natural fish spawning in the morning and raised at 28 °C in Danieau (30%) medium on a 14 h light to 10 h dark cycle were used for all experiments. To inhibit pigmentation, zfl were treated with PTU (1-phenyl-2-thiourea, final concentration 0.003% in Danieau) from 1 dpf. All experimental groups were kept at the same density, temperature/light conditions and medium change cycles. All fish experiments were carried out before 5 dpf.

Microinjections for morpholino oligonucleotide zfhx4 knockdown (MO-KD)

A morpholino oligonucleotide (GeneTools, LLC, Philomath, OR, USA) targeting the zfhx4 translational start site was used for the specific knockdown of all three transcripts (MO-KD). Zebrafish eggs were collected within 20 min after breeding and the yolk of one-cell staged embryos was pressure injected with 6 ng of MO-KD (1.7 nl/embryo) and 6 ng of standard control MO (Ctrl-MO). Uninjected embryos (UIMO) were kept as controls.

Microinjections for zfhx4 CRISPR/Cas9 F0-knockout (F0-KO)

All substances were ordered from Integrated DNA Technologies (IDT, Leuven, Belgium). Five crRNAs targeting exon 3 of zfhx4 were designed using CRISPRscan [35] and equal amounts of each crRNA were combined to obtain a 100 μM crRNA stock. Then, 3 μl of crRNA stock and 3 μl of tracrRNA (100 μM) were diluted in 44 μl of nuclease-free IDT duplex buffer and annealed to form gRNAs at 95 °C for 5 min. gRNAs (6 μl) were incubated with equal volumes of Cas9 protein (1 μg/μl, diluted in PBS) at 37 °C for 10 min. Once cooled to room temperature, 1 μl of Phenol red was added. As a negative control, a mixture of three crRNAs not matching any genomic locus (Scrambled control, Ctrl-Scr) was prepared as described above [36]. Then, ~1.7 nl Cas9/gRNA/phenol red mix was pressure injected per one-cell staged zebrafish embryo (see Table S1 for gRNA sequences).

DNA extraction and PCR

To determine the efficiency of F0-KO, DNA was isolated from 10–15 zfl at 4 dpf from (i) zfl injected with the F0-KO mix, (ii) zfl injected with the Ctrl-Scr mix, and (iii) uninjected zfl (UICRISPR). DNA was isolated following Meeker et al. [37] using 30 μl of 50 mM NaOH per zfl. PCR was performed using 2 μl of DNA. PCR primers were designed to bind to introns adjacent to exon 3 and to amplify a product of 720 bp (see Table S1 for primer sequences).

Protein isolation and concentration measurement

Protein was isolated at 3 dpf. Zfl from six groups (F0-KO, Ctrl-Scr, UICRISPR, MO-KD, Ctrl-MO, UIMO) were collected and decapitated. Then, 80–100 zfl heads per group were pooled in RIPA Lysis and Extraction Buffer (Thermo Fisher Scientific, Bremen, Germany) with 1× concentrated Protease inhibitor (Pierce Inhibitor Cocktail Tablets; Roche) (1 µl of RIPA buffer/zfl head) and frozen on dry ice. After the tissue was ruptured mechanically, the suspension was incubated for 2 h on a roller incubator at 4 °C and centrifuged at 13,000 rpm for 20 min at 4 °C. The supernatant was stored at –80 °C. The concentration of protein lysates was measured using the Pierce BCA Protein Assay Kit (Thermo Fisher Scientific).

Peptide preparation for liquid chromatography mass spectrometry (LC-MS)

Protein solutions were processed using the SP3-approach [38]. Peptides (10 µg) were further desalted with C18 ZipTips (Merck Millipore, Darmstadt, Germany) to ensure the complete removal of beads.

Directional measurement of Zfhx4 using LC-MS

All chemicals were obtained from Sigma (Taufkirchen, Germany) unless otherwise noted. Dried peptides were dissolved in 10 µl of 0.1% formic acid including retention standards. Peptide separation was performed on a Dionex Ultimate 3000 RSLC nano HPLC system (Dionex GmbH, Idstein, Germany) coupled to an Orbitrap Fusion Lumos mass spectrometer (Thermo Fisher Scientific). Peptides were injected onto a C18 analytical column (400 mm length, 100 µm inner diameter, ReproSil-Pur 120 C18-AQ, 3 µm).

The samples were analyzed first by a standard data-dependent acquisition (DDA) method. Peptides were separated using a linear gradient from 5% to 35% solvent B (90% acetonitrile, 0.1% FA) at 300 nl/min within 60 min. Scans were performed between 330 and 1600 m/z (standard gain control and injection time settings); ions were subjected to higher-energy collision-induced dissociation (HCD: 1.0 Da isolation, threshold intensity 25,000, collision energy 28%) and fragments were analyzed in the Orbitrap.

Fourteen specific peptides were selected for targeted analyses of Zfhx4, Zfhx3 (paralog of Zfhx4), and 60S ribosomal protein L4 (Rpl4; loading control) based on detection in DDA mode (see Table S1 for peptide sequences). Peptides were separated during a 120 min gradient. MS1 spectra were acquired every 3 s. Target ions were subjected to HCD fragmentation (1.6 Da window) and product ions were analyzed in the Orbitrap with a maximum injection time of 100 ms.

LC-MS data analysis

Raw processing of DDA data and database searches were performed using Proteome Discoverer 2.5.0.400 (Thermo Fisher Scientific). Peptides were identified using Mascot server version 2.8.1 (Matrix Science Ltd., London, UK) against the UniProt reference proteome for Danio rerio (as of 04/12/23) and a collection of common contaminants [39]. For details, refer to the Supplementary Materials and Methods.

Data from targeted measurements were analyzed using Skyline [40]. Validation of MS2 spectra was aided by a spectral library created on the PROSIT server [41]. Protein quantification was performed at the MS2 level. Zfhx3 and Zfhx4 levels were normalized against the Rpl4 abundance in each sample.

Cartilage staining

At 4 dpf, zfl were fixed and cartilage-stained with Alcian Blue according to Walker & Kimmel [42].

Imaging and morphometric measurements of zfl

Zfl imaging and Meckel’s-palatoquadrate angle (M-PQ) [43] and ethmoidal plate and Meckel’s cartilage lengths were measured using a ZEISS Stemi 508 stereomicroscope with a Nikon DSFi2 camera and NIS Elements.

Statistical analyses

Craniofacial phenotypes and Zfhx4 fold changes detected by LC-MS were compared among groups using two-way analysis of variance (ANOVA). Survival was compared using Kaplan-Meier survival curves. The M-PQ angle was compared using two-sided t-tests. GraphPad Prism 9 was used for analyses and to generate graphs. Values of p ≤ 0.05 were considered significant.

Literature and database searches

A search for patients carrying ZFHX4 variants was performed against PubMed using the search terms “ZFHX4” and “ZFHX4 orofacial cleft” and against the DECIPHER database for ZFHX4.

Web resources

Please refer to Supplementary Materials and Methods.

Results

CNV calling and filtering

Among the 50 nsCL/P patients, XHMM (our primary calling algorithm) detected 1277 CNVs and 733 were retained after the elimination of common CNVs. Of those, 36 CNVs were not detected in the respective parents and were identified as de novo. Eleven CNVs were also called using either CoNIFER and/or EXCAVATOR2. Ultimately, after excluding CNVs that did not meet additional filtering criteria described in Fig. 1A IV-VI, a heterozygous 86 kb deletion affecting exons 4–11 of ZFHX4 remained and its de novo status was confirmed (Fig. 1A–C). The CNV (or a comparable CNV with significant overlap) was absent from DGV.

Additional variants identified by targeted-sequencing and clinical exome data

Post-collapsing, the coverage of ZFHX4 was 587×. While no truncating variants were observed in the controls, targeted-sequencing revealed three truncating variants in patients, two frameshift variants leading to a premature stop, and a splice donor variant (Fig. 1D, Table 1). One of three truncating variants was identified as de novo. The segregation status of the other two variants could not be investigated owing to lack of parental DNA. The rare variant association test in the targeted-sequencing cohort revealed an association of nsCL/P with truncating variants in ZFHX4 (p = 0.01575). In addition, a homozygous ZFHX4 missense variant was identified by a clinical exome analysis of an individual with CL/P, microcephaly, and micrognathia born to unaffected parents carrying the variant in the heterozygous state (Table 1). This variant (NM_024721.4:c.1957T>C, NP_078997.4:Tyr653His) is located in a canonical zinc-finger motif of ZFHX4 which allowed to model its proposed conformation with high confidence [44] (Fig. 1E). Modeling of the zinc-finger structure showed that the hydroxyl group of the Tyr653 side chain could form a hydrogen bond with the imidazole ring of His662, which is part of the C2H2-type zinc-finger motif (Cys646, Cys649, His662, His667) that coordinates the metal ligand (Fig. 1F).

Table 1.

Genetic evidence for the role of ZFHX4 in orofacial clefting.

Source Mutation type Affected exon ZFHX4 Nucleotide change (NM_024721.4) a Amino acid change (NP_078997.4) Zygosity De novo (y/n) Size Syndromic (y/n) OFC phenotype Micrognathia (y/n)
Present study CNV, In-frame deletion 4–11 c.3105_10835del p.(Lys1035_Asp3612delinsAsn) HET y 86,33 kb n CL/P n
Present study (Zametica) Frameshift deletion 2 c.1863_1864del p.(Leu622GlyfsTer26) HET na 2 bp n CL/P Retrognathia
Present study (Zametica) SNV, splice donor Intron 9 c.3964+2 T > A p.? HET na 1 bp n CL/P n
Present study (Zametica) SNV, frameshift deletion 2 c.1927del p.(Thr643ProfsTer21) HET y 1 bp n CL/P n
Present study (Girisha) SNV, missense 2 c.1957T > C p.(Tyr653His) HOM n 1 bp y CL/P y
Palomares et al. [45] Deletion, start loss c.−3231464_*6395221del p.Met1? HET y 9.80 Mb y Highly arched palate y
Palomares et al. [45] Deletion, start loss c.−2993631_*2514645del p.Met1? HET y 5.69 Mb y Highly arched palate y
Palomares et al. [45] Deletion, start loss c.−495025_7082del p.Met1? HET n 0.66 Mb y CPb n
Bishop et al. [14] SNV, frameshift deletion 10 c.5595del p.(Glu1866LysfsTer8) HET y 1 bp n CL/P Not known
Bishop et al. [14] SNV, stopgain 10 c.7990 C > T p.(Gln2664Ter) HET y 1 bp n CL/P Not known
Fontana et al. [46] SNV, splice donor Intron 3 c.3093+1 G > T p.? HET y 1 bp y Highly arched palate y
Créton et al. [47] SNV, frameshift deletion 2 c.2513del p.(Asn838ThrfsTer7) HET y 1 bp y CL/P yc
Sorrentino et al. [48] Frameshift deletion-insertion 9 c.3943_3945delinsAA p.(Glu1315LysfsTer29) HET y 2 bp nd CL/P n
DECIPHER v11.13, Patient 412653 SNV, missense 10 c.8882 T > C p.(Met2961Thr) HET y 1 bp y CP n
DECIPHER v11.13, Patient 284919 SNV, stopgain 2 c.2041 C > T p.(Gln681Ter) HET ne 1 bp y CL/P n
DECIPHER v11.13, Patient 268410 SNV, missense 5 c.3326 A > T p.(Glu1109Val) HET n 1 bp y Highly arched palate y

na not available, y yes, n no.

aAll variants are absent from GnomAD v2.1.1.

bIndividual from Belligni et al. 2010. In this study, four additional family members with syndromal phenotypes are reported, including CPO (maternal grandmother and sister of maternal grandmother) and nasal voice (mother and maternal great-grandmother).

cAlso in the mother without a ZFHX4 mutation.

dThe pregnancy was terminated after an orofacial cleft was detected on ultrasound examination. The status was apparently nonsyndromic. Postmortem examination of the fetus confirmed the absence of other major congenital anomalies. However, other syndromic features that could not be assessed postmortem, such as developmental delay or mental retardation, could not be completely excluded.

eInherited from the mother with CL/P (unilateral cleft lip).

Literature search of ZFHX4 variants in individuals with orofacial clefting

PubMed and DECIPHER database searches revealed nine individuals [14, 4548] with syndromic/nonsyndromic forms of orofacial clefts carrying ZFHX4 variants or deletions affecting ZFHX4 (Table 1).

Zfhx4/zfhx4 expression in mouse and zf during craniofacial development

At the amino acid level we found 92% conservation between human and mouse ZFHX4 and 72% between human and zf (Fig. S1). Using published RNA-Seq datasets for mouse palatal shelves, expression levels of Zfhx4 were higher than those of other OFC genes (CDH1, IRF6, and GRHL3) at all five timepoints examined (Fig. 2A, Fig. S2). Grhl3 showed the lowest expression levels among the four genes (maximum value: Grhl3E13.5 = 100 RPKM).

Fig. 2. Expression pattern of Zfhx4 in mouse embryos.

Fig. 2

A Heatmap of mean replicates in Reads Per Kilobase Million (RPKM) of Zfhx4 and the established orofacial clefting genes (Cdh1, Irf6, and Grhl3) from bulk RNA-Seq data for mouse secondary palatal shelves (embryonic days E10.5–E14.5). Dot plots of gene expression from single-cell RNA-Seq data for the B whole mouse embryo and C mouse embryo face at E11.5. The color of the dots corresponds to the average scaled expression level. The size of the dots corresponds to the percentage of cells that expressed the gene in the respective cell type.

Zfhx4 was expressed in many cell types, especially in neuronal cells, chondrocytes, and connective tissue progenitors, based on scRNA-Seq data for whole mouse embryos at E11.5 (for more stages refer to Fig. S3). The other three OFC genes were mainly expressed in epithelial cells (Fig. 2B). ScRNA-Seq data from mouse face at E11.5 confirmed these findings. Whereas the established OFC genes were mainly expressed in peridermal/epithelial cells, Zfhx4 showed a broad expression pattern (Fig. 2C). In situ hybridization and scRNA-Seq data from different timepoints of zf development supported these findings (Fig. S4S7).

zfhx4 F0-KO and MO-KD zfl present with craniofacial anomalies

To assess the role of zfhx4 in zfl development, we generated F0-KO and MO-KD and performed cartilage staining at 4 dpf. Controls (UICRISPR, UIMO, Ctrl-Scr, and Ctrl-MO zfl) generally showed normal physiological development, including craniofacial development (Fig. 3A upper two panels). Zfl injected with the F0-KO mix and MO-KD resulted in anomalies with various degrees of severity and penetrance (Fig. 3A lower three panels, C). Survival rates did not differ significantly between F0-KO and Ctrl-Scr groups (p = 0.5417, Mantel-Cox test) but were slightly lower in MO-KD than in the Ctrl-MO group (p ≤ 0.01, Mantel-Cox test) (Fig. 3B). The penetrance of craniofacial anomalies was higher in the MO-KD group than in the F0-KO group (Fig. 3C). We observed relevant phenotypes, such as a reduced head size, shortened and misshaped jaw, and ethmoidal plate structures, in 19% of F0-KO zfl compared with 3% of Ctrl-Scr zfl (p ≤ 0.01, two-way ANOVA) and in 38% of MO-KD zfl compared with 0% of Ctrl-MO zfl (p ≤ 0.001, two-way ANOVA). The phenotypes described are similar to those seen in other studies disrupting OFC genes in zfl [49]. Zfl with a severe craniofacial phenotype may also present with reduced bodylength (Fig. 3A).

Fig. 3. Effect of F0-generation knock-out (F0-KO) and translation-blocking morpholino knock-down (MO-KD) of zfhx4 in zebrafish larvae (zfl).

Fig. 3

A Ventral view of whole-mount Alcian blue-stained zfl at 4 dpf. For F0-KO (left) and MO-KD (right), the spectrum of the phenotypes is shown in blue (– = no phenotype, + = mild phenotype, ++ = strong phenotype). Scale bar = 113 µm. B Survival curves for each group (N = 3). Survival of F0-KO did not differ significantly from that of the Ctrl-Scr group, whereas survival was lower in MO-KD than in Ctrl-MO. Comparisons were performed using Mantel-Cox tests. Survival rates at 4 dpf: UICRISPR: 81.1%, Ctrl-Scr: 77.2%, F0-KO: 75.7%, UIMO: 89.6%, Ctrl-MO: 84.4%, MO-KD: 78.1%. C Percentages of surviving zfl at 4 dpf with craniofacial anomalies (phenotype) and no phenotype in different groups (N = 3). No distinction between phenotype severities was made. F0-KO and MO-KD injection resulted in craniofacial anomalies in 19.13% and 38.17% of zfl, respectively, compared with rates of anomalies of 3.09% in Ctrl-Scr zfl and 0% in UICRISPR/MO and Ctrl-MO zfl. Comparisons were performed using two-way analysis of variance (ANOVA) and Tukey’s multiple comparisons tests. Data are presented as means with standard error of the mean (SEM). D PCR validation of the disruption of zfhx4 exon 3 in F0-KO zfl at 4 dpf. Primer binding sites are located on adjacent introns. Expected wt band size, 720 bp. E Liquid chromatography mass spectrometry (LC-MS) analysis of protein lysates from heads of 3 dpf zfl (N = 2). Depicted are Zfhx4 fold changes relative to the 60S ribosomal protein L4 (Rpl4) abundance in each sample. Zfhx4 levels were decreased by 77.1% in F0-KO compared with levels in Ctrl-Scr and by 41.6% in MO-KD compared with levels in Ctrl-MO. Individual data for LC-MS run1 (stars) and run2 (dots) are plotted. Comparisons were performed using two-way analysis of variance (ANOVA) and Tukey’s multiple comparisons tests. Data are presented as means with standard deviation (SD). ns, p > 0.05, **p ≤ 0.01, ***p ≤ 0.001; dpf days post-fertilization, UICRISPR/MO uninjected wild-type zfl, Ctrl-MO Control MO-injected zfl, Ctrl-Scr Scrambled control-injected zfl, wt wild-type.

Genotyping revealed efficient mutagenesis of zfl (Fig. 3D). LC-MS analysis of protein lysates of zfl at 3 dpf demonstrated that Zfhx4 levels were 77.1% lower in F0-KO than in Ctrl-Scr zfl (p ≤ 0.001, two-way ANOVA) and 41.6% lower in MO-KD than in Ctrl-MO zfl (p ≤ 0.01, two-way ANOVA) (Fig. 3E), while no significant reduction in the Zfhx4 paralog Zfhx3 was detected (Fig. S8).

To further evaluate the consequences of zfhx4 disruption on craniofacial development, we specifically assessed the ethmoid (homolog of the human palate; Fig. 4A, B) and Meckel’s cartilage (Fig. 4C) in zfl at 4 dpf at higher magnification (Fig. 4D, E). There were no differences in parameter values among UICRISPR, UIMO, Ctrl-Scr, and Ctrl-MO zfl. F0-KO and MO-KD zfl exhibited shorter Meckel’s cartilage and reduced ethmoid plate lengths (Fig. 4D, E).

Fig. 4. Craniofacial outcomes of zfhx4 F0-generation knock-out (F0-KO) and translation-blocking morpholino knock-down (MO-KD) in zebrafish larvae (zfl).

Fig. 4

A Schematic overview of the human primary (green) and secondary palate (light blue) (ventral view). B Schematic overview of the zfl neurocranium with medial (green) and lateral ethmoid (light blue) (ventral view). Both structures are homologous structures to the human palate as indicated by the corresponding colors. C Ventral view of the zfl cartilage structures of the neurocranium (green and light blue) and viscerocranium (dark blue). The eyes are outlined in light gray. D Ventral view of the craniofacial region of whole-mount Alcian blue stained zfl at 4 dpf as represented schematically in C (for a different zfl than that shown in Fig. 3A). Comparison of craniofacial phenotypes between F0-KO zfl against controls (UICRISPR and Ctrl-Scr). Scale bar = 200 µm. E Ventral view of the craniofacial region of whole-mount Alcian blue-stained zfl at 4 dpf as represented schematically in C. Comparison of craniofacial phenotypes between MO-KD-injected zfl against controls (UIMO and Ctrl-MO). Scale bar = 200 µm. F Length of Meckel’s cartilage and ethmoid plate measured as indicated by the pink lines of 4 dpf old, Alcian blue-stained F0-KO- and Ctrl-Scr-injected zfl (mean length Meckel’s: F0-KO: 96.9 μm, Ctrl-Scr: 107.7 μm, difference between means: 10.8 ± 5.4 μm; mean length ethmoid: F0-KO: 103.4 μm, Ctrl-Scr: 108.9 μm, difference between means 5.5 ± 3.1 μm) and MO-KD- and Ctrl-MO-injected zfl (mean length Meckel’s: MO-KD: 79.5 μm, Ctrl-MO: 102.9 μm, difference between means: 23.5 ± 6.3 μm; mean length ethmoid: MO-KD: 91.1 μm, Ctrl-MO: 101.5 μm, difference between means: 10.4 ± 3.2 μm). Comparisons were performed using unpaired, two-tailed Welch’s t tests. G Measurement of the Meckel’s-palatoquadrate (M-PQ) angle as represented by the pink angle, which is a reliable parameter to assess craniofacial outcomes as suggested by Raterman et al. [43]. Comparison of M-PQ angles in 4 dpf old, Alcian blue-stained F0-KO- and Ctrl-Scr-injected zfl (mean M-PQ angle: F0-KO: 45.6°, Ctrl-Scr: 43.1°, difference between means 2.5° ± 1.1°) and MO-KD- and Ctrl-MO-injected zfl (mean M-PQ angle: MO-KD: 58.8°, Ctrl-MO: 46.4°, difference between means 12.5° ± 2.0°). Comparisons were performed using unpaired, two-tailed Student’s t tests. ns, p > 0.05, *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001. m Meckel’s, pq Palatoquadrate, ch Ceratohyal, hs Hyosympletic, dpf days post-fertilization, UICRISPR/MO uninjected wild-type zfl, Ctrl-MO Control MO-injected zfl, Ctrl-Scr Scrambled control-injected zfl.

We performed a quantitative analysis of Meckel’s cartilage and ethmoid plate lengths (Fig. 4F) and the M-PQ angle (Fig. 4G) using randomly chosen zfl within each group (without considering the presence or severity of craniofacial phenotypes). Meckel’s cartilage length, measured from the level of the interpupillary distance, was significantly shorter in F0-KO zfl (mean 96.9 μm) than in Ctrl-Scr zfl (mean 107.7 μm) (p ≤ 0.05, unpaired, two-tailed t test). A stronger effect was observed in MO-KD zfl (mean length: MO-KD: 79.5 μm, Ctrl-MO: 102.9 μm; p ≤ 0.001, unpaired, two-tailed t test) (Fig. 4F). The ethmoid plate length, measured from the level of the interpupillary distance, was significantly shorter in MO-KD zfl (mean 91.1 μm) than in Ctrl-MO zfl (mean 101.5 μm) (p ≤ 0.01, unpaired, two-tailed t test), but did not differ significantly between F0-KO (mean 103.4 μm) and Ctrl-Scr zfl (mean 108.9 μm) (p > 0.05, unpaired, two-tailed t test) (Fig. 4F). The M-PQ angle was significantly larger in F0-KO zfl (mean 45.6°) than in Ctrl-Scr zfl (mean 43.1°) (p ≤ 0.05, unpaired, two-tailed t test). The difference was greater between MO-KD (mean 58.8°) and Ctrl-MO zfl (mean 46.4°) (p ≤ 0.001, unpaired, two-tailed t test) (Fig. 4G).

Discussion

We attempted to identify highly penetrant candidate genes for nsCL/P. ES data provided a valid basis for identifying CNVs, although very conservative filtering and validation are required. In particular, we identified a de novo heterozygous 86 kb deletion affecting major parts of ZFHX4, which encodes a transcription factor with 23 zinc finger motifs and four DNA-binding homeodomains (Fig. 1C) [50]. Targeted-sequencing of ZFHX4 in our in-house nsCL/P and the AGORA cohort revealed three additional truncating variants.

To analyze the functions of ZFHX4 in early craniofacial development, we established F0-KO and MO-KD zf models. ZFHX4 was identified as suitable candidate for functional studies in zf, with only one orthologue (zfhx4) and high amino acid conservation (Fig. S1). Zfhx4/zfhx4 was expressed in mouse/zf craniofacial tissues and cell lineages at time points relevant to craniofacial development (Fig. 2, Fig. S3S7).

The disruption (F0-KO) and transient suppression (MO-KD) of zfhx4 resulted in craniofacial changes in zfl at 4 dpf, albeit with differences in penetrance (Figs. 3, 4). These differences could be explained by the ability of MO-KD to bind maternal zfhx4 RNA possibly present in the zf zygote. Since CRISPR acts at the DNA level, such maternal mRNA would not be affected by F0-KO and could lead to a rescue-like effect. However, two studies that analyzed the transcriptome of early zf zygotes, 2-cell stage –9 h post fertilization and 0.75–5.5 h post fertilization, respectively, with different methods, could not detect any significant expression of zfhx4, making the mentioned hypothesis obsolete [51, 52]. Nevertheless, the discrepancies in phenotype penetrance and severity between knockdowns and knockouts have been described and also assessed for some loci [53]. In our opinion, the most probable reason for the discrepancies in phenotype penetrance is the generation of hypomorphic alleles with F0-KO that still lead to functional protein or more complex compensatory mechanisms.

While the initial patient in this study presented with nsCL/P, individuals with ZFHX4 variants or CNVs that disrupt ZFHX4 exhibit both nonsyndromic and syndromic orofacial clefts, including nervous system abnormalities (e.g., developmental delay or intellectual disability), musculoskeletal or limb abnormalities, and craniofacial abnormalities in addition to the cleft such as microcephaly or micrognathia, with the latter being present in the majority of individuals with syndromic forms of clefting (Table 1). Regarding our zf models, the ethmoidal plate, which is structurally homologous to the human palate, was significantly shorter in MO-KD and slightly shorter in F0-KO, and Meckel’s cartilage was significantly shorter in both F0-KO and MO-KD (Fig. 4F). Of note, this may resemble micrognathia in humans [54]. The M-PQ angle, a well-established measure for craniofacial outcomes [43], was larger in F0-KO and MO-KD (Fig. 4G), indicative of micrognathia but also microcephaly [43]. These observations further support the causal role of the gene in craniofacial anomalies and a broader phenotypic spectrum (beyond nsCL/P). This was recently confirmed in a study by Nakamura et al., revealing that Zfhx4-deficient mice died within one day and presented with skeletal abnormalities and a cleft palate resulting from a failure in elevation and fusion of the palatal shelves [17]. Interestingly, mouse and zf orthologues of CDH1, IRF6, and GRHL3, associated with autosomal dominant nonsyndromic or syndromic forms of clefts with apparently nonsyndromic appearance, showed a narrow expression pattern during embryonic development, while mouse and zf orthologues of ZFHX4 were much more widely expressed (Fig. 2, Figure S3S7). This could explain, in part, why ZFHX4 leads to a much wider phenotypic spectrum than just nsCL/P. DECIPHER also lists individuals with ZFHX4 variants or CNVs that disrupt ZFHX4 that have a syndrome without orofacial clefting (not included in Table 1), which may be explained by incomplete penetrance and modifying factors.

Collectively, the data compiled in this study demonstrate that different types of variants, from small missense and truncating variants in ZFHX4 to larger CNVs affecting only ZFHX4 or additional genes, either inherited or acquired de novo, could lead to OFC. The type of cleft caused by ZFHX4 variants or CNVs affecting ZFHX4 is variable, including CL/P and CPO, either nonsyndromic or syndromic. Currently, it appears that large deletions are more likely to result in CPO or a highly arched palate. Of note, there is evidence that CPO and a highly arched palate share genetic risk factors [55]. By contrast, small variants may be more likely to cause CL/P than CPO. Of note, the fact that genetic testing is more likely to be performed for individuals with complex syndromes than individuals with apparently nonsyndromic forms of OFC may lead to a reporting bias towards syndromic forms of OFC.

Regarding the postulated mode of inheritance, all individuals from the literature and DECIPHER, and four of five of the newly presented individuals, had heterozygous variants or CNVs affecting ZFHX4. With a pLI score of 1, ZFHX4 is highly constrained, suggesting that the loss of substantial portions of one allele results in haploinsufficiency. However, one individual presented in this study carried a novel homozygous missense variant in ZFHX4, whereas her parents were heterozygous carriers with no malformations or abnormalities (Table 1, Supplementary Material). This is the first study to present an individual with an apparent recessive inheritance of ZFHX4 variants. The structural modelling of ZFHX4 showed that the respective amino acid change Tyr653His might disrupt the formation of a hydrogen bond with the imidazole ring of His662, which is part of the C2H2-type zinc-finger motif (Fig. 1F). This disruption might alter the structural conformation as well as the ability for zinc-coordination in this part of the protein, both of which could be important for the ability of the transcription factor ZFHX4 to bind DNA. This observation raises the possibility that, in addition to heterozygous ZFHX4 variants, homozygous or biallelic ZFHX4 variants might also contribute to OFC risk.

In conclusion, our results support the role of ZFHX4 in OFC. Variants in ZFHX4 or CNVs can lead to both typical subtypes of OFC − CL/P and CPO. Also, ZFHX4 disruption can lead to both nonsyndromic and syndromic OFC. Based on our findings and recent studies, ZFHX4 variants are associated with an extremely broad phenotypic spectrum, from complex malformation syndromes to nonsyndromic isolated malformations. Accordingly, more data are needed to systematically assess genotype–phenotype relationships and to identify modifying factors. ZFHX4 should be considered a causative factor not only in patients with severe complex syndromes but also in patients with mild syndromic and even nonsyndromic OFC.

Supplementary information

Supplementary Figures (1.5MB, pdf)
Table S1 (533.8KB, pdf)

Acknowledgements

We thank all patients, relatives, and control individuals for their participation. We thank the German support group for individuals with cleft lip and/or palate (Wolfgang Rosenthal Gesellschaft) for assistance with recruitment. We acknowledge the invaluable assistance of all clinical, laboratory and bioinformatics personnel. We are thankful for the support of Jayne Y. Hehir-Kwa (Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands) and Joris A. Veltman (Biosciences Institute, Faculty of Medical Sciences, Newcastle University, UK) with CNV calling from Exome data, Nuria Ortega Ibañez and Hartmut Engels (Institute of Human Genetics, Medical Faculty, University of Bonn, Germany) with CNV validation via qPCR, the NGS Core Facility (Medical Faculty, University of Bonn, Germany) for sequencing the smMIP libraries, the Zebrafish Core Facility (Medical Faculty, University of Bonn, Germany) for the assistance with the zebrafish work, and the Core Facility for Analytical Proteomics (Medical Faculty, University of Bonn, Germany) for Zfhx4 detection in protein lysates. This study made use of data generated by the DECIPHER community. A full list of centers who contributed to the generation of data is available at https://deciphergenomics.org/about/stats and via email from contact@deciphergenomics.org. Funding for the DECIPHER project was provided by Wellcome [grant number WT223718/Z/21/Z] (PMID: 19344873). Finally, the authors thank the Genome Aggregation Database (gnomAD) and all groups that provided exome and genome variant data to this resource. A full list of gnomAD contributors is provided in the gnomAD flagship paper (PMID: 32461654).

Author contributions

Study conception and design: NI, SH, BO, EMa.; Sample collection and clinical data analysis: TK, EO, KMG, SP, SN, CC, IvR, EMa; Genetic data acquisition: NI, SH, CG, FD, DD, BZ, KMG; Functional data acquisition: NI, SH, ÖY, TL, JL, EMi, JCK, KC, DK, AS, MD, GH, MG, KUL; Analysis and interpretation of genetic and zebrafish data: NI, SH, DD, BZ, KUL, BO, EMa; Manuscript drafting and critical revision: all authors.

Funding

The present study was supported by the Else Kröner-Fresenius Foundation (BonnNi grant, Q-614.1254 to S.H.) and by the German Research Foundation (DFG)-Grants BE 3828/8-1, LU 1944/2-1, MA 2546/5-1, and LU1944/3-1. This work was partially supported by the DBT/Wellcome Trust India Alliance Grant titled ‘Center for Rare Disease Diagnosis, Research and Training [Grant number: IA/CRC/20/1/600002]’ awarded to K.M.G. Open Access funding enabled and organized by Projekt DEAL.

Data availability

The data that support the findings of this study are available on request from the corresponding authors. The data are not publicly available due to privacy or ethical restrictions.

Competing interests

For the sake of completeness, we declare that D.D. provides compensated consulting services outside of academia as an independent consultant. Furthermore, K.U.L. is a co-founder of LAMPseq Diagnostics GmbH. Although past and current clients might include biotech, life science, and pharma companies, the services are not related to the presented work and all authors are unaware of any possible competing interests.

Ethical approval

Written informed consent was obtained from all participants, or the respective parents/legal guardians, prior to inclusion. The study was approved by the ethics committee of the Medical Faculty of the University of Bonn (ethics approval number 295/14, last dated June 20th, 2022). Animal husbandry and experimental setups were in accordance with European Legislation for the Protection of Animals used for Scientific Purposes (Directive 2010/62/EU). National law exempts all zebrafish experiments conducted at larval stages up to 5 dpf, before larvae begin to feed, from ethical approval.

Footnotes

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

These authors contributed equally: Nina Ishorst, Selina Hölzel.

These authors jointly supervised this work: Benjamin Odermatt, Elisabeth Mangold.

Contributor Information

Nina Ishorst, Email: nina.ishorst@uni-bonn.de.

Benjamin Odermatt, Email: b.odermatt@uni-bonn.de.

Elisabeth Mangold, Email: e.mangold@uni-bonn.de.

Supplementary information

The online version contains supplementary material available at 10.1038/s41431-024-01775-9.

References

  • 1.Mossey PA, Modell B Epidemiology of Oral Clefts 2012: An International Perspective Part 1: Descriptive Epidemiology for Orofacial Clefts. 2012. [DOI] [PubMed]
  • 2.Mangold E, Ludwig KU, Nöthen MM. Breakthroughs in the genetics of orofacial clefting. Trends Mol Med. 2011;17:725–33. [DOI] [PubMed] [Google Scholar]
  • 3.Marazita ML. The evolution of human genetic studies of cleft lip and cleft palate. Annu Rev Genomics Hum Genet. 2012;13:263–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Yu Y, Zuo X, He M, Gao J, Fu Y, Qin C, et al. Genome-wide analyses of non-syndromic cleft lip with palate identify 14 novel loci and genetic heterogeneity. Nat Commun. 2017;8:14364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Leslie EJ, Carlson JC, Shaffer JR, Butali A, Buxó CJ, Castilla EE, et al. Genome-wide meta-analyses of nonsyndromic orofacial clefts identify novel associations between FOXE1 and all orofacial clefts, and TP63 and cleft lip with or without cleft palate. Hum Genet. 2017;136:275–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ludwig KU, Böhmer AC, Bowes J, Nikolić M, Ishorst N, Wyatt N, et al. Imputation of orofacial clefting data identifies novel risk loci and sheds light on the genetic background of cleft lip ± cleft palate and cleft palate only. Hum Mol Genet. 2017;26:ddx012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Welzenbach J, Hammond NL, Nikolić M, Thieme F, Ishorst N, Leslie EJ, et al. Integrative approaches generate insights into the architecture of non-syndromic cleft lip with or without cleft palate. Hum Genet Genomics Adv. 2021;2:100038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Khandelwal KD, Ishorst N, Zhou H, Ludwig KU, Venselaar H, Gilissen C, et al. Novel IRF6 mutations detected in orofacial cleft patients by targeted massively parallel sequencing. J Dent Res. 2017;96:179–85. [DOI] [PubMed] [Google Scholar]
  • 9.Basha M, Demeer B, Revencu N, Helaers R, Theys S, Bou Saba S, 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;55:449–58. [DOI] [PubMed] [Google Scholar]
  • 10.Savastano CP, Brito LA, Faria ÁC, Setó-Salvia N, Peskett E, Musso CM, et al. Impact of rare variants in ARHGAP29 to the etiology of oral clefts: role of loss-of-function vs missense variants. Clin Genet. 2017;91:683–9. [DOI] [PubMed] [Google Scholar]
  • 11.Leslie EJ, Taub MA, Liu H, Steinberg KM, Koboldt DC, Zhang Q, et al. Identification of functional variants for cleft lip with or without cleft palate in or near PAX7, FGFR2, and NOG by targeted sequencing of GWAS loci. Am J Hum Genet. 2015;96:397–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Marini NJ, Asrani K, Yang W, Rine J, Shaw GM. 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:ajmg.a.61183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cox LL, Cox TC, Moreno Uribe LM, Zhu Y, Richter CT, Nidey N, et al. Mutations in the epithelial cadherin-p120-catenin complex cause mendelian non-syndromic cleft lip with or without cleft palate. Am J Hum Genet. 2018;102:1143–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Bishop MR, Diaz Perez KK, Sun M, Ho S, Chopra P, Mukhopadhyay N, et al. Genome-wide enrichment of de novo coding mutations in orofacial cleft trios. Am J Hum Genet. 2020;107:124–36. [DOI] [PMC free article] [PubMed]
  • 15.Ishorst N, Henschel L, Thieme F, Drichel D, Sivalingam S, Mehrem SL, et al. Identification of de novo variants in nonsyndromic cleft lip with/without cleft palate patients with low polygenic risk scores. Mol Genet Genom Med. 2023;11:e2109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zieger HK, Weinhold L, Schmidt A, Holtgrewe M, Juranek SA, Siewert A, et al. Prioritization of non-coding elements involved in non-syndromic cleft lip with/without cleft palate through genome-wide analysis of de novo mutations. HGG Adv. 2023;4:100166. [DOI] [PMC free article] [PubMed]
  • 17.Nakamura E, Hata K, Takahata Y, Kurosaka H, Abe M, Abe T, et al. Zfhx4 regulates endochondral ossification as the transcriptional platform of Osterix in mice. Commun Biol. 2021;4:1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Fromer M, Moran JL, Chambert K, Banks E, Bergen SE, Ruderfer DM, et al. Discovery and statistical genotyping of copy-number variation from whole-exome sequencing depth. Am J Hum Genet. 2012;91:597–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Krumm N, Sudmant PH, Ko A, O’Roak BJ, Malig M, Coe BP, et al. Copy number variation detection and genotyping from exome sequence data. Genome Res. 2012;22:1525–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.D’Aurizio R, Pippucci T, Tattini L, Giusti B, Pellegrini M, Magi A. Enhanced copy number variants detection from whole-exome sequencing data using EXCAVATOR2. Nucleic Acids Res. 2016;44:e154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fromer M, Purcell SM Using XHMM software to detect copy number variation in whole-exome sequencing data. Curr Protoc Hum Genet. 2014. 10.1002/0471142905.hg0723s81. [DOI] [PMC free article] [PubMed]
  • 22.Geoffroy V, Herenger Y, Kress A, Stoetzel C, Piton A, Dollfus H, et al. AnnotSV: an integrated tool for structural variations annotation. Bioinformatics. 2018;34:3572–4. [DOI] [PubMed] [Google Scholar]
  • 23.Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol 2011;29:24–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 2001;25:402–8. [DOI] [PubMed] [Google Scholar]
  • 25.Mangold E, Ludwig KU, Birnbaum S, Baluardo C, Ferrian M, Herms S, et al. Genome-wide association study identifies two susceptibility loci for nonsyndromic cleft lip with or without cleft palate. Nat Genet. 2010;42:24–6. [DOI] [PubMed] [Google Scholar]
  • 26.Van Rooij IALM, Ludwig KU, Welzenbach J, Ishorst N, Thonissen M, Galesloot TE, et al. Non-Syndromic Cleft Lip with or without Cleft Palate: Genome-Wide Association Study in Europeans Identifies a Suggestive Risk Locus at 16p12.1 and Supports SH3PXD2A as a Clefting Susceptibility Gene. Genes (Basel). 2019;10:1023. [DOI] [PMC free article] [PubMed]
  • 27.Abramson J, Adler J, Dunger J, Evans R, Green T, Pritzel A, et al. Accurate structure prediction of biomolecular interactions with AlphaFold 3. Nature. 2024;630:493–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Madeira F, Pearce M, Tivey AR, Basutkar P, Lee J, et al. Search and sequence analysis tools services from EMBL-EBI in 2022. Nucleic Acids Res. 2022;50:W276–9. [DOI] [PMC free article] [PubMed]
  • 29.Siewert A, Reiz B, Krug C, Heggemann J, Mangold E, Dickten H, et al. Analysis of candidate genes for cleft lip ± cleft palate using murine single-cell expression data. Front Cell Dev Biol. 2023;11:1091666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Thisse B, Thisse C. Fast release clones: a high throughput expression analysis. ZFIN Direct Data Submission. 2004 http://zfin.org.
  • 31.Lange M, Granados A, VijayKumar S, Bragantini J, Ancheta S, Santhosh S et al. Zebrahub – multimodal zebrafish developmental atlas reveals the state transition dynamics of late vertebrate pluripotent axial progenitors. bioRxiv 2023; 2023.03.06.531398. [DOI] [PubMed]
  • 32.Birnbaum S, Ludwig KU, Reutter H, Herms S, de Assis NA, Diaz-Lacava A, et al. IRF6 gene variants in Central European patients with non-syndromic cleft lip with or without cleft palate. Eur J Oral Sci. 2009;117:766–9. [DOI] [PubMed] [Google Scholar]
  • 33.Mangold E, Böhmer AC, Ishorst N, Hoebel A-K, Gültepe P, Schuenke H, 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;98:755–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Westerfield M The Zebrafish Book. A Guide for the Laboratory Use of Zebrafish (Danio rerio). 4th ed. 2000.
  • 35.Moreno-Mateos MA, Vejnar CE, Beaudoin JD, Fernandez JP, Mis EK, Khokha MK, et al. CRISPRscan: designing highly efficient sgRNAs for CRISPR-Cas9 targeting in vivo. Nat Methods 2015 1210. 2015;12:982–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kroll F, Powell GT, Ghosh M, Gestri G, Antinucci P, Hearn TJ, et al. A simple and effective f0 knockout method for rapid screening of behaviour and other complex phenotypes. Elife. 2021;10:1–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Meeker ND, Hutchinson SA, Ho L, Trede NS. Method for isolation of PCR-ready genomic DNA from zebrafish tissues. Biotechniques. 2007;43:610–4. [DOI] [PubMed] [Google Scholar]
  • 38.Hughes CS, Moggridge S, Müller T, Sorensen PH, Morin GB, Krijgsveld J. Single-pot, solid-phase-enhanced sample preparation for proteomics experiments. Nat Protoc. 2019;14:68–85. [DOI] [PubMed] [Google Scholar]
  • 39.Frankenfield AM, Ni J, Ahmed M, Hao L. Protein contaminants matter: building universal protein contaminant libraries for DDA and DIA proteomics. J Proteome Res. 2022;21:2104–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Pino LK, Searle BC, Bollinger JG, Nunn B, MacLean B, MacCoss MJ. The Skyline ecosystem: Informatics for quantitative mass spectrometry proteomics. Mass Spectrom Rev. 2020;39:229–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Gessulat S, Schmidt T, Zolg DP, Samaras P, Schnatbaum K, Zerweck J, et al. Prosit: proteome-wide prediction of peptide tandem mass spectra by deep learning. Nat Methods. 2019;16:509–18. [DOI] [PubMed] [Google Scholar]
  • 42.Walker MBM, Kimmel CBC. A two-color acid-free cartilage and bone stain for zebrafish larvae. Biotech Histochem. 2007;82:23–28. [DOI] [PubMed] [Google Scholar]
  • 43.Raterman ST, Metz JR, Wagener FADTG, Von den Hoff JW. Zebrafish models of craniofacial malformations: interactions of environmental factors. Front Cell Dev Biol. 2020;8:600926. [DOI] [PMC free article] [PubMed]
  • 44.Klug A. The discovery of zinc fingers and their applications in gene regulation and genome manipulation. Annu Rev Biochem. 2010;79:213–31. [DOI] [PubMed] [Google Scholar]
  • 45.Palomares M, Delicado A, Mansilla E, De Torres ML, Vallespín E, Fernandez L, et al. Characterization of a 8q21.11 microdeletion syndrome associated with intellectual disability and a recognizable phenotype. Am J Hum Genet. 2011;89:295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Fontana P, Ginevrino M, Bejo K, Cantalupo G, Ciavarella M, Lombardi C, et al. A ZFHX4 mutation associated with a recognizable neuropsychological and facial phenotype. Eur J Med Genet. 2021;64:104321. [DOI] [PubMed] [Google Scholar]
  • 47.Créton M, Wagener F, Massink M, Fennis W, Bloemen M, Schols J, et al. Concurrent de novo ZFHX4 variant and 16q24.1 deletion in a patient with orofacial clefting; a potential role of ZFHX4 and USP10. Am J Med Genet Part A. 2023;191:1083–8. [DOI] [PubMed] [Google Scholar]
  • 48.Sorrentino U, Fedrigo M, Calò AP, Perin M, Veronese P, Salviati L. ZFHX4 truncating variant and orofacial clefting. Am J Med Genet Part A. 2024;194:115–6. [DOI] [PubMed]
  • 49.Duncan KM, Mukherjee K, Cornell RA, Liao EC. Zebrafish models of orofacial clefts. Dev Dyn. 2017;246:897–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chudnovsky Y, Kim D, Zheng S, Whyte WA, Bansal M, Bray MA, et al. ZFHX4 interacts with the NuRD core member CHD4 and regulates the glioblastoma tumor-initiating cell state. Cell Rep. 2014;6:313–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Harvey SA, Sealy I, Kettleborough R, Fenyes F, White R, Stemple D, et al. Identification of the zebrafish maternal and paternal transcriptomes. Development. 2013;140:2703–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bhat P, Cabrera-Quio LE, Herzog VA, Fasching N, Pauli A, Ameres SL. SLAMseq resolves the kinetics of maternal and zygotic gene expression during early zebrafish embryogenesis. Cell Rep. 2023;42:112070. [DOI] [PubMed] [Google Scholar]
  • 53.Rossi A, Kontarakis Z, Gerri C, Nolte H, Hölper S, Krüger M, et al. Genetic compensation induced by deleterious mutations but not gene knockdowns. Nature. 2015;524:230–3. [DOI] [PubMed] [Google Scholar]
  • 54.Liu S, Narumi R, Ikeda N, Morita O, Tasaki J. Chemical-induced craniofacial anomalies caused by disruption of neural crest cell development in a zebrafish model. Dev Dyn. 2020;249:794–815. [DOI] [PMC free article] [PubMed]
  • 55.Conley ZR, Hague M, Kurosaka H, Dixon J, Dixon MJ, Trainor PA. A quantitative method for defining high-arched palate using the Tcof1+/− mutant mouse as a model. Dev Biol. 2016;415:296–305. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Figures (1.5MB, pdf)
Table S1 (533.8KB, pdf)

Data Availability Statement

The data that support the findings of this study are available on request from the corresponding authors. The data are not publicly available due to privacy or ethical restrictions.


Articles from European Journal of Human Genetics are provided here courtesy of Nature Publishing Group

RESOURCES